1
|
Raghani NR, Chorawala MR, Parekh K, Sharma A, Alsaidan OA, Alam P, Fareed M, Prajapati B. Exosomal miRNA-based theranostics in cervical cancer: bridging diagnostics and therapy. Med Oncol 2025; 42:193. [PMID: 40320487 DOI: 10.1007/s12032-025-02752-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Accepted: 04/28/2025] [Indexed: 06/01/2025]
Abstract
Cervical cancer (CC) remains a significant global health burden, particularly in low- and middle-income countries, where access to effective screening and treatment is limited. Despite advancements in conventional therapies, such as surgery, chemotherapy, and radiotherapy, challenges related to late-stage diagnosis, treatment resistance, and disease recurrence persist. The emergence of microRNAs (miRNAs) as key regulators of gene expression has revolutionized cancer diagnostics and therapeutics. Exosomal miRNAs, in particular, have garnered attention due to their stability, detectability in bodily fluids, and pivotal roles in tumor progression, metastasis, and immune modulation. This review provides a comprehensive overview of the role of exosomal miRNAs in the theranostic landscape of CC. We explore their involvement in disease pathogenesis, highlighting their potential as minimally invasive diagnostic biomarkers for early detection and disease monitoring. Furthermore, we examine their utility in therapeutic strategies, including miRNA-mediated drug delivery systems and miRNA-targeted interventions to overcome chemoresistance. Integrating exosomal miRNA profiling with current diagnostic modalities could enhance screening sensitivity and specificity, while miRNA-based therapies offer novel avenues to improve treatment efficacy. This review discusses recent advancements in miRNA research, current challenges in clinical translation, and future perspectives on leveraging exosomal miRNAs for personalized CC care.
Collapse
Affiliation(s)
- Neha R Raghani
- Department of Pharmacology and Pharmacy Practice, Saraswati Institute of Pharmaceutical Sciences, Gandhinagar, Gujarat, 382355, India
| | - Mehul R Chorawala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad, Gujarat, 380009, India.
| | - Kavya Parekh
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad, Gujarat, 380009, India
| | - Anvesha Sharma
- Department of Pharmacology and Pharmacy Practice, Saraswati Institute of Pharmaceutical Sciences, Gandhinagar, Gujarat, 382355, India
| | - Omar Awad Alsaidan
- Department of Pharmaceutics, College of Pharmacy, Jouf University, 72341, Sakaka, Saudi Arabia
| | - Prawez Alam
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj, 11942, Saudi Arabia
| | - Mohammad Fareed
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh, 13713, Saudi Arabia
| | - Bhupendra Prajapati
- Department of Pharmaceutics and Pharmaceutical Technology, Shree S K Patel College of Pharmaceutical Education and Research, Ganpat University, Kherva, Mahesana, Gujarat, 384012, India.
- Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand.
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
2
|
Moon KZ, Rahman MH, Alam MJ, Hossain MA, Hwang S, Kang S, Moon S, Park MN, Ahn CH, Kim B. Unraveling the interplay between cardiovascular diseases and alcohol use disorder: A bioinformatics and network-based exploration of shared molecular pathways and key biomarkers validation via western blot analysis. Comput Biol Chem 2025; 115:108338. [PMID: 39778286 DOI: 10.1016/j.compbiolchem.2024.108338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/02/2024] [Accepted: 12/26/2024] [Indexed: 01/11/2025]
Abstract
Clinical observations indicate a pronounced exacerbation of Cardiovascular Diseases (CVDs) in individuals grappling with Alcohol Use Disorder (AUD), suggesting an intricate interplay between these maladies. Pinpointing shared risk factors for both conditions has proven elusive. To address this, we pioneered a sophisticated bioinformatics framework and network-based strategy to unearth genes exhibiting aberrant expression patterns in both AUD and CVDs. In heart tissue samples from patients battling both AUD and CVDs, our study identified 76 Differentially Expressed Genes (DEGs) further used for retrieving important Gene Ontology (GO) keywords and metabolic pathways, highlighting mechanisms like proinflammatory cascades, T-cell cytotoxicity, antigen processing and presentation. By using Protein-Protein Interaction (PPI) analysis, we were able to identify key hub proteins that have a significant impact on the pathophysiology of these illnesses. Several hub proteins were identified include PTGS2, VCAM1, CCL2, CXCL8, IL7R, among these only CDH1 was covered in 10 algorithms of cytoHubba plugin. Furthermore, we pinpointed several Transcription Factors (TFs), including SOD2, CXCL8, THBS2, GREM1, CCL2, and PTGS2, alongside potential microRNAs (miRNAs) such as hsa-mir-203a-3p, hsa-mir-23a-3p, hsa-mir-98-5p, and hsa-mir-7-5p, which exert critical regulatory control over gene expression… In vitro study investigates the effect of alcohol on E-cadherin (CDH1) expression in HepG2 and Hep3B cells, showing a significant decrease in expression following ethanol treatment. These findings suggest that alcohol exposure may disrupt cell adhesion, potentially contributing to cellular changes associated with cardiovascular diseases. Our innovative approach has unveiled distinctive biomarkers delineating the dynamic interplay between AUD and various cardiovascular conditions for future therapeutic exploration.
Collapse
Affiliation(s)
- Kamelia Zaman Moon
- Department of Computer Science and Engineering, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh
| | - Md Habibur Rahman
- Department of Computer Science and Engineering, Islamic University, Kushita 7003, Bangladesh.
| | - Md Jahangir Alam
- Department of Computer Science and Engineering, Islamic University, Kushita 7003, Bangladesh
| | - Md Arju Hossain
- Department of Biochemistry and Biotechnology, Khwaja Yunus Ali University, Sirajganj 6751, Bangladesh
| | - Sungho Hwang
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Sojin Kang
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Seungjoon Moon
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Chi-Hoon Ahn
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea.
| |
Collapse
|
3
|
Tülüce Y, Köstekci S, Karakuş F, Keleş AY, Tunçyürekli M. Investigation the immunotherapeutic potential of miR-4477a targeting PD-1/PD-L1 in breast cancer cell line using a CD8 + co-culture model. Mol Biol Rep 2025; 52:326. [PMID: 40106025 DOI: 10.1007/s11033-025-10435-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND In the present study, we investigated the immunotherapeutic and anticancer activities of microRNA-4477a (miR-4477a) as a PD-L1 inhibitor in breast cancer cells (MCF-7). METHODS To this end, a series of analytical procedures were conducted, including bioinformatic analysis, RT-PCR analysis, PD-L1 ELISA, in vitro co-culture analysis, cytotoxicity assays, cell migration assays, and colony formation assays, with the objective of determining the anticancer activity of the compound in question. RESULTS The results demonstrated that miR-4477a can bind to three distinct regions of PD-L1 mRNA with high scores (94%, 88% and 80%), effectively targeting and suppressing the crucial regulatory pathways of cancer cells. In vitro studies demonstrated that a 25 nM dose of miR-4477a caused relatively high cytotoxicity in the MCF-7 cell line, suppressed PD-L1 gene expression, and decreased sPD-L1 protein levels, strongly inhibited cell migration, and significantly reduced colony formation. The in vitro co-culture analysis revealed that cancer cells were unable to evade the surveillance and cytotoxic activity of T cells (CD8+) due to the blockade of PD-L1 expression by miR-4477a. CONCLUSIONS In conclusion, miRNA-4477a has the capacity to regulate immune responses in breast cancer cells and may therefore be a promising candidate for use in cancer immunotherapy as a therapeutic agent.
Collapse
Affiliation(s)
- Yasin Tülüce
- Department of Medical Biology, Faculty of Medicine, Van Yüzüncü Yıl University, Van, 65080, Türkiye.
| | - Sedat Köstekci
- Department of Molecular Biology and Genetics, Institute of Natural and Applied Sciences, Van Yüzüncü Yıl University, Van, 65080, Türkiye
| | - Fuat Karakuş
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Van Yüzüncü Yıl University, Van, 65080, Türkiye
| | - Ahmet Yasin Keleş
- Department of Molecular Biology and Genetics, Institute of Natural and Applied Sciences, Van Yüzüncü Yıl University, Van, 65080, Türkiye
| | - Merve Tunçyürekli
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Van Yüzüncü Yıl University, Van, 65080, Türkiye
| |
Collapse
|
4
|
Krishnamoorthy VK, Hamdani F, Shukla P, Rao RA, Anaitullah S, Biligiri KK, Kadumuri RV, Pothula PR, Chavali S, Rampalli S. NSD3 protein methylation and stabilization transforms human ES cells into variant state. Life Sci Alliance 2025; 8:e202402871. [PMID: 39741006 PMCID: PMC11707394 DOI: 10.26508/lsa.202402871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 11/29/2024] [Accepted: 11/29/2024] [Indexed: 01/02/2025] Open
Abstract
Cultured human embryonic stem cells (hESCs) can develop genetic anomalies that increase their susceptibility to transformation. In this study, we characterized a variant hESC (vhESC) line and investigated the molecular mechanisms leading to the drift towards a transformed state. Our findings revealed that vhESCs up-regulate EMT-specific markers, accelerate wound healing, exhibit compromised lineage differentiation, and retain pluripotency gene expression in teratomas. Furthermore, we discovered an altered epigenomic landscape and overexpression of the lysine methyltransferases EHMT1, EHMT2, and NSD group of proteins in vhESCs. Remarkably, depleting NSD3 oncogene reversed the molecular and phenotypic changes in vhESCs. We identified a detailed mechanism where EHMT2 interacts and methylates NSD3 at lysine 477, stabilizing its protein levels in vhESCs. In addition, we showed that NSD3 levels are regulated by protein degradation in hESCs, and its stabilization leads to the emergence of the variant state. Overall, our study identify that misregulation of NSD3 in pluripotent stem cells, through methylation-mediated abrogation of its protein degradation, drives hESCs towards oncogenic transformation.
Collapse
Affiliation(s)
- Vignesh K Krishnamoorthy
- https://ror.org/05ef28661 Council of Scientific and Industrial Research (CSIR) - Institute of Genomics and Integrative Biology (IGIB), New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Fariha Hamdani
- https://ror.org/05ef28661 Council of Scientific and Industrial Research (CSIR) - Institute of Genomics and Integrative Biology (IGIB), New Delhi, India
| | - Pooja Shukla
- https://ror.org/05ef28661 Council of Scientific and Industrial Research (CSIR) - Institute of Genomics and Integrative Biology (IGIB), New Delhi, India
| | - Radhika Arasala Rao
- Institute for Stem Cell Science and Regenerative Medicine (DBT-inStem), GKVK Campus, Bangalore, India
| | - Shaikh Anaitullah
- Institute for Stem Cell Science and Regenerative Medicine (DBT-inStem), GKVK Campus, Bangalore, India
| | - Kriti Kestur Biligiri
- https://ror.org/05ef28661 Council of Scientific and Industrial Research (CSIR) - Institute of Genomics and Integrative Biology (IGIB), New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Rajashekar Varma Kadumuri
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, India
| | | | - Sreenivas Chavali
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, India
| | - Shravanti Rampalli
- https://ror.org/05ef28661 Council of Scientific and Industrial Research (CSIR) - Institute of Genomics and Integrative Biology (IGIB), New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
5
|
Tsunematsu T, Mouri Y, Shao W, Arakaki R, Ruppert JG, Murano K, Ishimaru N, Guardavaccaro D, Pagano M, Kudo Y. Sustained chromosomal passenger complex activity preserves the pluripotency of human embryonic carcinoma cells. Sci Signal 2025; 18:eadg4626. [PMID: 40136047 DOI: 10.1126/scisignal.adg4626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 01/30/2025] [Indexed: 03/27/2025]
Abstract
Human embryonic carcinoma (hEC) cells are derived from teratocarcinomas, exhibit robust proliferation, have a high differentiation potential, are the malignant counterparts of human embryonic stem cells (hESCs), and are considered hESC-like. The chromosomal passenger complex (CPC), made up of the microtuble binding protein Borealin, the kinase Aurora-B, the CPC-stabilizing inner centromere protein (INCENP), and the inhibitor of apoptosis family member Survivin, regulates cell division and is active exclusively during mitosis in somatic cells. The anaphase-promoting complex/cyclosome and its cofactor Cdh1 (APC/CCdh1) is a ubiquitylating complex that catalyzes the degradation of Aurora-B and Borealin in somatic cells but has low activity during interphase in hESCs. Here, we found that Borealin and Aurora-B exhibited sustained stability throughout the cell cycle of hEC cells due to low APC/CCdh1 activity. In contrast with somatic cells, CPC activity persisted across the cell cycle of hEC cells because of diminished APC/CCdh1 activity. Disrupting the CPC complex by depleting its constituents triggered spontaneous differentiation in hEC cells. As hEC cells differentiated, APC/CCdh1 activation curtailed CPC activity. Inactivating the CPC by pharmacologically inhibiting Aurora-B induced hEC cell differentiation by activating the epithelial-to-mesenchymal transition (EMT) program. Hence, APC/CCdh1-mediated termination of CPC activity triggered hEC cell differentiation. Collectively, these findings demonstrate a role for the CPC in governing hESC cell fate.
Collapse
Affiliation(s)
- Takaaki Tsunematsu
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8504, Japan
| | - Yasuhiro Mouri
- Department of Oral Bioscience, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8504, Japan
| | - Wenhua Shao
- Department of Oral Bioscience, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8504, Japan
| | - Rieko Arakaki
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8504, Japan
| | - Jan G Ruppert
- Wellcome Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Edinburgh EH9 3BF, Scotland, UK
| | - Kensaku Murano
- Department of Molecular Biology, Keio University School of Medicine, Tokyo, Japan
| | - Naozumi Ishimaru
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8504, Japan
| | | | - Michele Pagano
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, 522 First Avenue, SRB 1107, New York, NY 10016, USA
- NYU Perlmutter Cancer Center, New York University School of Medicine, 522 First Avenue, SRB 1107, New York, NY 10016, USA
- Howard Hughes Medical Institute, New York University School of Medicine, 522 First Avenue, SRB 1107, New York, NY 10016, USA
| | - Yasusei Kudo
- Department of Oral Bioscience, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8504, Japan
| |
Collapse
|
6
|
Alaca Y. Machine learning via DARTS-Optimized MobileViT models for pancreatic Cancer diagnosis with graph-based deep learning. BMC Med Inform Decis Mak 2025; 25:81. [PMID: 39955532 PMCID: PMC11830204 DOI: 10.1186/s12911-025-02923-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 02/07/2025] [Indexed: 02/17/2025] Open
Abstract
The diagnosis of pancreatic cancer presents a significant challenge due to the asymptomatic nature of the disease and the fact that it is frequently detected at an advanced stage. This study presents a novel approach combining graph-based data representation with DARTS-optimised MobileViT models, with the objective of enhancing diagnostic accuracy and reliability. The images of the pancreatic CT were transformed into graph structures using the Harris Corner Detection algorithm, which enables the capture of complex spatial relationships. Subsequently, the graph representations were processed using MobileViT models that had been optimised with Differentiable Architecture Search (DARTS), thereby enabling dynamic architectural adaptation. To further enhance classification accuracy, advanced machine learning algorithms, including K-Nearest Neighbours (KNN), Support Vector Machines (SVM), Random Forest (RF), and XGBoost, were applied. The MobileViTv2_150 and MobileViTv2_200 models demonstrated remarkable performance, with an accuracy of 97.33% and an F1 score of 96.25%, surpassing the capabilities of traditional CNN and Vision Transformer models. This innovative integration of graph-based deep learning and machine learning techniques demonstrates the potential of the proposed method to establish a new standard for early pancreatic cancer diagnosis. Furthermore, the study highlights the scalability of this approach for broader applications in medical imaging, which could lead to improved patient outcomes.
Collapse
Affiliation(s)
- Yusuf Alaca
- Department of Computer Engineering, Hitit University, Çorum, 19500, Turkey.
| |
Collapse
|
7
|
Meng X, Liu YP, Dai JW, Bai Y, Hu X, Azhar M, Huang XJ. The preventive effect and mechanism of Tibetan medicine Aconitum tanguticum (Maxim.) Stapf on acute lung injury. Chin Med 2025; 20:21. [PMID: 39939884 DOI: 10.1186/s13020-025-01072-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 01/23/2025] [Indexed: 02/14/2025] Open
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Aconitum tanguticum (Maxim.) Stapf (ATS) is a rare Tibetan medicinal plant that belongs to the Ranunculaceae family. This herb is mainly distributed in the high-altitude areas of Qinghai, Gansu provinces, and Tibetan Autonomous Region in China. In Tibetan medicine, ATS is mainly used to treat lung inflammation, hepatitis, gastrointestinal diseases, influenza, fever caused by infectious diseases, food poisoning, snake and scorpion bites, and yellow water disease. ATS has anti-inflammatory, antiviral, and other pharmacological effects, according to recent research. It is welltolerated by individuals from diverse ethnic groups and has a long history of use in Tibetan medicine. AIM OF THE STUDY This study investigated the preventive effects of ATS alcoholic extract on acute lung injury (ALI) in mice and aimed to elucidate its possible mechanism of action. MATERIALS AND METHODS Alveolar epithelial cells A549 and specific pathogen-free C57BL/6 mice were induced with lipopolysaccharide (LPS) to establish ALI models both in vivo and in vitro and to explore the pharmacological effects and therapeutic mechanisms of ATS. RESULTS ATS down-regulated the mRNA levels of inflammatory factors NF-κB p65, TNF-α, IL-1β, and IL-8, inhibited the release of reactive oxygen species, inhibited epithelial-mesenchymal transition caused by sustained cell injury, promoted the Keap1/Nrf2/HO-1 signalling pathway, reduced the degree of oxidative stress in vivo, and inhibited the production of proteins associated with LPS-induced ferroptosis. CONCLUSION The Tibetan medicine ATS reduced pulmonary haemorrhage and oedema in ALI mice, alleviated the degree of lung tissue lesions, inhibited the expression of inflammatory factors and apoptosis, and plays a preventive role against acute lung injury in mice.
Collapse
Affiliation(s)
- Xiang Meng
- College of Pharmaceutical Science, South-Central Minzu University, 182 Minyuan Road, Wuhan, 430074, People's Republic of China
| | - Yu-Peng Liu
- College of Pharmaceutical Science, South-Central Minzu University, 182 Minyuan Road, Wuhan, 430074, People's Republic of China
| | - Jia-Wei Dai
- College of Pharmaceutical Science, South-Central Minzu University, 182 Minyuan Road, Wuhan, 430074, People's Republic of China
| | - Yuan Bai
- College of Pharmaceutical Science, South-Central Minzu University, 182 Minyuan Road, Wuhan, 430074, People's Republic of China
| | - Xin Hu
- College of Pharmaceutical Science, South-Central Minzu University, 182 Minyuan Road, Wuhan, 430074, People's Republic of China
| | - Muhammad Azhar
- Department of Genomics and Bioinformatics, Cholistan University of Vertienary Sciences Bahwalpur, 63221, Bahwalpur, Pakistan
| | - Xian-Ju Huang
- College of Pharmaceutical Science, South-Central Minzu University, 182 Minyuan Road, Wuhan, 430074, People's Republic of China.
- Hubei International Science and Technology Cooperation Base (SH2311), Wuhan, 430074, China.
| |
Collapse
|
8
|
Lee JG, Park I, Lee H, Nam S, Kim J, Lee WS, Kang M, Kim JH. Integrating E-cadherin expression levels with TNM staging for enhanced prognostic prediction in colorectal cancer patients. BMC Cancer 2025; 25:150. [PMID: 39871234 PMCID: PMC11770905 DOI: 10.1186/s12885-025-13539-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/16/2025] [Indexed: 01/29/2025] Open
Abstract
PURPOSE This study aimed to investigate the clinical significance of E-cadherin expression levels in colorectal cancer tissues and explore the relationship between E-cadherin expression and tumor node metastasis (TNM) stage. The goal was to establish a more accurate prognostic prediction for colorectal cancer patients by analyzing E-cadherin expression levels alongside TNM staging. METHODS The study examined colorectal cancer patients by dividing them into groups based on E-cadherin expression levels. It then assessed their 5-year event-free survival (EFS) and disease-specific survival (DSS) hazard ratios (HRs). Additionally, the prognosis of patients was analyzed by combining E-cadherin expression levels with TNM staging, particularly focusing on patients in stages III and IV. RESULTS The E-cadherinLow group had significantly worse outcomes, with HRs of 2.30 for EFS and 2.76 for DSS compared to the E-cadherinHigh group. When combined with TNM stage III/IV, patients with E-cadherinLow expression showed a poor prognosis, with HRs of 1.93 for EFS and 2.35 for DSS compared to those with E-cadherinHigh expression at the same TNM stage. CONCLUSIONS Low levels of E-cadherin expression are associated with a poor prognosis and decreased survival in colorectal cancer patients. Combining E-cadherin expression levels with TNM staging provides a more precise prediction of patient prognosis and survival, potentially guiding personalized treatment strategies.
Collapse
Affiliation(s)
- Jae-Ghi Lee
- Gachon Medical Research Institute, Gachon Biomedical Convergence Institute, College of Medicine, Gachon University Gil Medical Center, Gachon University, Incheon, 21565, Republic of Korea
| | - Ilkyu Park
- Gachon Medical Research Institute, Gachon Biomedical Convergence Institute, College of Medicine, Gachon University Gil Medical Center, Gachon University, Incheon, 21565, Republic of Korea
| | - Hannah Lee
- Department of Internal Medicine, College of Medicine, Gachon University Gil Medical Center, Gachon University, Incheon, 21565, Republic of Korea
| | - Seungyoon Nam
- AI Convergence Center for Medical Science, Department of Genome Medicine and Science, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, 21565, Republic of Korea
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology, Gachon University, Incheon, 21999, Republic of Korea
| | - Jisup Kim
- Department of Pathology, College of Medicine, Gachon University Gil Medical Center, Gachon University, Incheon, 21565, Republic of Korea
| | - Won-Suk Lee
- Department of Surgery, College of Medicine, Gachon University Gil Medical Center, Gachon University, Incheon, 21565, Republic of Korea
| | - Myunghee Kang
- Department of Pathology, Dongkang Medical Center, Ulsan, 44455, Korea.
| | - Jung Ho Kim
- Gachon Medical Research Institute, Gachon Biomedical Convergence Institute, College of Medicine, Gachon University Gil Medical Center, Gachon University, Incheon, 21565, Republic of Korea.
- Department of Internal Medicine, College of Medicine, Gachon University Gil Medical Center, Gachon University, Incheon, 21565, Republic of Korea.
- Department of Translational-Clinical Medicine, Gachon Advanced Institute for Health Sciences and Technology, Gachon University, Incheon, 21999, Republic of Korea.
| |
Collapse
|
9
|
Li X, Zhang H, Dong J, Wang J. Tyrosine phosphatase SHP2 accelerated ovarian cancer via modulating integrin/ E-Cadherin/ ZEB1 induced EMT. Sci Rep 2025; 15:1535. [PMID: 39789103 PMCID: PMC11718206 DOI: 10.1038/s41598-025-85445-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025] Open
Abstract
This article focusing on examining the function and further, molecular function of SHP2 in ovarian cancer (OC). For the molecular mechanism, bioinformatics was applied to study the specifically expressed genes in ovarian cancer ; the western blotting was applied to identify the EGF, p-SHP2, ZEB1, and E-Cadherin expressions in ovarian cancer tissue and pair adjacent tissue; then SKOV3 cells were treated with EGF and infected with E-Cadherin overexpression lentivirus, and then cells were treated with benzyl butyl phthalate and IRS-1 respectively. Detection of expression of p-SHP2, ZEB1, E-Cadherin, α3-integrin, p-Src, p-SMAD2, Snail, Slug and SKOV3 cells of migration and invasion abilities were detected using Western blot method and cell scratch assay and Transwell assay; Progression of ovarian cancer was detected using subcutaneous tumor transplantation assay in nude mice and HE staining method and immunocyto. The bioinformatics analysis results suggested that SHP2 is highly specifically expressed and E-Cadherin, which is low specifically expressed in ovarian cancer tissues, while EGF, p-SHP2 or ZEB1 are highly expressed and E-Cadherin is low expressed in ovarian cancer tissues; Overexpression of E-Cadherin could reduce the expressions of p-SHP2, ZEB1, α3-integrin, p-Src, p-SMAD2, Snail and Slug might has roles in alleviating the ovarian cancer development and decreasing the levels of p-SHP2 and ZEB1 in tumor samples. And E-Cadherin overexpression reduced the migration and invasion ability of SKOV3 cells. SHP2 tyrosine phosphatase enhances the ovarian cancer cells' motility and invasiveness by upregulation of the integrin/E-Cadherin switch through ZEB1 signal.
Collapse
Affiliation(s)
- Xiaofei Li
- Department of Obstetrics and Gynecology, The Fourth Hospital of Hebei Medical University, No.12, Health Road, Shijiazhuang City, 050011, Hebei Province, China
| | - Haibo Zhang
- Department of Obstetrics and Gynecology, The Fourth Hospital of Hebei Medical University, No.12, Health Road, Shijiazhuang City, 050011, Hebei Province, China
| | - Jianan Dong
- Department of Obstetrics and Gynecology, The Fourth Hospital of Hebei Medical University, No.12, Health Road, Shijiazhuang City, 050011, Hebei Province, China
| | - Juan Wang
- Department of Obstetrics and Gynecology, The Fourth Hospital of Hebei Medical University, No.12, Health Road, Shijiazhuang City, 050011, Hebei Province, China.
| |
Collapse
|
10
|
Matsuo-Takasaki M, Kambayashi S, Hemmi Y, Wakabayashi T, Shimizu T, An Y, Ito H, Takeuchi K, Ibuki M, Kawashima T, Masayasu R, Suzuki M, Kawai Y, Umekage M, Kato TM, Noguchi M, Nakade K, Nakamura Y, Nakaishi T, Nishishita N, Tsukahara M, Hayashi Y. Complete suspension culture of human induced pluripotent stem cells supplemented with suppressors of spontaneous differentiation. eLife 2024; 12:RP89724. [PMID: 39529479 PMCID: PMC11556790 DOI: 10.7554/elife.89724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) are promising resources for producing various types of tissues in regenerative medicine; however, the improvement in a scalable culture system that can precisely control the cellular status of hiPSCs is needed. Utilizing suspension culture without microcarriers or special materials allows for massive production, automation, cost-effectiveness, and safety assurance in industrialized regenerative medicine. Here, we found that hiPSCs cultured in suspension conditions with continuous agitation without microcarriers or extracellular matrix components were more prone to spontaneous differentiation than those cultured in conventional adherent conditions. Adding PKCβ and Wnt signaling pathway inhibitors in the suspension conditions suppressed the spontaneous differentiation of hiPSCs into ectoderm and mesendoderm, respectively. In these conditions, we successfully completed the culture processes of hiPSCs, including the generation of hiPSCs from peripheral blood mononuclear cells with the expansion of bulk population and single-cell sorted clones, long-term culture with robust self-renewal characteristics, single-cell cloning, direct cryopreservation from suspension culture and their successful recovery, and efficient mass production of a clinical-grade hiPSC line. Our results demonstrate that precise control of the cellular status in suspension culture conditions paves the way for their stable and automated clinical application.
Collapse
Affiliation(s)
- Mami Matsuo-Takasaki
- iPS Cell Advanced Characterization and Development Team, RIKEN BioResource Research CenterIbarakiJapan
| | - Sho Kambayashi
- Regenerative Medicine and Cell Therapy Laboratories, KANEKA CORPORATIONKobeJapan
| | - Yasuko Hemmi
- iPS Cell Advanced Characterization and Development Team, RIKEN BioResource Research CenterIbarakiJapan
| | - Tamami Wakabayashi
- iPS Cell Advanced Characterization and Development Team, RIKEN BioResource Research CenterIbarakiJapan
| | - Tomoya Shimizu
- iPS Cell Advanced Characterization and Development Team, RIKEN BioResource Research CenterIbarakiJapan
| | - Yuri An
- iPS Cell Advanced Characterization and Development Team, RIKEN BioResource Research CenterIbarakiJapan
| | - Hidenori Ito
- iPS Cell Advanced Characterization and Development Team, RIKEN BioResource Research CenterIbarakiJapan
| | - Kazuhiro Takeuchi
- Regenerative Medicine and Cell Therapy Laboratories, KANEKA CORPORATIONKobeJapan
| | - Masato Ibuki
- Regenerative Medicine and Cell Therapy Laboratories, KANEKA CORPORATIONKobeJapan
| | - Terasu Kawashima
- Regenerative Medicine and Cell Therapy Laboratories, KANEKA CORPORATIONKobeJapan
| | - Rio Masayasu
- Regenerative Medicine and Cell Therapy Laboratories, KANEKA CORPORATIONKobeJapan
| | - Manami Suzuki
- Regenerative Medicine and Cell Therapy Laboratories, KANEKA CORPORATIONKobeJapan
| | - Yoshikazu Kawai
- Regenerative Medicine and Cell Therapy Laboratories, KANEKA CORPORATIONKobeJapan
| | | | - Tomoaki M Kato
- Research and Development Center, CiRA FoundationKyotoJapan
| | - Michiya Noguchi
- Cell Engineering Division, RIKEN BioResource Research CenterIbarakiJapan
| | - Koji Nakade
- Gene Engineering Division, RIKEN BioResource Research CenterIbarakiJapan
| | - Yukio Nakamura
- Cell Engineering Division, RIKEN BioResource Research CenterIbarakiJapan
| | - Tomoyuki Nakaishi
- Regenerative Medicine and Cell Therapy Laboratories, KANEKA CORPORATIONKobeJapan
| | - Naoki Nishishita
- Regenerative Medicine and Cell Therapy Laboratories, KANEKA CORPORATIONKobeJapan
| | | | - Yohei Hayashi
- iPS Cell Advanced Characterization and Development Team, RIKEN BioResource Research CenterIbarakiJapan
- Faculty of Medicine and School of Integrative and Global Majors, University of TsukubaIbarakiJapan
| |
Collapse
|
11
|
Bonetto V, Pagano CA, Sabbatini M, Magnelli V, Donadelli M, Marengo E, Masini MA. Microgravity as a Tool to Investigate Cancer Induction in Pleura Mesothelial Cells. Curr Issues Mol Biol 2024; 46:10896-10912. [PMID: 39451527 PMCID: PMC11506709 DOI: 10.3390/cimb46100647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024] Open
Abstract
The present work shows that the exposure of mesothelial cells to simulated microgravity changes their cytoskeleton and adhesion proteins, leading to a cell switch from normal towards tumoral cells. Immunohistochemical and molecular data were obtained from both MeT-5A exposed to simulated microgravity and BR95 mesothelioma cell lines. Simulated microgravity was found to affect the expression of actin, vinculin, and connexin-43, altering their quantitative and spatial distribution pattern inside the cell. The analysis of the tumoral markers p27, CD44, Fibulin-3, and NANOG and the expression of genes related to cancer transformation such as NANOG, CDH-1, and Zeb-1 showed that the simulated microgravity environment led to expression patterns in MeT-5A cells similar to those observed in BR95 cells. The alteration in both quantitative expression and structural organization of the cytoskeleton and adhesion/communication proteins can thus be considered a pivotal mechanism involved in the cellular shift towards tumoral progression.
Collapse
Affiliation(s)
- Valentina Bonetto
- Department of Science and Innovation Technology (DISIT), Università del Piemonte Orientale, 15121 Alessandria, Italy; (V.B.); (C.A.P.); (V.M.); (E.M.); (M.A.M.)
| | - Corinna Anais Pagano
- Department of Science and Innovation Technology (DISIT), Università del Piemonte Orientale, 15121 Alessandria, Italy; (V.B.); (C.A.P.); (V.M.); (E.M.); (M.A.M.)
| | - Maurizio Sabbatini
- Department of Science and Innovation Technology (DISIT), Università del Piemonte Orientale, 15121 Alessandria, Italy; (V.B.); (C.A.P.); (V.M.); (E.M.); (M.A.M.)
| | - Valeria Magnelli
- Department of Science and Innovation Technology (DISIT), Università del Piemonte Orientale, 15121 Alessandria, Italy; (V.B.); (C.A.P.); (V.M.); (E.M.); (M.A.M.)
| | - Massimo Donadelli
- Department of Neurosciences, Biomedicine and Movement Sciences (DNBM), University of Verona, 37124 Verona, Italy;
| | - Emilio Marengo
- Department of Science and Innovation Technology (DISIT), Università del Piemonte Orientale, 15121 Alessandria, Italy; (V.B.); (C.A.P.); (V.M.); (E.M.); (M.A.M.)
| | - Maria Angela Masini
- Department of Science and Innovation Technology (DISIT), Università del Piemonte Orientale, 15121 Alessandria, Italy; (V.B.); (C.A.P.); (V.M.); (E.M.); (M.A.M.)
| |
Collapse
|
12
|
Wang R, Shu RR, Seldin L. Noncanonical functions of adhesion proteins in inflammation. Am J Physiol Cell Physiol 2024; 327:C505-C515. [PMID: 38981610 PMCID: PMC11427013 DOI: 10.1152/ajpcell.00292.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/24/2024] [Accepted: 07/01/2024] [Indexed: 07/11/2024]
Abstract
Cell adhesion proteins localize to epithelial and endothelial cell membranes to form junctional complexes between neighboring cells or between cells and the underlying basement membrane. The structural and functional integrities of these junctions are critical to establish cell polarity and maintain tissue barrier function, while also facilitating leukocyte migration and adhesion to sites of inflammation. In addition to their adhesive properties, however, junctional proteins can also serve important noncanonical functions in inflammatory signaling and transcriptional regulation. Intriguingly, recent work has unveiled novel roles for cell adhesion proteins as both signaling initiators and downstream targets during inflammation. In this review, we discuss both the traditional functions of junction proteins in cell adhesion and tissue barrier function as well as their noncanonical signaling roles that have been implicated in facilitating diverse inflammatory pathologies.
Collapse
Affiliation(s)
- Ruochong Wang
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Raphael R Shu
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Lindsey Seldin
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, United States
- Department of Dermatology, Emory University School of Medicine, Atlanta, Georgia, United States
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States
- Atlanta Veterans Affairs Medical Center, Decatur, Georgia, United States
| |
Collapse
|
13
|
Huang Y, Yang Y, Chen X, Zeng S, Chen Y, Wang H, Lv X, Hu X, Teng L. Downregulation of malic enzyme 3 facilitates progression of gastric carcinoma via regulating intracellular oxidative stress and hypoxia-inducible factor-1α stabilization. Cell Mol Life Sci 2024; 81:375. [PMID: 39212717 PMCID: PMC11364750 DOI: 10.1007/s00018-024-05388-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 07/04/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Gastric cancer (GC) is one of the most malignant cancers worldwide. Metabolism disorder is a critical characteristic of malignant tumors related to tumor progression and metastasis. However, the expression and molecular mechanism of malic enzyme 3 (ME3) in GC are rarely reported. In this study, we aim to investigate the molecular mechanism of ME3 in the development of GC and to explore its potential value as a prognostic and therapeutic target in GC. METHOD ME3 mRNA and protein expression were evaluated in patients with GC using RT-qPCR, WB, and immunohistochemistry, as well as their correlation with clinicopathological indicators. The effect of ME3 on proliferation and metastasis was evaluated using Cell Counting Kit-8 (CCK-8), 5-ethynyl-20-deoxyuridine (EdU) assay, transwell assay, wound healing assay, and subcutaneous injection or tail vein injection of tumor cells in mice model. The effects of ME3 knockdown on the level of metabolites and hypoxia-inducible factor-1α (HIF-1α) protein were determined in GC cells. Oxidative phosphorylation was measured to evaluate adenosine triphosphate (ATP) production. RESULTS ME3 was downregulated in human GC tissues (P < 0.001). The decreased ME3 mRNA expression was associated with younger age (P = 0.02), pathological staging (P = 0.049), and lymph node metastasis (P = 0.001), while low ME3 expression was associated with tumor size (P = 0.048), tumor invasion depth (P < 0.001), lymph node metastasis (P = 0.018), TNM staging (P < 0.001), and poor prognosis (OS, P = 0.0206; PFS P = 0.0453). ME3 knockdown promoted GC cell malignancy phenotypes. Moreover, α-ketoglutarate (α-KG) and NADPH/NADP+ ratios were reduced while malate was increased in the ME3 knockdown group under normoxia. When cells were incubated under hypoxia, the NADPH/NADP+ ratio and α-KG decreased while intracellular reactive oxygen species (ROS) increased significantly. The ME3 knockdown group exhibited an increase in ATP production and while ME3 overexpression group exhibited oppositely. We discovered that ME3 and HIF-1α expression were negatively correlated in GC cells and tissues, and proposed the hypothesis: downregulation of ME3 promotes GC progression via regulating intracellular oxidative stress and HIF-1α. CONCLUSION We provide evidence that ME3 downregulation is associated with poor prognosis in GC patients and propose a hypothesis for the ME3 regulatory mechanism in GC progression. The present study is of great scientific significance and clinical value for exploring the prognostic and therapeutic targets of GC, evaluating and improving the clinical efficacy of patients, reducing recurrence and metastasis, and improving the prognosis and quality of life of patients.
Collapse
Affiliation(s)
- Yingying Huang
- Department of Oncological Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Institute (Key Laboratory for Cancer Intervention and Prevention, China National Ministry of Education, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Gynecology, Guangzhou First People's Hospital, Guangzhou, China
| | - Yan Yang
- Department of Oncological Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiangliu Chen
- Department of Oncological Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Siying Zeng
- Cancer Institute (Key Laboratory for Cancer Intervention and Prevention, China National Ministry of Education, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiran Chen
- Department of Oncological Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Haiyong Wang
- Department of Oncological Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiadong Lv
- Department of Oncological Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xun Hu
- Cancer Institute (Key Laboratory for Cancer Intervention and Prevention, China National Ministry of Education, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lisong Teng
- Department of Oncological Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
14
|
Cheng YC, Zhang Y, Tripathi S, Harshavardhan BV, Jolly MK, Schiebinger G, Levine H, McDonald TO, Michor F. Reconstruction of single-cell lineage trajectories and identification of diversity in fates during the epithelial-to-mesenchymal transition. Proc Natl Acad Sci U S A 2024; 121:e2406842121. [PMID: 39093947 PMCID: PMC11317558 DOI: 10.1073/pnas.2406842121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 06/25/2024] [Indexed: 08/04/2024] Open
Abstract
Exploring the complexity of the epithelial-to-mesenchymal transition (EMT) unveils a diversity of potential cell fates; however, the exact timing and mechanisms by which early cell states diverge into distinct EMT trajectories remain unclear. Studying these EMT trajectories through single-cell RNA sequencing is challenging due to the necessity of sacrificing cells for each measurement. In this study, we employed optimal-transport analysis to reconstruct the past trajectories of different cell fates during TGF-beta-induced EMT in the MCF10A cell line. Our analysis revealed three distinct trajectories leading to low EMT, partial EMT, and high EMT states. Cells along the partial EMT trajectory showed substantial variations in the EMT signature and exhibited pronounced stemness. Throughout this EMT trajectory, we observed a consistent downregulation of the EED and EZH2 genes. This finding was validated by recent inhibitor screens of EMT regulators and CRISPR screen studies. Moreover, we applied our analysis of early-phase differential gene expression to gene sets associated with stemness and proliferation, pinpointing ITGB4, LAMA3, and LAMB3 as genes differentially expressed in the initial stages of the partial versus high EMT trajectories. We also found that CENPF, CKS1B, and MKI67 showed significant upregulation in the high EMT trajectory. While the first group of genes aligns with findings from previous studies, our work uniquely pinpoints the precise timing of these upregulations. Finally, the identification of the latter group of genes sheds light on potential cell cycle targets for modulating EMT trajectories.
Collapse
Affiliation(s)
- Yu-Chen Cheng
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA02215
- Center for Cancer Evolution, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA02138
| | - Yun Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing100021, China
| | - Shubham Tripathi
- Yale Center for Systems and Engineering Immunology and Department of Immunobiology, Yale School of Medicine, New Haven, CT06510
| | - B. V. Harshavardhan
- Interdisciplinary Mathematics Initiative, Indian Institute of Science, Bangalore560012, India
| | - Mohit Kumar Jolly
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore560012, India
| | - Geoffrey Schiebinger
- Department of Mathematics, University of British Columbia, Vancouver, BCV6T 1Z2, Canada
| | - Herbert Levine
- Center for Theoretical Biological Physics, Northeastern University, Boston, MA02115
- Department of Physics, Northeastern University, Boston, MA02115
| | - Thomas O. McDonald
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA02215
- Center for Cancer Evolution, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA02138
| | - Franziska Michor
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA02215
- Center for Cancer Evolution, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA02138
- The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA02138
- The Ludwig Center at Harvard, Boston, MA02115
| |
Collapse
|
15
|
Guo Q, Chen J, Bu Q, Zhang J, Ruan M, Chen X, Zhao M, Tu X, Zhao C. Establishing stable and highly osteogenic hiPSC-derived MSCs for 3D-printed bone graft through microenvironment modulation by CHIR99021-treated osteocytes. Mater Today Bio 2024; 26:101111. [PMID: 38933413 PMCID: PMC11201125 DOI: 10.1016/j.mtbio.2024.101111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 05/17/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
Human induced pluripotent stem cell (hiPSC)-derived mesenchymal stem cells (iMSCs) are ideal candidates for the production of standardised and scalable bioengineered bone grafts. However, stable induction and osteogenic differentiation of iMSCs pose challenges in the industry. We developed a precise differentiation method to produce homogeneous and fully differentiated iMSCs. In this study, we established a standardised system to prepare iMSCs with increased osteogenic potential and improved bioactivity by introducing a CHIR99021 (C91)-treated osteogenic microenvironment (COOME). COOME enhances the osteogenic differentiation and mineralisation of iMSCs via canonical Wnt signalling. Global transcriptome analysis and co-culturing experiments indicated that COOME increased the pro-angiogenesis/neurogenesis activity of iMSCs. The superior osteogenic differentiation and mineralisation abilities of COOME-treated iMSCs were also confirmed in a Bio3D module generated using a polycaprolactone (PCL) and cell-integrated 3D printing (PCI3D) system, which is the closest model to in vivo research. This COOME-treated iMSCs differentiation system offers a new perspective for generating highly osteogenic, bioactive, and anatomically matched grafts for clinical applications. Statement of significance Although human induced pluripotent stem cell-derived MSCs (iMSCs) are ideal seed cells for synthetic bone implants, the challenges of stable induction and osteogenic differentiation hinder their clinical application. This study established a standardised system for the scalable preparation of iMSCs with improved osteogenic potential by combining our precise iMSC differentiation method with the CHIR99021 (C91)-treated osteocyte osteogenic microenvironment (COOME) through the activation of canonical Wnt signalling. Moreover, COOME upregulated the pro-angiogenic and pro-neurogenic capacities of iMSCs, which are crucial for the integration of implanted bone grafts. The superior osteogenic ability of COOME-treated iMSCs was confirmed in Bio3D modules generated using PCL and cell-integrated 3D printing systems, highlighting their functional potential in vivo. This study contributes to tissue engineering by providing insights into the functional differentiation of iMSCs for bone regeneration.
Collapse
Affiliation(s)
- Qiuling Guo
- Laboratory of Skeletal Development and Regeneration, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Jingjing Chen
- Laboratory of Skeletal Development and Regeneration, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Qiqi Bu
- Laboratory of Skeletal Development and Regeneration, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Jinling Zhang
- Laboratory of Skeletal Development and Regeneration, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Minjie Ruan
- Laboratory of Skeletal Development and Regeneration, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Xiaoyu Chen
- Center for Medical Epigenetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Mingming Zhao
- Center for Medical Epigenetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Xiaolin Tu
- Laboratory of Skeletal Development and Regeneration, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Chengzhu Zhao
- Laboratory of Skeletal Development and Regeneration, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
16
|
Kulus M, Farzaneh M, Bryja A, Zehtabi M, Azizidoost S, Abouali Gale Dari M, Golcar-Narenji A, Ziemak H, Chwarzyński M, Piotrowska-Kempisty H, Dzięgiel P, Zabel M, Mozdziak P, Bukowska D, Kempisty B, Antosik P. Phenotypic Transitions the Processes Involved in Regulation of Growth and Proangiogenic Properties of Stem Cells, Cancer Stem Cells and Circulating Tumor Cells. Stem Cell Rev Rep 2024; 20:967-979. [PMID: 38372877 PMCID: PMC11087301 DOI: 10.1007/s12015-024-10691-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/01/2024] [Indexed: 02/20/2024]
Abstract
Epithelial-mesenchymal transition (EMT) is a crucial process with significance in the metastasis of malignant tumors. It is through the acquisition of plasticity that cancer cells become more mobile and gain the ability to metastasize to other tissues. The mesenchymal-epithelial transition (MET) is the return to an epithelial state, which allows for the formation of secondary tumors. Both processes, EMT and MET, are regulated by different pathways and different mediators, which affects the sophistication of the overall tumorigenesis process. Not insignificant are also cancer stem cells and their participation in the angiogenesis, which occur very intensively within tumors. Difficulties in effectively treating cancer are primarily dependent on the potential of cancer cells to rapidly expand and occupy secondarily vital organs. Due to the ability of these cells to spread, the concept of the circulating tumor cell (CTC) has emerged. Interestingly, CTCs exhibit molecular diversity and stem-like and mesenchymal features, even when derived from primary tumor tissue from a single patient. While EMT is necessary for metastasis, MET is required for CTCs to establish a secondary site. A thorough understanding of the processes that govern the balance between EMT and MET in malignancy is crucial.
Collapse
Affiliation(s)
- Magdalena Kulus
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Torun, Poland
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Artur Bryja
- Division of Anatomy, Department of Human Morphology and Embryology, Wroclaw Medical University, Wroclaw, Poland
| | - Mojtaba Zehtabi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shirin Azizidoost
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mahrokh Abouali Gale Dari
- Department of Obstetrics and Gynecology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Afsaneh Golcar-Narenji
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC, USA
| | - Hanna Ziemak
- Veterinary Clinic of the Nicolaus Copernicus University in Torun, Torun, Poland
| | - Mikołaj Chwarzyński
- Veterinary Clinic of the Nicolaus Copernicus University in Torun, Torun, Poland
| | - Hanna Piotrowska-Kempisty
- Department of Toxicology, Poznan University of Medical Sciences, Poznan, Poland
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Torun, Poland
| | - Piotr Dzięgiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, Wroclaw, Poland
- Department of Physiotherapy, Wroclaw University School of Physical Education, Wroclaw, Poland
| | - Maciej Zabel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, Wroclaw, Poland
- Division of Anatomy and Histology, University of Zielona Góra, Zielona Góra, Poland
| | - Paul Mozdziak
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC, USA
- Physiology Graduate Faculty, North Carolina State University, Raleigh, NC, USA
| | - Dorota Bukowska
- Department of Diagnostics and Clinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Torun, Poland
| | - Bartosz Kempisty
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Torun, Poland.
- Division of Anatomy, Department of Human Morphology and Embryology, Wroclaw Medical University, Wroclaw, Poland.
- Physiology Graduate Faculty, North Carolina State University, Raleigh, NC, USA.
- Department of Obstetrics and Gynecology, University Hospital and Masaryk University, Brno, Czech Republic.
| | - Paweł Antosik
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Torun, Poland
| |
Collapse
|
17
|
Husser MC, Pham NP, Law C, Araujo FRB, Martin VJJ, Piekny A. Endogenous tagging using split mNeonGreen in human iPSCs for live imaging studies. eLife 2024; 12:RP92819. [PMID: 38652106 PMCID: PMC11037917 DOI: 10.7554/elife.92819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024] Open
Abstract
Endogenous tags have become invaluable tools to visualize and study native proteins in live cells. However, generating human cell lines carrying endogenous tags is difficult due to the low efficiency of homology-directed repair. Recently, an engineered split mNeonGreen protein was used to generate a large-scale endogenous tag library in HEK293 cells. Using split mNeonGreen for large-scale endogenous tagging in human iPSCs would open the door to studying protein function in healthy cells and across differentiated cell types. We engineered an iPS cell line to express the large fragment of the split mNeonGreen protein (mNG21-10) and showed that it enables fast and efficient endogenous tagging of proteins with the short fragment (mNG211). We also demonstrate that neural network-based image restoration enables live imaging studies of highly dynamic cellular processes such as cytokinesis in iPSCs. This work represents the first step towards a genome-wide endogenous tag library in human stem cells.
Collapse
Affiliation(s)
| | - Nhat P Pham
- Biology Department, Concordia University, Montreal, Canada
| | - Chris Law
- Biology Department, Concordia University, Montreal, Canada
- Center for Microscopy and Cellular Imaging, Concordia University, Montreal, Canada
| | - Flavia R B Araujo
- Center for Applied Synthetic Biology, Concordia University, Montreal, Canada
| | - Vincent J J Martin
- Biology Department, Concordia University, Montreal, Canada
- Center for Applied Synthetic Biology, Concordia University, Montreal, Canada
| | - Alisa Piekny
- Biology Department, Concordia University, Montreal, Canada
- Center for Microscopy and Cellular Imaging, Concordia University, Montreal, Canada
- Center for Applied Synthetic Biology, Concordia University, Montreal, Canada
| |
Collapse
|
18
|
Rodrigues DB, Moreira HR, Jarnalo M, Horta R, Marques AP, Reis RL, Pirraco RP. Generation of 3D melanoma models using an assembloid-based approach. Acta Biomater 2024; 178:93-110. [PMID: 38382833 DOI: 10.1016/j.actbio.2024.02.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 01/31/2024] [Accepted: 02/13/2024] [Indexed: 02/23/2024]
Abstract
While 3D tumor models have greatly evolved over the past years, there is still a strong requirement for more biosimilar models which are capable of recapitulating cellular crosstalk within the tumor microenvironment while equally displaying representative levels of tumor aggressiveness and invasion. Herein, we disclose an assembloid melanoma model based on the fusion of individual stromal multicellular spheroids (MCSs). In contrast to more traditional tumor models, we show that it is possible to develop self-organizing, heterotypic melanoma models where tumor cells present stem-cell like features like up-regulated pluripotency master regulators SOX2, POU5F1 and NANOG. Additionally, these assembloids display high levels of invasiveness while embedded in 3D matrices as evidenced by stromal cell promotion of melanoma cell invasion via metalloproteinase production. Furthermore, sensitivity to anticancer drug doxorubicin was demonstrated for the melanoma assembloid model. These findings suggest that melanoma assembloids may play a significant role in the field of 3D cancer models as they more closely mimic the tumor microenvironment when compared to more traditional MCSs, opening the doors to a better understanding of the role of tumor microenvironment in supporting tumor progression. STATEMENT OF SIGNIFICANCE: The development of complex 3D tumor models that better recapitulate the tumor microenvironment is crucial for both an improved comprehension of intercellular crosstalk and for more efficient drug screening. We have herein developed a self-organizing heterotypic assembloid-based melanoma model capable of closely mimicking the tumor microenvironment. Key features recapitulated were the preservation of cancer cell stemness, sensitivity to anti-cancer agents and tumor cell invasion promoted by stromal cells. The approach of pre-establishing distinct stromal domains for subsequent combination into more complex tumor constructs provides a route for developing superior tumor models with a higher degree of similarity to native cancer tissues.
Collapse
Affiliation(s)
- Daniel B Rodrigues
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães 4805-017, Portugal
| | - Helena R Moreira
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães 4805-017, Portugal
| | - Mariana Jarnalo
- Department of Plastic and Reconstructive Surgery, and Burn Unity, Centro Hospitalar de São João, Porto, Portugal; Faculty of Medicine - University of Porto, Portugal
| | - Ricardo Horta
- Department of Plastic and Reconstructive Surgery, and Burn Unity, Centro Hospitalar de São João, Porto, Portugal; Faculty of Medicine - University of Porto, Portugal
| | - Alexandra P Marques
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães 4805-017, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães 4805-017, Portugal
| | - Rogério P Pirraco
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães 4805-017, Portugal.
| |
Collapse
|
19
|
Correia LFL, Leal GR, Brandão FZ, Batista RITP, Souza-Fabjan JMG. Effect of antifreeze protein I in the freezing solution on in vivo-derived sheep embryos. Res Vet Sci 2024; 168:105132. [PMID: 38183895 DOI: 10.1016/j.rvsc.2023.105132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/17/2023] [Accepted: 12/28/2023] [Indexed: 01/08/2024]
Abstract
This study evaluated the effects of different antifreeze protein type I (AFP I) concentrations added to a slow freezing solution in sheep in vivo-derived embryos. Good-quality embryos were allocated into: AFP-free (CONT); 0.1 μg/mL of AFP I (AFP0.1); or 0.5 μg/mL of AFP I (AFP0.5). After thawing, embryos were in vitro cultured (IVC) for 48 h. At 24 h and 48 h of IVC, dead cells and apoptosis, mitochondrial activity, intracellular reactive oxygen species (ROS), and glutathione (GSH) evaluations were performed. At 24 h, evaluated embryos were submitted to RT-qPCR for metabolism (SIRT2, PRDX1, OCT4, CDX2) and quality (AQP3, CDH1, HSP70, BAX, BCL2) genes. The in vitro survival rate was 56% (22/39) for CONT, 60% (32/53) for AFP0.1, and 53% (23/43) for AFP0.5 (p > 0.05). A tendency (p = 0.09) for a higher blastocyst hatching rate was noted in AFP0.1 (62%) compared to AFP0.5 (33%), and both groups were similar to CONT (50%). An increased (p < 0.05) mitochondrial activity at 24 h was observed in AFP0.1 compared to CONT. No differences (p > 0.05) were observed in oxidative stress homeostasis and viability between treatments. A downregulation (p < 0.05) of CDH1 in AFP0.1 and a downregulation of AQP3 in AFP0.5 were observed in comparison to the other groups. An upregulation (p < 0.05) was detected in HSP70 and BCL2 on AFP0.5 compared to AFP0.1 group. The addition of AFP I in slow freezing solution can benefit cryopreserved sheep in vivo-derived embryos, without affecting embryonic survival.
Collapse
Affiliation(s)
- Lucas F L Correia
- Faculdade de Veterinária, Universidade Federal Fluminense, Av. Vital Brazil Filho, 64, CEP 24230-340 Niterói, RJ, Brazil.
| | - Gabriela R Leal
- Faculdade de Veterinária, Universidade Federal Fluminense, Av. Vital Brazil Filho, 64, CEP 24230-340 Niterói, RJ, Brazil
| | - Felipe Z Brandão
- Faculdade de Veterinária, Universidade Federal Fluminense, Av. Vital Brazil Filho, 64, CEP 24230-340 Niterói, RJ, Brazil
| | - Ribrio I T P Batista
- Faculdade de Veterinária, Universidade Federal Fluminense, Av. Vital Brazil Filho, 64, CEP 24230-340 Niterói, RJ, Brazil
| | - Joanna M G Souza-Fabjan
- Faculdade de Veterinária, Universidade Federal Fluminense, Av. Vital Brazil Filho, 64, CEP 24230-340 Niterói, RJ, Brazil.
| |
Collapse
|
20
|
Ghorbanzadeh V, Hassan ALJAF KA, Wasman HM, Dariushnejad H. Crocin inhibit the metastasis of MDA-MB-231 cell line by suppressing epithelial to mesenchymal transition through WNT/β-catenin signalling pathway. Ann Med Surg (Lond) 2024; 86:1401-1407. [PMID: 38463069 PMCID: PMC10923327 DOI: 10.1097/ms9.0000000000001691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 12/27/2023] [Indexed: 03/12/2024] Open
Abstract
Background Triple-negative breast cancer has the poorest prognosis and survival rates compared to other breast cancer subtypes due to its invasive behaviours. This type of cancer does not respond to biological therapies and exhibits resistance to available treatment options. Therefore, it is imperative to discover new therapeutics to address this challenge. Methods In this study, a TNBC cell line was utilized to investigate the anti-metastatic effect of crocin on the Wnt/β-catenin pathway. Cell proliferation was assessed using the MTT assay, and the effects of crocin on migration were monitored through transwell and wound healing experiments. The expression of specific epithelial-mesenchymal transition marker genes was evaluated using real-time polymerase chain reaction, and β-catenin expression was also examined through real-time polymerase chain reaction. Results The findings revealed that crocin significantly inhibits cell proliferation and migration of tumour cells in a dose-dependent manner. Moreover, crocin decreased the expression of Vimentin, Snail, Zeb-1, and β-catenin. Additionally, crocin increased the expression of E-cadherin in the MDA-MB-231 cell line. Conclusions The results demonstrated an association between crocin and the Wnt/β-catenin signalling pathway. In conclusion, this study establishes that crocin holds promise as a potential therapeutic option for triple-negative breast cancer.
Collapse
Affiliation(s)
| | | | - Hunar Mustafa Wasman
- Medical Laboratory Science Department, University of Raparin, Kurdistan Region, Iraq
| | - Hassan Dariushnejad
- Razi Herbal Medicines Research Center
- Department of Medical Biotechnology, Faculty of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| |
Collapse
|
21
|
Thiery JP, Sheng G, Shu X, Runyan R. How studies in developmental epithelial-mesenchymal transition and mesenchymal-epithelial transition inspired new research paradigms in biomedicine. Development 2024; 151:dev200128. [PMID: 38300897 DOI: 10.1242/dev.200128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
Epithelial-mesenchymal transition (EMT) and its reverse mechanism, mesenchymal-epithelial transition (MET), are evolutionarily conserved mechanisms initially identified in studies of early metazoan development. EMT may even have been established in choanoflagellates, the closest unicellular relative of Metazoa. These crucial morphological transitions operate during body plan formation and subsequently in organogenesis. These findings have prompted an increasing number of investigators in biomedicine to assess the importance of such mechanisms that drive epithelial cell plasticity in multiple diseases associated with congenital disabilities and fibrosis, and, most importantly, in the progression of carcinoma. EMT and MET also play crucial roles in regenerative medicine, notably by contributing epigenetic changes in somatic cells to initiate reprogramming into stem cells and their subsequent differentiation into distinct lineages.
Collapse
Affiliation(s)
| | - Guojun Sheng
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Xiaodong Shu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Raymond Runyan
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
22
|
Rodrigues DB, Reis RL, Pirraco RP. Modelling the complex nature of the tumor microenvironment: 3D tumor spheroids as an evolving tool. J Biomed Sci 2024; 31:13. [PMID: 38254117 PMCID: PMC10804490 DOI: 10.1186/s12929-024-00997-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Cancer remains a serious burden in society and while the pace in the development of novel and more effective therapeutics is increasing, testing platforms that faithfully mimic the tumor microenvironment are lacking. With a clear shift from animal models to more complex in vitro 3D systems, spheroids emerge as strong options in this regard. Years of development have allowed spheroid-based models to better reproduce the biomechanical cues that are observed in the tumor-associated extracellular matrix (ECM) and cellular interactions that occur in both a cell-cell and cell-ECM manner. Here, we summarize some of the key cellular interactions that drive tumor development, progression and invasion, and how successfully are these interactions recapitulated in 3D spheroid models currently in use in the field. We finish by speculating on future advancements in the field and on how these can shape the relevance of spherical 3D models for tumor modelling.
Collapse
Affiliation(s)
- Daniel B Rodrigues
- 3B's Research Group, I3Bs, Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence On Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017, Guimarães, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga, 4805-017, Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs, Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence On Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017, Guimarães, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga, 4805-017, Guimarães, Portugal
| | - Rogério P Pirraco
- 3B's Research Group, I3Bs, Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence On Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017, Guimarães, Portugal.
- ICVS/3B's, PT Government Associate Laboratory, Braga, 4805-017, Guimarães, Portugal.
| |
Collapse
|
23
|
Tan F, Li X, Li X, Xu M, Shahzad KA, Hou L. GelMA/PEDOT:PSS Composite Conductive Hydrogel-Based Generation and Protection of Cochlear Hair Cells through Multiple Signaling Pathways. Biomolecules 2024; 14:95. [PMID: 38254695 PMCID: PMC10812993 DOI: 10.3390/biom14010095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 12/18/2023] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Recent advances in cochlear implantology are exemplified by novel functional strategies such as bimodal electroacoustic stimulation, in which the patient has intact low-frequency hearing and profound high-frequency hearing pre-operatively. Therefore, the synergistic restoration of dysfunctional cochlear hair cells and the protection of hair cells from ototoxic insults have become a persistent target pursued for this hybrid system. In this study, we developed a composite GelMA/PEDOT:PSS conductive hydrogel that is suitable as a coating for the cochlear implant electrode for the potential local delivery of otoregenerative and otoprotective drugs. Various material characterization methods (e.g., 1H NMR spectroscopy, FT-IR, EIS, and SEM), experimental models (e.g., murine cochlear organoid and aminoglycoside-induced ototoxic HEI-OC1 cellular model), and biological analyses (e.g., confocal laser scanning microscopy, real time qPCR, flow cytometry, and bioinformatic sequencing) were used. The results demonstrated decent material properties of the hydrogel, such as mechanical (e.g., high tensile stress and Young's modulus), electrochemical (e.g., low impedance and high conductivity), biocompatibility (e.g., satisfactory cochlear cell interaction and free of systemic toxicity), and biosafety (e.g., minimal hemolysis and cell death) features. In addition, the CDR medicinal cocktail sustainably released by the hydrogel not only promoted the expansion of the cochlear stem cells but also boosted the trans-differentiation from cochlear supporting cells into hair cells. Furthermore, hydrogel-based drug delivery protected the hair cells from oxidative stress and various forms of programmed cell death (e.g., apoptosis and ferroptosis). Finally, using large-scale sequencing, we enriched a complex network of signaling pathways that are potentially downstream to various metabolic processes and abundant metabolites. In conclusion, we present a conductive hydrogel-based local delivery of bifunctional drug cocktails, thereby serving as a potential solution to intracochlear therapy of bimodal auditory rehabilitation and diseases beyond.
Collapse
Affiliation(s)
- Fei Tan
- Department of ORL-HNS, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200070, China; (X.L.); (M.X.); (K.A.S.)
- Plasma Medicine and Surgical Implants Center, School of Medicine, Tongji University, Shanghai 200070, China
- Department of ORL-HNS, The Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
- Department of ORL-HNS, The Royal College of Surgeons of England, London WC2A 3PE, UK
| | - Xuran Li
- Department of ORL-HNS, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200070, China; (X.L.); (M.X.); (K.A.S.)
- Plasma Medicine and Surgical Implants Center, School of Medicine, Tongji University, Shanghai 200070, China
| | - Xiao Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology & Center for Advanced Low-Dimension Materials, Donghua University, Shanghai 200051, China; (X.L.); (L.H.)
| | - Maoxiang Xu
- Department of ORL-HNS, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200070, China; (X.L.); (M.X.); (K.A.S.)
- Plasma Medicine and Surgical Implants Center, School of Medicine, Tongji University, Shanghai 200070, China
| | - Khawar Ali Shahzad
- Department of ORL-HNS, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200070, China; (X.L.); (M.X.); (K.A.S.)
- Plasma Medicine and Surgical Implants Center, School of Medicine, Tongji University, Shanghai 200070, China
| | - Lei Hou
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology & Center for Advanced Low-Dimension Materials, Donghua University, Shanghai 200051, China; (X.L.); (L.H.)
| |
Collapse
|
24
|
den Hollander P, Maddela JJ, Mani SA. Spatial and Temporal Relationship between Epithelial-Mesenchymal Transition (EMT) and Stem Cells in Cancer. Clin Chem 2024; 70:190-205. [PMID: 38175600 PMCID: PMC11246550 DOI: 10.1093/clinchem/hvad197] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 11/02/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Epithelial-mesenchymal transition (EMT) is often linked with carcinogenesis. However, EMT is also important for embryo development and only reactivates in cancer. Connecting how EMT occurs during embryonic development and in cancer could help us further understand the root mechanisms of cancer diseases. CONTENT There are key regulatory elements that contribute to EMT and the induction and maintenance of stem cell properties during embryogenesis, tissue regeneration, and carcinogenesis. Here, we explore the implications of EMT in the different stages of embryogenesis and tissue development. We especially highlight the necessity of EMT in the mesodermal formation and in neural crest cells. Through EMT, these cells gain epithelial-mesenchymal plasticity (EMP). With this transition, crucial morphological changes occur to progress through the metastatic cascade as well as tissue regeneration after an injury. Stem-like cells, including cancer stem cells, are generated from EMT and during this process upregulate factors necessary for stem cell maintenance. Hence, it is important to understand the key regulators allowing stem cell awakening in cancer, which increases plasticity and promotes treatment resistance, to develop strategies targeting this cell population and improve patient outcomes. SUMMARY EMT involves multifaceted regulation to allow the fluidity needed to facilitate adaptation. This regulatory mechanism, plasticity, involves many cooperating transcription factors. Additionally, posttranslational modifications, such as splicing, activate the correct isoforms for either epithelial or mesenchymal specificity. Moreover, epigenetic regulation also occurs, such as acetylation and methylation. Downstream signaling ultimately results in the EMT which promotes tissue generation/regeneration and cancer progression.
Collapse
Affiliation(s)
- Petra den Hollander
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI, United States
- Department of Pathology and Lab Medicine, The Warren Alpert Medical School, Brown University, Providence, RI, United States
| | - Joanna Joyce Maddela
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI, United States
- Department of Pathology and Lab Medicine, The Warren Alpert Medical School, Brown University, Providence, RI, United States
| | - Sendurai A Mani
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI, United States
- Department of Pathology and Lab Medicine, The Warren Alpert Medical School, Brown University, Providence, RI, United States
| |
Collapse
|
25
|
Jiang Y, Li S, Shi R, Yin W, Lv W, Tian T, Lin Y. A Novel Bioswitchable miRNA Mimic Delivery System: Therapeutic Strategies Upgraded from Tetrahedral Framework Nucleic Acid System for Fibrotic Disease Treatment and Pyroptosis Pathway Inhibition. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305622. [PMID: 37984862 PMCID: PMC10767442 DOI: 10.1002/advs.202305622] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/16/2023] [Indexed: 11/22/2023]
Abstract
There has been considerable interest in gene vectors and their role in regulating cellular activities and treating diseases since the advent of nucleic acid drugs. MicroRNA (miR) therapeutic strategies are research hotspots as they regulate gene expression post-transcriptionally and treat a range of diseases. An original tetrahedral framework nucleic acid (tFNA) analog, a bioswitchable miR inhibitor delivery system (BiRDS) carrying miR inhibitors, is previously established; however, it remains unknown whether BiRDS can be equipped with miR mimics. Taking advantage of the transport capacity of tetrahedral framework nucleic acid (tFNA) and upgrading it further, the treatment outcomes of a traditional tFNA and BiRDS at different concentrations on TGF-β- and bleomycin-induced fibrosis simultaneously in vitro and in vivo are compared. An upgraded traditional tFNA is designed by successfully synthesizing a novel BiRDS, carrying a miR mimic, miR-27a, for treating skin fibrosis and inhibiting the pyroptosis pathway, which exhibits stability and biocompatibility. BiRDS has three times higher efficiency in delivering miRNAs than the conventional tFNA with sticky ends. Moreover, BiRDS is more potent against fibrosis and pyroptosis-related diseases than tFNAs. These findings indicate that the BiRDS can be applied as a drug delivery system for disease treatment.
Collapse
Affiliation(s)
- Yueying Jiang
- State Key Laboratory of Oral DiseasesNational Center for StomatologyNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduSichuan610041China
| | - Songhang Li
- State Key Laboratory of Oral DiseasesNational Center for StomatologyNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduSichuan610041China
| | - Ruijianghan Shi
- State Key Laboratory of Oral DiseasesNational Center for StomatologyNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduSichuan610041China
| | - Wumeng Yin
- State Key Laboratory of Oral DiseasesNational Center for StomatologyNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduSichuan610041China
| | - Weitong Lv
- State Key Laboratory of Oral DiseasesNational Center for StomatologyNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduSichuan610041China
| | - Taoran Tian
- State Key Laboratory of Oral DiseasesNational Center for StomatologyNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduSichuan610041China
| | - Yunfeng Lin
- State Key Laboratory of Oral DiseasesNational Center for StomatologyNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduSichuan610041China
- Sichuan Provincial Engineering Research Center of Oral BiomaterialsSichuan UniversityChengduSichuan610041China
| |
Collapse
|
26
|
Tekin C, Ercelik M, Dunaev P, Galembikova A, Tezcan G, Aksoy SA, Budak F, Isık O, Ugras N, Boichuk S, Tunca B. Leaf Extract from European Olive (Olea europaea L.) Post-Transcriptionally Suppresses the Epithelial-Mesenchymal Transition and Sensitizes Gastric Cancer Cells to Chemotherapy. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:97-115. [PMID: 38467548 DOI: 10.1134/s0006297924010061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/21/2023] [Accepted: 11/30/2023] [Indexed: 03/13/2024]
Abstract
The overall survival of patients with the advanced and recurrent gastric cancer (GC) remains unfavorable. In particular, this is due to cancer spreading and resistance to chemotherapy associated with the epithelial-mesenchymal transition (EMT) of tumor cells. EMT can be identified by the transcriptome profiling of GC for EMT markers. Indeed, analysis of the TCGA and GTEx databases (n = 408) and a cohort of GC patients (n = 43) revealed that expression of the CDH2 gene was significantly decreased in the tumors vs. non-tumor tissues and correlated with the overall survival of GC patients. Expression of the EMT-promoting transcription factors SNAIL and ZEB1 was significantly increased in GC. These data suggest that targeting the EMT might be an attractive therapeutic approach for patients with GC. Previously, we demonstrated a potent anti-cancer activity of the olive leaf extract (OLE). However, its effect on the EMT regulation in GC remained unknown. Here, we showed that OLE efficiently potentiated the inhibitory effect of the chemotherapeutic agents 5-fluorouracil (5-FU) and cisplatin (Cis) on the EMT and their pro-apoptotic activity, as was demonstrated by changes in the expression of the EMT markers (E- and N-cadherins, vimentin, claudin-1) in GC cells treated with the aforementioned chemotherapeutic agents in the presence of OLE. Thus, culturing GC cells with 5-FU + OLE or Cis + OLE attenuated the invasive properties of cancer cells. Importantly, upregulation of expression of the apoptotic markers (PARP cleaved form) and increase in the number of cells undergoing apoptosis (annexin V-positive) were observed for GC cells treated with a combination of OLE and 5-FU or Cis. Collectively, our data illustrate that OLE efficiently interferes with the EMT in GC cells and potentiates the pro-apoptotic activity of certain chemotherapeutic agents used for GC therapy.
Collapse
Affiliation(s)
- Cagla Tekin
- Department of Medical Biology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Melis Ercelik
- Department of Medical Biology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Pavel Dunaev
- Department of Pathology, Kazan State Medical University, Kazan, Russia
| | - Aigul Galembikova
- Department of Pathology, Kazan State Medical University, Kazan, Russia
| | - Gulcin Tezcan
- Department of Fundamental Sciences, Faculty of Dentistry, Bursa Uludag University, Bursa, Turkey
| | - Secil Ak Aksoy
- Inegol Vocation School, Bursa Uludag University, Bursa, Turkey
- Experimental Animal Breeding and Research Unit, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Ferah Budak
- Department of Immunology, Medical Faculty, Bursa Uludag University Bursa, Turkey
| | - Ozgen Isık
- Department of General Surgery, Medical Faculty, Bursa Uludag University Bursa, Turkey
| | - Nesrin Ugras
- Department of Pathology, Medical Faculty, Bursa Uludag University, Bursa, Turkey
| | - Sergei Boichuk
- Department of Pathology, Kazan State Medical University, Kazan, Russia.
- Department of Radiotherapy and Radiology, Russian Medical Academy of Continuous Professional Education, Moscow, Russia
- "Biomarker" Research Laboratory, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Berrin Tunca
- Department of Medical Biology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey.
| |
Collapse
|
27
|
Silveira DA, Gupta S, da Cunha Jaeger M, Brunetto de Farias C, Mombach JCM, Sinigaglia M. A logical model of Ewing sarcoma cell epithelial-to-mesenchymal transition supports the existence of hybrid cellular phenotypes. FEBS Lett 2023; 597:2446-2460. [PMID: 37597508 DOI: 10.1002/1873-3468.14724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 08/04/2023] [Indexed: 08/21/2023]
Abstract
Ewing sarcoma (ES) is a highly aggressive pediatric tumor driven by the RNA-binding protein EWS (EWS)/friend leukemia integration 1 transcription factor (FLI1) chimeric transcription factor, which is involved in epithelial-mesenchymal transition (EMT). EMT stabilizes a hybrid cell state, boosting metastatic potential and drug resistance. Nevertheless, the mechanisms underlying the maintenance of this hybrid phenotype in ES remain elusive. Our study proposes a logical EMT model for ES, highlighting zinc finger E-box-binding homeobox 2 (ZEB2), miR-145, and miR-200 circuits that maintain hybrid states. The model aligns with experimental findings and reveals a previously unknown circuit supporting the mesenchymal phenotype. These insights emphasize the role of ZEB2 in the maintenance of the hybrid state in ES.
Collapse
Affiliation(s)
- Daner A Silveira
- Children's Cancer Institute, Porto Alegre, Brazil
- National Science and Technology Institute for Children's Cancer Biology and Pediatric Oncology - INCT BioOncoPed, Porto Alegre, Brazil
| | | | - Mariane da Cunha Jaeger
- Children's Cancer Institute, Porto Alegre, Brazil
- National Science and Technology Institute for Children's Cancer Biology and Pediatric Oncology - INCT BioOncoPed, Porto Alegre, Brazil
| | - Caroline Brunetto de Farias
- Children's Cancer Institute, Porto Alegre, Brazil
- National Science and Technology Institute for Children's Cancer Biology and Pediatric Oncology - INCT BioOncoPed, Porto Alegre, Brazil
| | | | - Marialva Sinigaglia
- Children's Cancer Institute, Porto Alegre, Brazil
- National Science and Technology Institute for Children's Cancer Biology and Pediatric Oncology - INCT BioOncoPed, Porto Alegre, Brazil
| |
Collapse
|
28
|
Kshirsagar A, Doroshev SM, Gorelik A, Olender T, Sapir T, Tsuboi D, Rosenhek-Goldian I, Malitsky S, Itkin M, Argoetti A, Mandel-Gutfreund Y, Cohen SR, Hanna JH, Ulitsky I, Kaibuchi K, Reiner O. LIS1 RNA-binding orchestrates the mechanosensitive properties of embryonic stem cells in AGO2-dependent and independent ways. Nat Commun 2023; 14:3293. [PMID: 37280197 DOI: 10.1038/s41467-023-38797-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/15/2023] [Indexed: 06/08/2023] Open
Abstract
Lissencephaly-1 (LIS1) is associated with neurodevelopmental diseases and is known to regulate the molecular motor cytoplasmic dynein activity. Here we show that LIS1 is essential for the viability of mouse embryonic stem cells (mESCs), and it governs the physical properties of these cells. LIS1 dosage substantially affects gene expression, and we uncovered an unexpected interaction of LIS1 with RNA and RNA-binding proteins, most prominently the Argonaute complex. We demonstrate that LIS1 overexpression partially rescued the extracellular matrix (ECM) expression and mechanosensitive genes conferring stiffness to Argonaute null mESCs. Collectively, our data transforms the current perspective on the roles of LIS1 in post-transcriptional regulation underlying development and mechanosensitive processes.
Collapse
Affiliation(s)
- Aditya Kshirsagar
- Departments of Molecular Genetics and Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Svetlana Maslov Doroshev
- Departments of Molecular Genetics and Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Anna Gorelik
- Departments of Molecular Genetics and Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Tsviya Olender
- Departments of Molecular Genetics and Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Tamar Sapir
- Departments of Molecular Genetics and Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Daisuke Tsuboi
- International Center for Brain Science, Fujita Health University, Toyoake, Japan
| | - Irit Rosenhek-Goldian
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot, Israel
| | - Sergey Malitsky
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Maxim Itkin
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Amir Argoetti
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | | | - Sidney R Cohen
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot, Israel
| | - Jacob H Hanna
- Departments of Molecular Genetics and Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Igor Ulitsky
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Kozo Kaibuchi
- International Center for Brain Science, Fujita Health University, Toyoake, Japan
| | - Orly Reiner
- Departments of Molecular Genetics and Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
29
|
Wu DT, Diba M, Yang S, Freedman BR, Elosegui‐Artola A, Mooney DJ. Hydrogel viscoelasticity modulates migration and fusion of mesenchymal stem cell spheroids. Bioeng Transl Med 2023; 8:e10464. [PMID: 37206235 PMCID: PMC10189430 DOI: 10.1002/btm2.10464] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 10/17/2022] [Accepted: 11/23/2022] [Indexed: 08/01/2023] Open
Abstract
Multicellular spheroids made of stem cells can act as building blocks that fuse to capture complex aspects of native in vivo environments, but the effect of hydrogel viscoelasticity on cell migration from spheroids and their fusion remains largely unknown. Here, we investigated the effect of viscoelasticity on migration and fusion behavior of mesenchymal stem cell (MSC) spheroids using hydrogels with a similar elasticity but different stress relaxation profiles. Fast relaxing (FR) matrices were found to be significantly more permissive to cell migration and consequent fusion of MSC spheroids. Mechanistically, inhibition of ROCK and Rac1 pathways prevented cell migration. Moreover, the combination of biophysical and biochemical cues provided by fast relaxing hydrogels and platelet-derived growth factor (PDGF) supplementation, respectively, resulted in a synergistic enhancement of migration and fusion. Overall, these findings emphasize the important role of matrix viscoelasticity in tissue engineering and regenerative medicine strategies based on spheroids.
Collapse
Affiliation(s)
- David T. Wu
- Laboratory for Cell and Tissue Engineering, John A. Paulson School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute for Biologically Inspired Engineering, Harvard UniversityBostonMassachusettsUSA
- Department of Oral Medicine, Infection, and ImmunityHarvard School of Dental MedicineBostonMassachusettsUSA
| | - Mani Diba
- Laboratory for Cell and Tissue Engineering, John A. Paulson School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute for Biologically Inspired Engineering, Harvard UniversityBostonMassachusettsUSA
- Department of Dentistry‐Regenerative BiomaterialsRadboud Institute for Molecular Life Sciences, Radboud University Medical CenterNijmegenthe Netherlands
| | - Stephanie Yang
- Laboratory for Cell and Tissue Engineering, John A. Paulson School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Department of Oral Medicine, Infection, and ImmunityHarvard School of Dental MedicineBostonMassachusettsUSA
| | - Benjamin R. Freedman
- Laboratory for Cell and Tissue Engineering, John A. Paulson School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute for Biologically Inspired Engineering, Harvard UniversityBostonMassachusettsUSA
| | - Alberto Elosegui‐Artola
- Laboratory for Cell and Tissue Engineering, John A. Paulson School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute for Biologically Inspired Engineering, Harvard UniversityBostonMassachusettsUSA
- Present address:
Cell and Tissue Mechanobiology LaboratoryThe Francis Crick InstituteLondonUK
- Present address:
Department of PhysicsKing's College LondonLondonUK
| | - David J. Mooney
- Laboratory for Cell and Tissue Engineering, John A. Paulson School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute for Biologically Inspired Engineering, Harvard UniversityBostonMassachusettsUSA
| |
Collapse
|
30
|
Otte ML, Lama Tamang R, Papapanagiotou J, Ahmad R, Dhawan P, Singh AB. Mucosal healing and inflammatory bowel disease: Therapeutic implications and new targets. World J Gastroenterol 2023; 29:1157-1172. [PMID: 36926666 PMCID: PMC10011951 DOI: 10.3748/wjg.v29.i7.1157] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/16/2022] [Accepted: 02/14/2023] [Indexed: 02/21/2023] Open
Abstract
Mucosal healing (MH) is vital in maintaining homeostasis within the gut and protecting against injury and infections. Multiple factors and signaling pathways contribute in a dynamic and coordinated manner to maintain intestinal homeostasis and mucosal regeneration/repair. However, when intestinal homeostasis becomes chronically disturbed and an inflammatory immune response is constitutively active due to impairment of the intestinal epithelial barrier autoimmune disease results, particularly inflammatory bowel disease (IBD). Many proteins and signaling pathways become dysregulated or impaired during these pathological conditions, with the mechanisms of regulation just beginning to be understood. Consequently, there remains a relative lack of broadly effective therapeutics that can restore MH due to the complexity of both the disease and healing processes, so tissue damage in the gastrointestinal tract of patients, even those in clinical remission, persists. With increased understanding of the molecular mechanisms of IBD and MH, tissue damage from autoimmune disease may in the future be ameliorated by developing therapeutics that enhance the body’s own healing response. In this review, we introduce the concept of mucosal healing and its relevance in IBD as well as discuss the mechanisms of IBD and potential strategies for altering these processes and inducing MH.
Collapse
Affiliation(s)
- Megan Lynn Otte
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Raju Lama Tamang
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Julia Papapanagiotou
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Rizwan Ahmad
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Punita Dhawan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Amar B Singh
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, United States
| |
Collapse
|
31
|
Fan L, Li W, Jiang H. Circ_0000395 Promoted CRC Progression via Elevating MYH9 Expression by Sequestering miR-432-5p. Biochem Genet 2023; 61:116-137. [PMID: 35759156 DOI: 10.1007/s10528-022-10245-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 06/07/2022] [Indexed: 01/24/2023]
Abstract
Colorectal cancer (CRC) has been listed as the fourth deadly cancer. Circular RNA hsa_circRNA_001046, also termed as hsa_circ_0000395 (circ_0000395), has been shown to be upregulated in CRC. Nevertheless, the function of circ_0000395 in CRC progression is unclear. 42 CRC patients were enrolled in the study. Detection of circ_0000395 expression in tissues and cells was executed using real-time quantitative polymerase chain reaction (RT-qPCR). Evaluation of circ_0000395 function was performed using loss-of-function experiments in vitro and in vivo. The regulatory mechanism of circ_0000395 was predicted by bioinformatics analysis and validated by dual-luciferase reporter and RIP assays. Exosomes were isolated by ultracentrifugation and validated by western blotting, TEM, and NTA. Circ_0000395 was strongly expressed in CRC samples and cell lines. Also, circ_0000395 repressed CRC growth in mouse models in vivo and induced CRC cell apoptosis, restrained CRC cell proliferation, migration, invasion, and EMT in vitro. Mechanistically, circ_0000395 sequestered miR-432-5p to regulate MYH9 expression. Furthermore, miR-432-5p knockdown reversed circ_0000395 silencing-mediated effects on CRC cell malignant phenotypes. MYH9 overexpression counteracted the inhibiting effects of miR-432-5p upregulation on CRC cell malignant phenotypes. Additionally, CRC cells derived from exosomal circ_0000395 promoted cancer cell malignant phenotypes. Our findings demonstrated that circ_0000395 sequestered miR-432-5p to elevate MYH9 expression, resulting in facilitating CRC progression, manifesting a potential therapeutic target for CRC.
Collapse
Affiliation(s)
- Leilei Fan
- Department of Gastrointestinal Surgery, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, No.158 Wuyang Avenue, Enshi City, Hubei Province, China
| | - Weiwei Li
- Department of Gastrointestinal Surgery, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, No.158 Wuyang Avenue, Enshi City, Hubei Province, China
| | - Hongsheng Jiang
- Department of Gastrointestinal Surgery, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, No.158 Wuyang Avenue, Enshi City, Hubei Province, China.
| |
Collapse
|
32
|
Yu HJ, Shin JA, Cho SD. Inhibition of focal adhesion kinase/paxillin axis by caffeic acid phenethyl ester restrains aggressive behaviors of head and neck squamous cell carcinoma in vitro. Arch Oral Biol 2023; 146:105611. [PMID: 36577313 DOI: 10.1016/j.archoralbio.2022.105611] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/06/2022] [Accepted: 12/23/2022] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Caffeic acid phenethyl ester (CAPE), one of the components of propolis that is produced by honeybees, reportedly suppresses multiple diseases, including bacterial infection, inflammation, and cancer. We aimed to investigate the inhibitory effects of CAPE on epithelial-mesenchymal transition (EMT) status and aggressive behaviors of human head and neck squamous cell carcinoma (HNSCC) in vitro and the underlying signaling pathway. DESIGN To examine the cell growth and in vitro tumorigenic potential of HNSCC cells, cell viability and soft agar colony formation assays, respectively, were performed. Transwell migration and invasion assays were conducted to monitor HNSCC cells' aggressive behaviors. Western blotting and immunocytochemistry analyses were done to investigate the signaling pathway responsible for relieving EMT progression and HNSCC cell aggressiveness. RESULTS CAPE inhibited the in vitro tumorigenic potential of SNU-1041 cells stimulated by epidermal growth factor and suppressed the migratory and invasive capacities of SNU-1041 cells, irrespective of their cell proliferation state. CAPE was, at least partially, capable of inhibiting EMT progression by upregulating E-cadherin expression, which was accompanied by the reduction of phosphorylated focal adhesion kinase (FAK) and Paxillin. The inhibition of the FAK/Paxillin axis by PF-562271 was sufficient to alleviate the EMT progression through the induction of E-cadherin and aggressive behaviors of SNU-1041 cells. CONCLUSIONS CAPE has a therapeutic potential as an anti-metastatic drug candidate for HNSCC therapy targeting the FAK/Paxillin axis.
Collapse
Affiliation(s)
- Hyun-Ju Yu
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Republic of Korea
| | - Ji-Ae Shin
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Republic of Korea.
| | - Sung-Dae Cho
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Republic of Korea.
| |
Collapse
|
33
|
Stavrou A, Ortiz A, Costa M. Cadmium Activates EGFR/STAT5 Signaling to Overcome Calcium Chelation and Promote Epithelial to Mesenchymal Transition. Biomolecules 2023; 13:116. [PMID: 36671501 PMCID: PMC9855692 DOI: 10.3390/biom13010116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 12/31/2022] [Accepted: 01/04/2023] [Indexed: 01/10/2023] Open
Abstract
Cadmium (Cd) is a heavy metal found in cigarette smoke, as well as in air and drinking water due to agricultural and industrial activities, and it poses a health risk to the general population. Prolonged low-dose Cd exposure via inhalation or ingestion causes lung and kidney cancers in humans and in animal models. While high doses of Cd exposure are correlated with the occupational setting and are cytotoxic, low doses of Cd are mainly correlated with exposure in the general population and induce carcinogenesis. The mechanism by which Cd-exposed cells overcome calcium chelation and induce malignant transformation remains unclear. This study examines how cells exposed to low doses of Cd survive loss of E-cadherin cell-cell adhesion via activation of the epidermal growth factor receptor (EGFR) and signal transducer and activator of transcription 5 (STAT5), which work to upregulate genes associated with survival and proliferation. To demonstrate the role of Cd in EGFR/STAT5 activation, we exposed two epithelial cell lines, BEAS-2B and HEK293, to two different doses (0.4 µM and 1.6 µM) of Cadmium chloride hemipentahydrate (CdCl2·2.5H2O) that are environmentally relevant to levels of Cd found in food and cigarettes for 24 h (hours) and 9 weeks (wks). When comparing cells treated with Cd with control cells, the Cd treated cells exhibited faster proliferation; therefore, we studied activation of EGFR via the STAT5 pathway using immunofluorescence (IF) for protein expression and localization and, in addition, RT-qPCR to examine changes in EGFR/STAT5 inducible genes. Our results showed an increase in EGFR and phosphorylated EGFR (p-EGFR) protein, with 1.6 µM of Cadmium having the highest expression at both 24-hour (hr) and 9-week (wk) exposures. Moreover, the IF analysis also demonstrated an increase of STAT5 and phosphorylated STAT5 (pSTAT5) in both short-term and long-term exposure, with 0.4 µM having the highest expression at 24 h. Finally, via Western blot analysis, we showed that there was a dose-dependent decrease in E-cadherin protein expression and increased N-cadherin in cells treated with low doses of Cd. These data demonstrate that epithelial cells can overcome Cd-mediated toxicity via activation of EGFR pathway to induce cell proliferation and survival and promote epithelial to mesenchymal transition.
Collapse
Affiliation(s)
| | | | - Max Costa
- Department of Medicine, Division of Environmental Medicine, New York University Grossman School of Medicine, New York, NY 10010, USA
| |
Collapse
|
34
|
The effect of mesenchymal stem cells-derived exosomes on the prostate, bladder, and renal cancer cell lines. Sci Rep 2022; 12:20924. [PMID: 36463254 PMCID: PMC9719468 DOI: 10.1038/s41598-022-23204-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 10/26/2022] [Indexed: 12/07/2022] Open
Abstract
We aimed to explain the role of mesenchymal stem cells (MSC-exosomes) on gene expressions of epithelial to mesenchymal transition (EMT), angiogenesis, and apoptosis. Four different cell lines were employed, including ACHN, 5637, LNCaP, and PC3, as well-known representatives for renal, bladder, hormone-sensitive, and hormone-refractory prostate cancers, respectively. Cell lines were exposed to diverse concentrations of mesenchymal stem cells-derived exosomes to find IC50 values. Percentages of apoptotic cells were evaluated by Annexin/P.I. staining. Micro Culture Tetrazolium Test assessed proliferative inhibitory effect; and prostate biomarker (KLK2), EMT (E-cadherin and Snail), angiogenesis genes (VEGF-A/VEGF-C), apoptosis genes (BAX/BCL2, P53) and Osteopontin variants (OPNa/b, and c) mRNA levels were studied by realtime PCR method. All 5637, LNCaP, and PC3 following treatment with exosomes illustrated specific responses with changes in expression of different genes. The increased TP53 and decreased BCL2 expressions were seen in 5637, LNCaP, and PC3. In PC3, OPNb and OPNc have raised more than P53; in LNCap, the increase was in VEGF-c. In 5637 cells, more than TP53 and BCL2 changes, two other genes, VEGFa and B.A.X., have decreased, suggesting exosomes' anti-apoptotic and anti-angiogenic effects. The kidney tumor cell line saw no significant gene expression change in ten targeted genes. MSC-exosomes therapy has augmented some interesting antitumor effects on prostate, bladder, and kidney cancer cell lines. This effect which originates from exosomes' potency to persuade apoptosis and prevent the proliferation of cancer cells simultaneously, was more substantial in bladder cancer, moderate in prostate cancer, and mild in renal cancer.
Collapse
|
35
|
Funato N, Yanagisawa H. TBX1 targets the miR-200-ZEB2 axis to induce epithelial differentiation and inhibit stem cell properties. Sci Rep 2022; 12:20188. [PMID: 36418889 PMCID: PMC9684448 DOI: 10.1038/s41598-022-24604-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/17/2022] [Indexed: 11/25/2022] Open
Abstract
TBX1, which encodes a T-box transcription factor, is considered a candidate gene for DiGeorge syndrome, velocardiofacial syndrome, and conotruncal anomaly face syndrome. Transduction of TBX1 decreases cell proliferation in epithelial cancer cells and Tbx1 ablation induces epithelial proliferation during palatal development. Here, we report that TBX1 regulates stem cell properties and epithelial differentiation through the transcriptional activation of microRNAs. Stable expression of TBX1 induces microRNA-200 (miR-200), whose members repress the epithelial-to-mesenchymal transition and induce epithelial differentiation. TBX1 rescues ZEB2-dependent transcriptional inhibition of the miR-200b/200a/429 cluster, whose promoter region contains conserved overlapping cis-regulatory motifs of the ZEB-binding E-box and TBX-binding element. Consequently, TBX1 activates the expression of both miR-200 and stemness-inhibitor miR-203 to inhibit their common targets, BMI1 and ZEB2. Moreover, Tbx1 ablation affects the differentiation of the palatal epithelium and perturbs the expression of miR-200, miR-203, and their target genes. We propose that TBX1 links stem cell properties and epithelial differentiation by inducing miR-200 and miR-203. Thus, targeting of the ZEB2-miR-200 axis by TBX1 may have potential therapeutic implications in miR-200-associated tumors and cleft palate.
Collapse
Affiliation(s)
- Noriko Funato
- grid.265073.50000 0001 1014 9130Department of Signal Gene Regulation, Tokyo Medical and Dental University (TMDU), Tokyo, 113-8510 Japan ,grid.265073.50000 0001 1014 9130Research Core, Tokyo Medical and Dental University (TMDU), Tokyo, 113-8510 Japan
| | - Hiromi Yanagisawa
- grid.20515.330000 0001 2369 4728Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Ibaraki, 305-8577 Japan
| |
Collapse
|
36
|
Thanuthanakhun N, Kim MH, Kino-oka M. Cell Behavioral Dynamics as a Cue in Optimizing Culture Stabilization in the Bioprocessing of Pluripotent Stem Cells. Bioengineering (Basel) 2022; 9:669. [PMID: 36354580 PMCID: PMC9687444 DOI: 10.3390/bioengineering9110669] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 10/28/2022] [Accepted: 11/05/2022] [Indexed: 04/23/2024] Open
Abstract
Pluripotent stem cells (PSCs) are important for future regenerative medicine therapies. However, in the production of PSCs and derivatives, the control of culture-induced fluctuations in the outcome of cell quality remains challenging. A detailed mechanistic understanding of how PSC behaviors are altered in response to biomechanical microenvironments within a culture is necessary for rational bioprocessing optimization. In this review, we discuss recent insights into the role of cell behavioral and mechanical homeostasis in modulating the states and functions of PSCs during culture processes. We delineate promising ways to manipulate the culture variability through regulating cell behaviors using currently developed tools. Furthermore, we anticipate their potential implementation for designing a culture strategy based on the concept of Waddington's epigenetic landscape that may provide a feasible solution for tuning the culture quality and stability in the bioprocessing space.
Collapse
Affiliation(s)
- Naruchit Thanuthanakhun
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Mee-Hae Kim
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Masahiro Kino-oka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita 565-0871, Osaka, Japan
- Research Base for Cell Manufacturability, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita 565-0871, Osaka, Japan
| |
Collapse
|
37
|
Hazra R, Brine L, Garcia L, Benz B, Chirathivat N, Shen MM, Wilkinson JE, Lyons SK, Spector DL. Platr4 is an early embryonic lncRNA that exerts its function downstream on cardiogenic mesodermal lineage commitment. Dev Cell 2022; 57:2450-2468.e7. [PMID: 36347239 PMCID: PMC9680017 DOI: 10.1016/j.devcel.2022.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 08/22/2022] [Accepted: 10/07/2022] [Indexed: 11/09/2022]
Abstract
The mammalian genome encodes thousands of long non-coding RNAs (lncRNAs), many of which are developmentally regulated and differentially expressed across tissues, suggesting their potential roles in cellular differentiation. Despite this expression pattern, little is known about how lncRNAs influence lineage commitment at the molecular level. Here, we demonstrate that perturbation of an embryonic stem cell/early embryonic lncRNA, pluripotency-associated transcript 4 (Platr4), directly influences the specification of cardiac-mesoderm-lineage differentiation. We show that Platr4 acts as a molecular scaffold or chaperone interacting with the Hippo-signaling pathway molecules Yap and Tead4 to regulate the expression of a downstream target gene, Ctgf, which is crucial to the cardiac-lineage program. Importantly, Platr4 knockout mice exhibit myocardial atrophy and valve mucinous degeneration, which are both associated with reduced cardiac output and sudden heart failure. Together, our findings provide evidence that Platr4 is required in cardiac-lineage specification and adult heart function in mice.
Collapse
Affiliation(s)
- Rasmani Hazra
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Lily Brine
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Libia Garcia
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Brian Benz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Napon Chirathivat
- Departments of Medicine, Genetics and Development, Urology, and Systems Biology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Michael M Shen
- Departments of Medicine, Genetics and Development, Urology, and Systems Biology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA
| | | | - Scott K Lyons
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - David L Spector
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
38
|
Satala CB, Jung I, Kovacs Z, Stefan-Van Staden RI, Molnar C, Bara T, Patrichi AI, Gurzu S. V-set and immunoglobulin domain containing 1 (VSIG1) as an emerging target for epithelial-mesenchymal transition of gastric cancer. Sci Rep 2022; 12:16241. [PMID: 36171238 PMCID: PMC9519899 DOI: 10.1038/s41598-022-19883-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 09/06/2022] [Indexed: 11/09/2022] Open
Abstract
V-set and Immunoglobulin domain containing 1 (VSIG1) is a cell-cell adhesion molecule which role in the genesis and evolution of gastric cancer (GC) is not understood. Only three Medline-indexed papers have focused on the role of VSIG1 in GC. The clinicopathological features of 94 GCs were examined in association with immunohistochemical (IHC) patterns of VSIG1, E-cadherin, and β-catenin which were assessed in the tumor core (central) vs. invasive edge. Cases were classified depending on the VSIG1 expression: membrane/membrane in both core and invasive front; null/negative staining in both core and invasive front; and cases with translocational patterns: membrane core/cytoplasmic buds and cytoplasmic core/null buds. Most of the tumors showed null pattern (n = 54). Cases with translocational patterns (n = 20) were GCs with a high lymph node ratio value (≥ 0.26) and advanced Dukes-MAC-like stage. Of the 20 total cases, 9 showed membrane-to-nuclear translocation of β-catenin and loss of E-cadherin, as indicators of epithelial-mesenchymal transition. All cases with membrane/membrane pattern (n = 20) involved the distal stomach. The poorest overall survival was registered in patients with subcellular translocation of VSIG1, compared to those with either membrane/membrane or null patterns (p = 0.002). In GC, VSIG1 acts as an adhesion membrane protein but its membrane-cytoplasmic translocation can be an indicator of epithelial-mesenchymal transition due to cytoplasmic VSIG1-mediated activation of canonical Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Catalin-Bogdan Satala
- Department of Pathology, George Emil Palade University of Medicine, Pharmacy, Science and Technology, Targu Mures, Romania
| | - Ioan Jung
- Department of Pathology, George Emil Palade University of Medicine, Pharmacy, Science and Technology, Targu Mures, Romania
| | - Zsolt Kovacs
- Department of Biochemistry, George Emil Palade University of Medicine, Pharmacy, Science and Technology, Targu Mures, Romania
| | | | - Calin Molnar
- Department of Surgery, George Emil Palade University of Medicine, Pharmacy, Science and Technology, Targu Mures, Romania
| | - Tivadar Bara
- Department of Surgery, George Emil Palade University of Medicine, Pharmacy, Science and Technology, Targu Mures, Romania
| | - Andrei-Ionut Patrichi
- Department of Pathology, George Emil Palade University of Medicine, Pharmacy, Science and Technology, Targu Mures, Romania
- Research Center for Oncopathology and Translational Medicine (CCOMT), George Emil Palade University of Medicine, Pharmacy, Science and Technology, Targu Mures, Romania
| | - Simona Gurzu
- Department of Pathology, George Emil Palade University of Medicine, Pharmacy, Science and Technology, Targu Mures, Romania.
- Research Center for Oncopathology and Translational Medicine (CCOMT), George Emil Palade University of Medicine, Pharmacy, Science and Technology, Targu Mures, Romania.
| |
Collapse
|
39
|
Cessna H, Baritaki S, Zaravinos A, Bonavida B. The Role of RKIP in the Regulation of EMT in the Tumor Microenvironment. Cancers (Basel) 2022; 14:cancers14194596. [PMID: 36230521 PMCID: PMC9559516 DOI: 10.3390/cancers14194596] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/21/2022] [Accepted: 09/21/2022] [Indexed: 12/03/2022] Open
Abstract
Simple Summary Raf kinase inhibitor protein (RKIP) expression in cancer cells is significantly reduced and promoting cancer cells growth and invasiveness. Overexpresssion of RKIP has been reported to mediate pleiotropic anti-cancer activities including the inhibition of survival signaling pathways, sensitization to cell death by cytotoxic drugs, inhibition of invasion, EMT and metastasis. The molecular mechanism by which RKIP inhibits EMT is not clear. In this review, we have examined how RKIP inhibits the selected EMT gene products (Snail, vimentin, N-cadherin, laminin alpha) and found that it involves signaling cross-talks between RKIP and each of the EMT gene products. These findings were validated by bioinformatic analyses demonstrating in various human cancers a negative correlation between the expression of RKIP and the expression of the EMT gene products. These findings suggest that targeting RKIP induction in cancer cells will result in multiple hits by inhibiting tumor growth, metastasis and reversal of chemo-immuno resistance. Abstract The Raf Kinase Inhibitor Protein (RKIP) is a unique gene product that directly inhibits the Raf/Mek/Erk and NF-kB pathways in cancer cells and resulting in the inhibition of cell proliferation, viability, EMT, and metastasis. Additionally, RKIP is involved in the regulation of cancer cell resistance to both chemotherapy and immunotherapy. The low expression of RKIP expression in many cancer types is responsible, in part, for the pathogenesis of cancer and its multiple properties. The inhibition of EMT and metastasis by RKIP led to its classification as a tumor suppressor. However, the mechanism by which RKIP mediates its inhibitory effects on EMT and metastases was not clear. We have proposed that one mechanism involves the negative regulation by RKIP of the expression of various gene products that mediate the mesenchymal phenotype as well as the positive regulation of gene products that mediate the epithelial phenotype via signaling cross talks between RKIP and each gene product. We examined several EMT mesenchymal gene products such as Snail, vimentin, N-cadherin, laminin and EPCAM and epithelial gene products such as E-cadherin and laminin. We have found that indeed these negative and positive correlations were detected in the signaling cross-talks. In addition, we have also examined bioinformatic data sets on different human cancers and the findings corroborated, in large part, the findings observed in the signaling cross-talks with few exceptions in some cancer types. The overall findings support the underlying mechanism by which the tumor suppressor RKIP regulates the expression of gene products involved in EMT and metastasis. Hence, the development of agent that can selectively induce RKIP expression in cancers with low expressions should result in the activation of the pleiotropic anti-cancer activities of RKIP and resulting in multiple effects including inhibition of tumor cell proliferation, EMT, metastasis and sensitization of resistant tumor cells to respond to both chemotherapeutics and immunotherapeutics.
Collapse
Affiliation(s)
- Hannah Cessna
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine, Jonsson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Stavroula Baritaki
- Laboratory of Experimental Oncology, Division of Surgery, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Apostolos Zaravinos
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia 2404, Cyprus
- Basic and Translational Cancer Research Center (BTCRC), Cancer Genetics, Genomics and Systems Biology Laboratory, Nicosia 1516, Cyprus
| | - Benjamin Bonavida
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine, Jonsson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA 90095, USA
- Correspondence:
| |
Collapse
|
40
|
Abstract
Neural crest cells (NCCs) are a dynamic, multipotent, vertebrate-specific population of embryonic stem cells. These ectodermally-derived cells contribute to diverse tissue types in developing embryos including craniofacial bone and cartilage, the peripheral and enteric nervous systems and pigment cells, among a host of other cell types. Due to their contribution to a significant number of adult tissue types, the mechanisms that drive their formation, migration and differentiation are highly studied. NCCs have a unique ability to transition from tightly adherent epithelial cells to mesenchymal and migratory cells by altering their polarity, expression of cell-cell adhesion molecules and gaining invasive abilities. In this Review, we discuss classical and emerging factors driving NCC epithelial-to-mesenchymal transition and migration, highlighting the role of signaling and transcription factors, as well as novel modifying factors including chromatin remodelers, small RNAs and post-translational regulators, which control the availability and longevity of major NCC players.
Collapse
Affiliation(s)
| | - Crystal D. Rogers
- Department of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, 1089 Veterinary Medicine Drive, Davis, CA 95616, USA
| |
Collapse
|
41
|
FOXM1-CD44 Signaling Is Critical for the Acquisition of Regorafenib Resistance in Human Liver Cancer Cells. Int J Mol Sci 2022; 23:ijms23147782. [PMID: 35887129 PMCID: PMC9324640 DOI: 10.3390/ijms23147782] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 12/21/2022] Open
Abstract
Regorafenib is a multikinase inhibitor that was approved by the US Food and Drug administration in 2017. Cancer stem cells (CSCs) are a small subset of cancer-initiating cells that are thought to contribute to therapeutic resistance. The forkhead box protein M1 (FOXM1) plays an important role in the regulation of the stemness of CSCs and mediates resistance to chemotherapy. However, the relationship between FOXM1 and regorafenib resistance in liver cancer cells remains unknown. We found that regorafenib-resistant HepG2 clones overexpressed FOXM1 and various markers of CSCs. Patients with hepatocellular carcinoma also exhibited an upregulation of FOXM1 and resistance to regorafenib, which were correlated with a poor survival rate. We identified a close relationship between FOXM1 expression and regorafenib resistance, which was correlated with the survival of patients with hepatocellular carcinoma. Thus, a strategy that antagonizes FOXM1–CD44 signaling would enhance the therapeutic efficacy of regorafenib in these patients.
Collapse
|
42
|
Zhou Y, Qiu S, Kim JT, Lee SB, Park HJ, Son MJ, Lee HJ, Chen J. Garcinone C Suppresses Tumorsphere Formation and Invasiveness by Hedgehog/Gli1 Signaling in Colorectal Cancer Stem-like Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:7941-7952. [PMID: 35749593 DOI: 10.1021/acs.jafc.2c01891] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Hyperactivation of hedgehog signaling occurs in colorectal cancer stem-like cells (CSCs), a rare subpopulation, potentially involved in metastasis, chemotherapy resistance, and cancer relapse. Garcinone C, a xanthone isolated from mangosteen (Garcinia mangostana), suppresses colorectal cancer in vivo and in vitro by inhibiting Gli1-dependent noncanonical hedgehog signaling. Herein, we investigated the effect of garcinone C on cancer stemness and invasiveness in colorectal cancer; Gli1 was noted as pivotal in maintaining stemness and invasiveness in HCT116 and HT29 CSCs. Garcinone C inhibited the proliferation and self-renewal of HCT116 and HT29 CSCs. Colon cancer stemness markers such as CD44, CD133, ALDH1, and Nanog were significantly decreased by garcinone C. Computational studies showed that garcinone C showed a high affinity with the Gli1 protein ZF domain by forming hydrogen bonds with amino acid residues of ASP244, ARG223, and ASP216. Besides, MG132 blocked the effects of garcinone C on Gli1. Thus, garcinone C suppressed colorectal CSCs by binding to Gli1 and enhancing its degradation. MMP2 and MMP9 levels, invasive-related markers, were increased in HCT116 CSCs but decreased by garcinone C. E-cadherin level was reduced in HCT116 CSCs, while the presence of garcinone C was restored. Garcinone C inhibited the proliferation and invasiveness of colorectal CSCs by targeting Gli1-dependent Hh signaling. Garcinone C may be a potent natural agent against colorectal cancer relapse.
Collapse
Affiliation(s)
- Yimeng Zhou
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong 17546, South Korea
| | - Shuai Qiu
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong 17546, South Korea
| | - Jin Tae Kim
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong 17546, South Korea
| | - Seung Beom Lee
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong 17546, South Korea
| | - Ho Jin Park
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong 17546, South Korea
| | - Moon Jeong Son
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong 17546, South Korea
| | - Hong Jin Lee
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong 17546, South Korea
| | - Jing Chen
- Institute for Advanced and Applied Chemical Synthesis, Jinan University, Guangzhou 510632, China
| |
Collapse
|
43
|
Ivey A, Pratt H, Boone BA. Molecular pathogenesis and emerging targets of gastric adenocarcinoma. J Surg Oncol 2022; 125:1079-1095. [PMID: 35481910 PMCID: PMC9069999 DOI: 10.1002/jso.26874] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/15/2022] [Accepted: 03/19/2022] [Indexed: 12/24/2022]
Abstract
Gastric adenocarcinoma (GC) is a devastating disease and is the third leading cause of cancer deaths worldwide. This heterogeneous disease has several different classification systems that consider histological appearance and genomic alterations. Understanding the etiology of GC, including infection, hereditary conditions, and environmental factors, is of particular importance and is discussed in this review. To improve survival in GC, we also must improve our therapeutic strategies. Here, we discuss new targets that warrant further exploration.
Collapse
Affiliation(s)
- Abby Ivey
- Department of Cancer Cell Biology, West Virginia University Cancer Institute, West Virginia University, Morgantown, West Virginia, USA
| | - Hillary Pratt
- Department of Cancer Cell Biology, West Virginia University Cancer Institute, West Virginia University, Morgantown, West Virginia, USA
| | - Brian A Boone
- Department of Cancer Cell Biology, West Virginia University Cancer Institute, West Virginia University, Morgantown, West Virginia, USA
- Department of Surgery, Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
| |
Collapse
|
44
|
E-Cadherin Signaling in Salivary Gland Development and Autoimmunity. J Clin Med 2022; 11:jcm11082241. [PMID: 35456333 PMCID: PMC9031707 DOI: 10.3390/jcm11082241] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/04/2022] [Accepted: 04/15/2022] [Indexed: 12/12/2022] Open
Abstract
E-cadherin, the major epithelial cadherin, is located in regions of cell–cell contact known as adherens junctions. E-cadherin contributes to the maintenance of the epithelial integrity through homophylic interaction; the cytoplasmic tail of E-cadherin directly binds catenins, forming a dynamic complex that regulates several intracellular signal transduction pathways, including epithelial-to-mesenchymal transition (EMT). Recent progress uncovered a novel and critical role for this adhesion molecule in salivary gland (SG) development and in SG diseases. We summarize the structure and regulation of the E-cadherin gene and transcript in view of the role of this remarkable protein in SG morphogenesis, focusing, in the second part of the review, on altered E-cadherin expression in EMT-mediated SG autoimmunity.
Collapse
|
45
|
Guo S, Zhou J, Zhang L, Bao CH, Zhao JM, Gao YL, Wu P, Weng ZJ, Shi Y. Acupuncture and Moxibustion Inhibited Intestinal Epithelial-Mesenchymal Transition in Patients with Crohn's Disease Induced by TGF- β 1/Smad3/Snail Pathway: A Clinical Trial Study. Chin J Integr Med 2022; 28:823-832. [PMID: 35419729 DOI: 10.1007/s11655-022-2888-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2021] [Indexed: 11/03/2022]
Abstract
OBJECTIVE To explore whether acupuncture combined with moxibustion could inhibit epithelialmesenchymal transition in Crohn's disease by affecting the transforming growth factor β 1 (TGF- β 1)/Smad3/Snail pathway. METHODS Sixty-three patients with Crohn's disease were randomly divided into an observation group (31 cases) receiving moxibustion at 43 °C combined with acupuncture, and a control group (32 cases) receiving moxibustion at 37 °C combined with sham acupuncture using a random number table. Patients were treated for 12 weeks. Crohn's Disease Activity Index (CDAI) was used to evaluate disease activity. Hematoxylin-eosin staining and transmission electron microscopy were utilized to observe the morphological and ultrastructural changes. Immunohistochemistry was used to detect the expression of transforming growth factor β 1 (TGF-β 1), T β R1, T β R2, Smad3, Snail, E-cadherin and fibronectin in intestinal mucosal tissues. RESULTS The decrease of the CDAI score, morphological and ultrastructural changes were more significant in observation group. The expression levels of TGF- β 1, Tβ R2, Smad3, and Snail in the observation group were significantly lower than those before the treatment (P<0.05 or P<0.01). After treatment, the expression levels of TGF-β 1, TβR2, and Snail in the observation group were significantly lower than those in the control group (all P<0.05); compared with the control group, the expression of fibronectin in the observation group was significantly decreased, and the expression of E-cadherin was significantly increased (all P<0.05). CONCLUSIONS Moxibustion at 43 °C combined with acupuncture may suppress TGF-β 1/Smad3/Snail pathway-mediated epithelial-mesenchymal transition of intestinal epithelial cells in Crohn's disease patients by inhibiting the expression levels of TGF-β 1, Tβ R2, Smad3, and Snail. (Registration No. ChiCTR-IIR-16007751).
Collapse
Affiliation(s)
- Sen Guo
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jing Zhou
- Department of Acupuncture and Moxibustion, Shanghai Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200082, China
| | - Liang Zhang
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chun-Hui Bao
- Department of Acupuncture and Moxibustion, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China.,Key Laboratory of Acupuncture and Immunological Effects, Shanghai Research Institute of Acupuncture and Meridian, Shanghai, 200030, China
| | - Ji-Meng Zhao
- Department of Acupuncture and Moxibustion, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China.,Key Laboratory of Acupuncture and Immunological Effects, Shanghai Research Institute of Acupuncture and Meridian, Shanghai, 200030, China
| | - Yan-Ling Gao
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Pin Wu
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zhi-Jun Weng
- Department of Acupuncture and Moxibustion, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China.,Key Laboratory of Acupuncture and Immunological Effects, Shanghai Research Institute of Acupuncture and Meridian, Shanghai, 200030, China
| | - Yin Shi
- Department of Acupuncture and Moxibustion, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China. .,Outpatient Department, Shanghai Research Institute of Acupuncture and Meridian, Shanghai, 200030, China.
| |
Collapse
|
46
|
Liu J, Qian K, Qin Z, Alshehri MD, Li Q, Tai Y. Cloud computing-enabled IIOT system for neurosurgical simulation using augmented reality data access. Exp Eye Res 2022; 220:109085. [DOI: 10.1016/j.exer.2022.109085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 03/15/2022] [Accepted: 04/13/2022] [Indexed: 12/18/2022]
|
47
|
Lin T, Tao J, Chen Y, Zhang Y, Li F, Zhang Y, Han X, Zhao Z, Liu G, Li H. Selenium Deficiency Leads to Changes in Renal Fibrosis Marker Proteins and Wnt/β-Catenin Signaling Pathway Components. Biol Trace Elem Res 2022; 200:1127-1139. [PMID: 33895963 DOI: 10.1007/s12011-021-02730-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 04/18/2021] [Indexed: 01/03/2023]
Abstract
Renal fibrosis is the final result of the progression of chronic kidney disease (CKD) to end-stage renal disease (ESRD). Earlier studies confirmed that selenium (Se) displays a close association with kidney diseases. However, the correlation between Se and fibrosis has rarely been explored. Thus, this article mainly aimed to investigate the effect of Se deficiency on renal fibrosis and the Wnt/β-catenin signaling pathway. Twenty BALB/c mice were fed a diet containing 0.02-mg/kg Se (Se-deficient diet) or 0.18-mg/kg Se (standard diet) for 20 weeks. A human glomerular mesangial cell (HMC) cell line was transfected with lentiviral TRNAU1AP-shRNA vector to establish a stable Se deficiency model in vitro. As indicated in this study, the glutathione (GSH) content in the Se-deficient group displayed an obvious decline compared with that in the control group, whereas the content of malondialdehyde (MDA) was obviously elevated. The results of Masson staining showed fibrosis around the renal tubules, and the results of immunohistochemistry showed that the area of positive fibronectin expression increased. In the Se-deficient group, the levels of collagen I, collagen III, matrix metalloproteinase 9 (MMP9), and other fibrosis-related proteins changed significantly in vivo and in vitro. Compared with the control group, the TRNAU1AP-shRNA group showed markedly reduced cell proliferation and migration abilities. Our data indicate that Se deficiency can cause kidney damage and renal fibrosis. Furthermore, the Wnt pathway is critical for the development of tissue and organ fibrosis. The data of this study demonstrated that the expression of Wnt5a, β-catenin, and dishevelled 1 (Dvl-1) was significantly upregulated in the Se-deficient group. Therefore, the Wnt/β-catenin pathway may play an important role in renal fibrosis caused by Se deficiency.
Collapse
Affiliation(s)
- Tingting Lin
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Baojian Road 157, Nangang District, Harbin City, 150086, Heilongjiang, China
| | - Jiaqi Tao
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Baojian Road 157, Nangang District, Harbin City, 150086, Heilongjiang, China
| | - Ying Chen
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Baojian Road 157, Nangang District, Harbin City, 150086, Heilongjiang, China
| | - Yitong Zhang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Baojian Road 157, Nangang District, Harbin City, 150086, Heilongjiang, China
| | - Fenglan Li
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Baojian Road 157, Nangang District, Harbin City, 150086, Heilongjiang, China
| | - Yutong Zhang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Baojian Road 157, Nangang District, Harbin City, 150086, Heilongjiang, China
| | - Xueqing Han
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Baojian Road 157, Nangang District, Harbin City, 150086, Heilongjiang, China
| | - Zihui Zhao
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Baojian Road 157, Nangang District, Harbin City, 150086, Heilongjiang, China
| | - Guiyan Liu
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Baojian Road 157, Nangang District, Harbin City, 150086, Heilongjiang, China
| | - Hui Li
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Baojian Road 157, Nangang District, Harbin City, 150086, Heilongjiang, China.
| |
Collapse
|
48
|
Sağraç D, Şenkal S, Hayal TB, Şahin F, Çobandede Z, Doğan A. Surface coating materials regulates the attachment and differentiation of mouse embryonic stem cell derived embryoid bodies into mesoderm at culture conditions. Cytotechnology 2022; 74:293-307. [PMID: 35464166 PMCID: PMC8976036 DOI: 10.1007/s10616-022-00529-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 02/11/2022] [Indexed: 11/03/2022] Open
Abstract
Abstract Pluripotent stem cells as a promising cell source with unlimited proliferation and differentiation capacity hold great promise for cell-based therapies in regenerative medicine. Establishment of appropriate culture conditions might enable the control of cellular fate decision in cell culture. Transfer of three-dimensional (3D) embryoid bodies to two-dimensional (2D) monolayer culture systems for initiation of cell differentiation and specialization requires an adaptation of cells which can be managed by extracellular matrix (ECM) materials. Here we compare the characteristics of four different cell culture coating materials and their effect on attachment and differentiation of cells spreading from mouse embryonic stem cell (mESC) derived embryoid bodies (EBs) in mesoderm inducing culture conditions. Atomic force microscope (AFM) and scanning electron microscope (SEM) analysis along with Water Contact Angle technique were used to analyze physical properties of ECM materials and to evaluate cellular behavior on surfaces. Cell migration and differentiation were performed initially by using mesoderm inducing culture conditions and then three germ layer specification conditions. We investigated properties of coating materials such as roughness and wettability control cell attachment, migration and differentiation of mESCs. Matrigel-Gelatin combination is suitable for cell attachment and migration of cells spreading from 3D EBs followed by transfer onto coated surfaces. Matrigel-Gelatin coating enhanced differentiation of cells into mesoderm like cells via EMT process. Our data demonstrated that the Matrigel-Gelatin combination as a cell culture coating matrix might serve as a suitable platform to transfer EBs for differentiation and might influence pluripotent stem cell fate decision into mesoderm and further mesoderm derivative cell populations. Graphical abstract Supplementary Information The online version contains supplementary material available at 10.1007/s10616-022-00529-z.
Collapse
|
49
|
Zhou H, Zou J, Shao C, Zhou A, Yu J, Chen S, Xu C. Prolyl 4-hydroxylase subunit alpha 3 facilitates human colon cancer growth and metastasis through the TGF-β/Smad signaling pathway. Pathol Res Pract 2022; 230:153749. [PMID: 34959098 DOI: 10.1016/j.prp.2021.153749] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/21/2021] [Accepted: 12/21/2021] [Indexed: 02/07/2023]
Abstract
Prolyl 4-hydroxylase subunit alpha 3 (P4HA3) has been known to be associated with a variety of human cancers. However, the role of P4HA3 on colon cancer growth and metastasis is unclear. In this study, we investigated the effect of P4HA3 on the growth and metastasis of colon cancer and its possible molecular mechanism. First of all, we demonstrated that P4HA3 expression was greatly higher in cells and tissues of colon cancer than that in non-tumor tissues and cells, and the prognosis of patients who had higher P4HA3 was distinctively poorer than patients who had lower level of P4HA3. Second, it was shown that P4HA3 knockdown strongly inhibited the migration, proliferation and invasion ability of colon cancer cells. However, P4HA3 over-expression accelerated the abilities. Meanwhile, P4HA3 could promote subcutaneous tumorigenesis in nude mice in vivo. In addition, P4HA3 knockdown significantly decreased mesenchymal markers Vimentin, N-cadherin and Snail expression and increased epithelial marker E-cadherin expression. And conversely, over-expression of P4HA3 produced the opposite effects. In the current study, there was further evidence that down-regulating P4HA3 significantly reduced both TGF-β and its following molecules including p-Smad2 as well as p-Smad3. However, overexpression of P4HA3 showed the opposite effect. In conclusion, this study shows that P4HA3 promotes the human colon cancer growth and metastasis by affecting TGF-β/Smad signaling pathway. P4HA3 may become a new target for early diagnosis, treatment and prognosis assessment of colon cancer.
Collapse
Affiliation(s)
- Hailang Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, PR China; Department of Gastroenterology, Lianshui People's Hospital Affiliated to Kangda College of Nanjing Medical University, Huaian, Jiangsu 223400, PR China
| | - Junwei Zou
- Department of General Surgery, The Second Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241000, PR China
| | - Changjiang Shao
- Department of Gastroenterology, The Second People's Hospital of Lianyungang, Lianyungang, Jiangsu 222006, PR China
| | - Aijun Zhou
- Department of Gastroenterology, Lianshui People's Hospital Affiliated to Kangda College of Nanjing Medical University, Huaian, Jiangsu 223400, PR China
| | - Jiufeng Yu
- Department of Traditional Chinese Medicine, Lianshui People's Hospital Affiliated to Kangda College of Nanjing Medical University, Huaian, Jiangsu 223400, PR China
| | - Song Chen
- The Institute of Life Sciences, Jiangsu College of Nursing,Huaian, Jiangsu 223300, PR China
| | - Chunfang Xu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, PR China.
| |
Collapse
|
50
|
Kretzmann JA, Irving KL, Smith NM, Evans CW. Modulating gene expression in breast cancer via DNA secondary structure and the CRISPR toolbox. NAR Cancer 2022; 3:zcab048. [PMID: 34988459 PMCID: PMC8693572 DOI: 10.1093/narcan/zcab048] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/26/2021] [Accepted: 12/01/2021] [Indexed: 12/11/2022] Open
Abstract
Breast cancer is the most commonly diagnosed malignancy in women, and while the survival prognosis of patients with early-stage, non-metastatic disease is ∼75%, recurrence poses a significant risk and advanced and/or metastatic breast cancer is incurable. A distinctive feature of advanced breast cancer is an unstable genome and altered gene expression patterns that result in disease heterogeneity. Transcription factors represent a unique therapeutic opportunity in breast cancer, since they are known regulators of gene expression, including gene expression involved in differentiation and cell death, which are themselves often mutated or dysregulated in cancer. While transcription factors have traditionally been viewed as 'undruggable', progress has been made in the development of small-molecule therapeutics to target relevant protein-protein, protein-DNA and enzymatic active sites, with varying levels of success. However, non-traditional approaches such as epigenetic editing, transcriptional control via CRISPR/dCas9 systems, and gene regulation through non-canonical nucleic acid secondary structures represent new directions yet to be fully explored. Here, we discuss these new approaches and current limitations in light of new therapeutic opportunities for breast cancers.
Collapse
Affiliation(s)
- Jessica A Kretzmann
- Laboratory for Biomolecular Nanotechnology, Department of Physics, Technical University of Munich, Am Coulombwall 4a, 85748 Garching, Germany
| | - Kelly L Irving
- School of Molecular Sciences, The University of Western Australia, 35 Stirling Hwy, Crawley, WA 6009, Australia
| | - Nicole M Smith
- School of Molecular Sciences, The University of Western Australia, 35 Stirling Hwy, Crawley, WA 6009, Australia
| | - Cameron W Evans
- School of Molecular Sciences, The University of Western Australia, 35 Stirling Hwy, Crawley, WA 6009, Australia
| |
Collapse
|