1
|
Jin J, Wang M, Liu Y, Li W, Zhang X, Cheng Z. Mitochondrial permeability transition drives the expression, identification and validation of necrosis-related genes in prognostic risk models of hepatocellular carcinoma. Transl Cancer Res 2025; 14:1037-1052. [PMID: 40104732 PMCID: PMC11912029 DOI: 10.21037/tcr-24-1442] [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: 08/20/2024] [Accepted: 12/17/2024] [Indexed: 03/20/2025]
Abstract
Background Hepatocellular carcinoma (HCC) is a prevalent malignant tumor, and the current treatment methods exhibit various limitations. In recent years, the role of mitochondrial permeability transition-driven necrosis-related genes (MPT-DNRGs) in the pathogenesis and progression of severe diseases, particularly tumors, has garnered significant attention. This study aimed to identify new targets and concepts for MPT-DNRG-targeted therapy in HCC. Methods In this study, we utilized HCC-related datasets and MPT-DNRGs to identify differentially expressed genes (DEGs) between HCC patients and control groups. By conducting a cross-analysis of the results of DEGs and MPT-DNRGs, we screened candidate genes. Subsequently, univariate Cox regression and least absolute shrinkage and selection operator (LASSO) regression analysis methods were employed to identify prognostic genes, which were used to construct a risk model and calculate individual risk scores for HCC patients. Additionally, we performed univariate and multivariate Cox regression analyses to identify independent prognostic factors and constructed a column chart based on these factors to predict the survival probability of HCC patients. Furthermore, gene set enrichment analysis (GSEA), the immune microenvironment, chemotherapy drugs, and the expression of prognostic genes between the two groups were analyzed. Finally, the expression of these prognostic genes was further confirmed using reverse transcription-quantitative polymerase chain reaction (RT-qPCR) technology. Results In this study, we identified 8,515 DEGs between HCC and control samples. By performing intersection analysis between DEGs and MPT-DNRGs, we pinpointed 15 candidate genes. Subsequently, through univariate Cox regression and LASSO regression analysis, we identified six genes (LMNB2, LMNB1, BAK1, CASP7, LMNA, and AKT1) that were significantly associated with overall survival (OS) in patients. Based on the median risk score, we categorized HCC patients into high-risk and low-risk groups. Kaplan-Meier (KM) survival analysis results demonstrated a significant difference in OS between the two groups, which was further validated through additional assessment. Furthermore, we constructed a nomogram to predict the survival probability of HCC patients. Moreover, GSEA revealed a crucial correlation between these genes and HCC, and highlighted a close association between risk scores and regulatory T cells. We also identified four chemotherapy drugs related to HCC. Finally, in both the training and validation cohorts, LMNB2, LMNB1, and LMNA exhibited high expression levels in tumor samples. Further validation using RT-qPCR confirmed that the expression of all prognostic genes was significantly higher in HCC group compared to the control group. Conclusions This study explored six prognostic genes (LMNB2, LMNB1, BAK1, CASP7, LMNA and AKT1) associated with MPT-DNRGs in HCC, which provides a reference for further research on HCC.
Collapse
Affiliation(s)
- Jiaxuan Jin
- Department of Gastroenterology, The First Affiliated Hospital of Jiamusi University, Jiamusi, China
- Department of Gastroenterology, Jiamusi Central Hospital, Jiamusi, China
| | - Mengyuan Wang
- Department of Gastroenterology, The First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Yinuo Liu
- Department of Gastroenterology, The First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Wei Li
- Department of Gastroenterology, The First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Xuemei Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Zhuoxin Cheng
- Department of Gastroenterology, The First Affiliated Hospital of Jiamusi University, Jiamusi, China
| |
Collapse
|
2
|
Zhao J, Zhang H, Pan C, He Q, Zheng K, Tang Y. Advances in research on the relationship between the LMNA gene and human diseases (Review). Mol Med Rep 2024; 30:236. [PMID: 39422026 PMCID: PMC11529173 DOI: 10.3892/mmr.2024.13358] [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: 06/18/2024] [Accepted: 08/29/2024] [Indexed: 10/19/2024] Open
Abstract
The LMNA gene, which is responsible for encoding lamin A/C proteins, is recognized as a primary constituent of the nuclear lamina. This protein serves crucial roles in various cellular physiological activities, including the maintenance of cellular structural stability, regulation of gene expression, mechanosensing and cellular motility. A significant association has been established between the LMNA gene and several major human diseases. Mutations, dysregulated expression of the LMNA gene, and improper processing of its encoded protein can result in a spectrum of pathological conditions. These diseases, collectively termed laminopathies, are directly attributed to LMNA gene dysfunction. The present review examines the recent advancements in research concerning the LMNA gene and its association with human diseases, while exploring its pathological roles. Particular emphasis is placed on the current status of LMNA gene research in the context of tumors. This includes an analysis of the abundance of LMNA alterations in cancer and its interplay with various signaling pathways. The aim of the present review was to provide novel perspectives for studying the development of LMNA‑related diseases and additional theoretical insights for basic and clinical translational research in this field.
Collapse
Affiliation(s)
- Jiumei Zhao
- Department of Laboratory, Chongqing Nanchuan District People's Hospital, Chongqing Medical University, Chongqing 408400, P.R. China
| | - Huijuan Zhang
- Forensic Science Centre, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Chenglong Pan
- Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Qian He
- School of Biomedical Engineering, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Kepu Zheng
- Forensic Science Centre, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Yu Tang
- Department of Pathology, The Third Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
3
|
Singh H, Gonzalez-Juarbe N, Pieper R, Yu Y, Vashee S. Predictive biomarkers for latent Mycobacterium tuberculosis infection. Tuberculosis (Edinb) 2024; 147:102399. [PMID: 37648595 PMCID: PMC10891298 DOI: 10.1016/j.tube.2023.102399] [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: 06/14/2023] [Revised: 08/16/2023] [Accepted: 08/23/2023] [Indexed: 09/01/2023]
Abstract
Tuberculosis is a leading cause of infectious death worldwide, with almost a fourth of the world's population latently infected with its causative agent, Mycobacterium tuberculosis. Current diagnostic methods are insufficient to differentiate between healthy and latently infected populations. Here, we used a machine learning approach to analyze publicly available proteomic data from saliva and serum in Ethiopia's healthy, latent TB (LTBI) and active TB (ATBI) people. Our analysis discovered a profile of six proteins, Mast Cell Expressed Membrane Protein-1, Hemopexin, Lamin A/C, Small Proline Rich Protein 2F, Immunoglobulin Kappa Variable 4-1, and Voltage Dependent Anion Channel 2 that can precisely differentiate between the healthy and latently infected populations. This data suggests that a combination of six host proteins can serve as accurate biomarkers to diagnose latent infection. This is important for populations living in high-risk areas as it may help in the surveillance and prevention of severe disease.
Collapse
Affiliation(s)
- Harinder Singh
- Infectious Diseases and Genomic Medicine Group, J Craig Venter Institute, 9605 Medical Center Drive Suite 150, Rockville, MD, USA.
| | - Norberto Gonzalez-Juarbe
- Infectious Diseases and Genomic Medicine Group, J Craig Venter Institute, 9605 Medical Center Drive Suite 150, Rockville, MD, USA
| | - Rembert Pieper
- Infectious Diseases and Genomic Medicine Group, J Craig Venter Institute, 9605 Medical Center Drive Suite 150, Rockville, MD, USA
| | - Yanbao Yu
- Infectious Diseases and Genomic Medicine Group, J Craig Venter Institute, 9605 Medical Center Drive Suite 150, Rockville, MD, USA
| | - Sanjay Vashee
- Infectious Diseases and Genomic Medicine Group, J Craig Venter Institute, 9605 Medical Center Drive Suite 150, Rockville, MD, USA
| |
Collapse
|
4
|
Chauhan R, Gupta A, Dagar G, Sharma S, Sadida HQ, Hashem S, Verghese AM, Tanwar M, Macha MA, Uddin S, Al-Shabeeb Akil AS, Pandita TK, Bhat AA, Singh M. Role of lamins in cellular physiology and cancer. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 143:119-153. [PMID: 39843134 DOI: 10.1016/bs.apcsb.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
Lamins, which are crucial type V intermediate filament proteins found in the nuclear lamina, are essential for maintaining the stability and function of the nucleus in higher vertebrates. They are classified into A- and B-types, and their distinct expression patterns contribute to cellular survival, development, and functionality. Lamins emerged during the transition from open to closed mitosis, with their complexity increasing alongside organism evolution. Derived from the LMNA, LMNB1, and LMNB2 genes, lamins undergo alternative splicing to produce seven variants, influencing cellular processes such as stiffness, chromatin condensation, and cell cycle regulation. The lamin network interacts with the cytoskeleton via Linkers of the nucleoskeleton to the cytoskeleton (LINC) complexes, playing a critical role in cellular stability and mechanotransduction. Lamins also regulate active transport into and out of the nucleus, affecting nuclear integrity, positioning, DNA maintenance, and gene expression. Genetic mutations in lamin genes lead to laminopathies, highlighting their functional significance and organizational roles. Changes in lamin subtype composition within the nuclear lamina have significant implications for cancer development, impacting cellular stiffness, mobility, and the Epithelial-to-Mesenchymal Transition (EMT). Lamin A/C, in particular, plays multifaceted roles in cancer biology, influencing progression, metastasis, and therapy response through interactions with various proteins and pathways. Dysregulated lamin expression is commonly observed in cancers, suggesting their potential as diagnostic and prognostic markers. This chapter underscores the pivotal roles of lamins in nuclear architecture and cancer biology, emphasizing their impact on cellular functions and disease pathology. Understanding lamin behavior and regulation mechanisms holds promise for developing novel diagnostic tools and targeted therapies in cancer treatment.
Collapse
Affiliation(s)
- Ravi Chauhan
- Department of Medical Oncology (Lab), Dr. B.R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Ashna Gupta
- Department of Medical Oncology (Lab), Dr. B.R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Gunjan Dagar
- Department of Medical Oncology (Lab), Dr. B.R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Shalini Sharma
- Department of Medical Oncology (Lab), Dr. B.R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Hana Q Sadida
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Sheema Hashem
- Department of Human Genetics, Sidra Medicine, Doha, Qatar
| | - Ann M Verghese
- Department of Human Genetics, Sidra Medicine, Doha, Qatar
| | - Mukesh Tanwar
- Department of Genetics, Maharishi Dayanand University Rohtak, Haryana, India
| | - Muzafar A Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Jammu and Kashmir, India
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Ammira S Al-Shabeeb Akil
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Tej K Pandita
- Center for Genomics and Precision Medicine, Texas A&M College of Medicine, Houston, TX, United States
| | - Ajaz A Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar.
| | - Mayank Singh
- Department of Medical Oncology (Lab), Dr. B.R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
5
|
Keuper K, Bartek J, Maya-Mendoza A. The nexus of nuclear envelope dynamics, circular economy and cancer cell pathophysiology. Eur J Cell Biol 2024; 103:151394. [PMID: 38340500 DOI: 10.1016/j.ejcb.2024.151394] [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: 10/29/2023] [Revised: 02/01/2024] [Accepted: 02/02/2024] [Indexed: 02/12/2024] Open
Abstract
The nuclear envelope (NE) is a critical component in maintaining the function and structure of the eukaryotic nucleus. The NE and lamina are disassembled during each cell cycle to enable an open mitosis. Nuclear architecture construction and deconstruction is a prime example of a circular economy, as it fulfills a highly efficient recycling program bound to continuous assessment of the quality and functionality of the building blocks. Alterations in the nuclear dynamics and lamina structure have emerged as important contributors to both oncogenic transformation and cancer progression. However, the knowledge of the NE breakdown and reassembly is still limited to a fraction of participating proteins and complexes. As cancer cells contain highly diverse nuclei in terms of DNA content, but also in terms of nuclear number, size, and shape, it is of great interest to understand the intricate relationship between these nuclear features in cancer cell pathophysiology. In this review, we provide insights into how those NE dynamics are regulated, and how lamina destabilization processes may alter the NE circular economy. Moreover, we expand the knowledge of the lamina-associated domain region by using strategic algorithms, including Artificial Intelligence, to infer protein associations, assess their function and location, and predict cancer-type specificity with implications for the future of cancer diagnosis, prognosis and treatment. Using this approach we identified NUP98 and MECP2 as potential proteins that exhibit upregulation in Acute Myeloid Leukemia (LAML) patients with implications for early diagnosis.
Collapse
Affiliation(s)
- Kristina Keuper
- DNA Replication and Cancer Group, Danish Cancer Institute, Copenhagen, Denmark; Genome Integrity Group, Danish Cancer Institute, Copenhagen, Denmark
| | - Jiri Bartek
- Genome Integrity Group, Danish Cancer Institute, Copenhagen, Denmark; Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SciLifeLab, Stockholm, Sweden
| | | |
Collapse
|
6
|
Alshaalan KS, Albawardi TK, Zhra M, Bin Sulaiman N, Jnied OY, Saleem RA, Aljada A. Differential Expression of LMNA/C and Insulin Receptor Transcript Variants in Peripheral Blood Mononuclear Cells of Leukemia Patients. J Clin Med 2024; 13:2568. [PMID: 38731097 PMCID: PMC11084221 DOI: 10.3390/jcm13092568] [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: 03/18/2024] [Revised: 04/16/2024] [Accepted: 04/21/2024] [Indexed: 05/13/2024] Open
Abstract
Background: Recent research has identified alternative transcript variants of LMNA/C (LMNA, LMNC, LMNAΔ10, and LMNAΔ50) and insulin receptors (INSRs) as potential biomarkers for various types of cancer. The objective of this study was to assess the expression of LMNA/C and INSR transcript variants in peripheral blood mononuclear cells (PBMCs) of leukemia patients to investigate their potential as diagnostic biomarkers. Methods: Quantitative TaqMan reverse transcriptase polymerase chain reaction (RT-qPCR) was utilized to quantify the mRNA levels of LMNA/C (LMNA, LMNC, LMNAΔ10, and LMNAΔ50) as well as INSR (IR-A and IR-B) variants in PBMCs obtained from healthy individuals (n = 32) and patients diagnosed with primary leukemias (acute myeloid leukemia (AML): n = 17; acute lymphoblastic leukemia (ALL): n = 8; chronic myeloid leukemia (CML): n = 5; and chronic lymphocytic leukemia (CLL): n = 15). Results: Only LMNA and LMNC transcripts were notably present in PBMCs. Both exhibited significantly decreased expression levels in leukemia patients compared to the healthy control group. Particularly, the LMNC:LMNA ratio was notably higher in AML patients. Interestingly, IR-B expression was not detectable in any of the PBMC samples, precluding the calculation of the IR-A:IR-B ratio as a diagnostic marker. Despite reduced expression across all types of leukemia, IR-A levels remained detectable, indicating its potential involvement in disease progression. Conclusions: This study highlights the distinct expression patterns of LMNA/C and INSR transcript variants in PBMCs of leukemia patients. The LMNC:LMNA ratio shows promise as a potential diagnostic indicator for AML, while further research is necessary to understand the role of IR-A in leukemia pathogenesis and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Khalid Saud Alshaalan
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia
| | - Turki Khalid Albawardi
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia
| | - Mahmoud Zhra
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (M.Z.)
| | - Norah Bin Sulaiman
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (M.Z.)
| | - Osama Yaheia Jnied
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (M.Z.)
| | - Rimah Abdullah Saleem
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (M.Z.)
| | - Ahmad Aljada
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (M.Z.)
| |
Collapse
|
7
|
Mishra J, Chakraborty S, Niharika, Roy A, Manna S, Baral T, Nandi P, Patra SK. Mechanotransduction and epigenetic modulations of chromatin: Role of mechanical signals in gene regulation. J Cell Biochem 2024; 125:e30531. [PMID: 38345428 DOI: 10.1002/jcb.30531] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 01/08/2024] [Accepted: 01/26/2024] [Indexed: 03/12/2024]
Abstract
Mechanical forces may be generated within a cell due to tissue stiffness, cytoskeletal reorganization, and the changes (even subtle) in the cell's physical surroundings. These changes of forces impose a mechanical tension within the intracellular protein network (both cytosolic and nuclear). Mechanical tension could be released by a series of protein-protein interactions often facilitated by membrane lipids, lectins and sugar molecules and thus generate a type of signal to drive cellular processes, including cell differentiation, polarity, growth, adhesion, movement, and survival. Recent experimental data have accentuated the molecular mechanism of this mechanical signal transduction pathway, dubbed mechanotransduction. Mechanosensitive proteins in the cell's plasma membrane discern the physical forces and channel the information to the cell interior. Cells respond to the message by altering their cytoskeletal arrangement and directly transmitting the signal to the nucleus through the connection of the cytoskeleton and nucleoskeleton before the information despatched to the nucleus by biochemical signaling pathways. Nuclear transmission of the force leads to the activation of chromatin modifiers and modulation of the epigenetic landscape, inducing chromatin reorganization and gene expression regulation; by the time chemical messengers (transcription factors) arrive into the nucleus. While significant research has been done on the role of mechanotransduction in tumor development and cancer progression/metastasis, the mechanistic basis of force-activated carcinogenesis is still enigmatic. Here, in this review, we have discussed the various cues and molecular connections to better comprehend the cellular mechanotransduction pathway, and we also explored the detailed role of some of the multiple players (proteins and macromolecular complexes) involved in mechanotransduction. Thus, we have described an avenue: how mechanical stress directs the epigenetic modifiers to modulate the epigenome of the cells and how aberrant stress leads to the cancer phenotype.
Collapse
Affiliation(s)
- Jagdish Mishra
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Subhajit Chakraborty
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Niharika
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Ankan Roy
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Soumen Manna
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Tirthankar Baral
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Piyasa Nandi
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Samir K Patra
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| |
Collapse
|
8
|
Singh S, Parthasarathi KTS, Bhat MY, Gopal C, Sharma J, Pandey A. Profiling Kinase Activities for Precision Oncology in Diffuse Gastric Cancer. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2024; 28:76-89. [PMID: 38271566 DOI: 10.1089/omi.2023.0173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Abstract
Gastric cancer (GC) remains a leading cause of cancer-related mortality globally. This is due to the fact that majority of the cases of GC are diagnosed at an advanced stage when the treatment options are limited and prognosis is poor. The diffuse subtype of gastric cancer (DGC) under Lauren's classification is more aggressive and usually occurs in younger patients than the intestinal subtype. The concept of personalized medicine is leading to the identification of multiple biomarkers in a large variety of cancers using different combinations of omics technologies. Proteomic changes including post-translational modifications are crucial in oncogenesis. We analyzed the phosphoproteome of DGC by using paired fresh frozen tumor and adjacent normal tissue from five patients diagnosed with DGC. We found proteins involved in the epithelial-to-mesenchymal transition (EMT), c-MYC pathway, and semaphorin pathways to be differentially phosphorylated in DGC tissues. We identified three kinases, namely, bromodomain adjacent to the zinc finger domain 1B (BAZ1B), WNK lysine-deficient protein kinase 1 (WNK1), and myosin light-chain kinase (MLCK) to be hyperphosphorylated, and one kinase, AP2-associated protein kinase 1 (AAK1), to be hypophosphorylated. LMNA hyperphosphorylation at serine 392 (S392) was demonstrated in DGC using immunohistochemistry. Importantly, we have detected heparin-binding growth factor (HDGF), heat shock protein 90 (HSP90), and FTH1 as potential therapeutic targets in DGC, as drugs targeting these proteins are currently under investigation in clinical trials. Although these new findings need to be replicated in larger study samples, they advance our understanding of signaling alterations in DGC, which could lead to potentially novel actionable targets in GC.
Collapse
Affiliation(s)
- Smrita Singh
- Manipal Academy of Higher Education (MAHE), Manipal, India
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- Center for Molecular Medicine, National Institute of Mental Health and Neuro-Sciences (NIMHANS), Bangalore, India
| | - K T Shreya Parthasarathi
- Manipal Academy of Higher Education (MAHE), Manipal, India
- Institute of Bioinformatics, International Technology Park, Bangalore, India
| | - Mohd Younis Bhat
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- Amrita School of Biotechnology, Amrita Vishwapeetham University, Kollam, India
| | - Champaka Gopal
- Department of Pathology, Kidwai Memorial Institute of Oncology, Bangalore, India
| | - Jyoti Sharma
- Manipal Academy of Higher Education (MAHE), Manipal, India
- Institute of Bioinformatics, International Technology Park, Bangalore, India
| | - Akhilesh Pandey
- Manipal Academy of Higher Education (MAHE), Manipal, India
- Center for Molecular Medicine, National Institute of Mental Health and Neuro-Sciences (NIMHANS), Bangalore, India
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
9
|
Das S, Dey MK, Devireddy R, Gartia MR. Biomarkers in Cancer Detection, Diagnosis, and Prognosis. SENSORS (BASEL, SWITZERLAND) 2023; 24:37. [PMID: 38202898 PMCID: PMC10780704 DOI: 10.3390/s24010037] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 11/27/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024]
Abstract
Biomarkers are vital in healthcare as they provide valuable insights into disease diagnosis, prognosis, treatment response, and personalized medicine. They serve as objective indicators, enabling early detection and intervention, leading to improved patient outcomes and reduced costs. Biomarkers also guide treatment decisions by predicting disease outcomes and facilitating individualized treatment plans. They play a role in monitoring disease progression, adjusting treatments, and detecting early signs of recurrence. Furthermore, biomarkers enhance drug development and clinical trials by identifying suitable patients and accelerating the approval process. In this review paper, we described a variety of biomarkers applicable for cancer detection and diagnosis, such as imaging-based diagnosis (CT, SPECT, MRI, and PET), blood-based biomarkers (proteins, genes, mRNA, and peptides), cell imaging-based diagnosis (needle biopsy and CTC), tissue imaging-based diagnosis (IHC), and genetic-based biomarkers (RNAseq, scRNAseq, and spatial transcriptomics).
Collapse
Affiliation(s)
| | | | | | - Manas Ranjan Gartia
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA; (S.D.); (M.K.D.); (R.D.)
| |
Collapse
|
10
|
Wallace M, Fedorchak GR, Agrawal R, Gilbert RM, Patel J, Park S, Paszek M, Lammerding J. The lamin A/C Ig-fold undergoes cell density-dependent changes that alter epitope binding. Nucleus 2023; 14:2180206. [PMID: 36809122 PMCID: PMC9980629 DOI: 10.1080/19491034.2023.2180206] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
Abstract
Lamins A/C are nuclear intermediate filament proteins that are involved in diverse cellular mechanical and biochemical functions. Here, we report that recognition of Lamins A/C by a commonly used antibody (JOL-2) that binds the Lamin A/C Ig-fold and other antibodies targeting similar epitopes is highly dependent on cell density, even though Lamin A/Clevels do not change. We propose that the effect is caused by partial unfolding or masking of the C'E and/or EF loops of the Ig-fold in response to cell spreading. Surprisingly, JOL-2 antibody labeling was insensitive to disruption of cytoskeletal filaments or the Linker of Nucleoskeleton and Cytoskeleton (LINC) complex. Furthermore, neither nuclear stiffness nor nucleo-cytoskeletal force transmission changed with cell density. These findings are important for the interpretation of immunofluorescence data for Lamin A/C and also raise the intriguing prospect that the conformational changes may play a role in Lamin A/C mediated cellular function.
Collapse
Affiliation(s)
- Melanie Wallace
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA,Weill Institute for Cell and Molecular Biology, Ithaca, NY, USA
| | - Gregory R. Fedorchak
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA,Weill Institute for Cell and Molecular Biology, Ithaca, NY, USA
| | - Richa Agrawal
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA,Weill Institute for Cell and Molecular Biology, Ithaca, NY, USA
| | - Rachel M. Gilbert
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA,Weill Institute for Cell and Molecular Biology, Ithaca, NY, USA
| | - Jineet Patel
- Weill Institute for Cell and Molecular Biology, Ithaca, NY, USA
| | - Sangwoo Park
- Graduate Field of Biophysics, Cornell University, Ithaca, NY, USA
| | - Matthew Paszek
- Graduate Field of Biophysics, Cornell University, Ithaca, NY, USA,Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, USA
| | - Jan Lammerding
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA,Weill Institute for Cell and Molecular Biology, Ithaca, NY, USA,CONTACT Jan Lammerding Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY14853, USA
| |
Collapse
|
11
|
Xin H, Tang Y, Jin YH, Li HL, Tian Y, Yu C, Zhao ZJ, Wu MS, Pan YF. Knockdown of LMNA inhibits Akt/β-catenin-mediated cell invasion and migration in clear cell renal cell carcinoma cells. Cell Adh Migr 2023; 17:1-14. [PMID: 37749865 PMCID: PMC10524799 DOI: 10.1080/19336918.2023.2260644] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 04/18/2023] [Indexed: 09/27/2023] Open
Abstract
The LMNA gene encoding lamin A/C is amplified in some clear cell renal cell carcinoma (ccRCC) samples. Our data showed that depletion of the tumor suppressor PBRM1 can upregulate lamin A/C levels, and lamin A/C could interact with PBRM1. However, the role of lamin A/C in ccRCC is not yet fully understood. Our functional assays showed that although the proliferation ability was slightly impaired after LMNA depletion, the migration and invasion of ccRCC cells were significantly inhibited. This suppression was accompanied by a reduction in MMP2, MMP9, AKT/p-AKT, and Wnt/β-catenin protein levels. Our data therefore suggest that lamin A/C, as an interaction partner of the tumor suppressor PBRM1, plays a crucial role in tumor invasion and metastasis in ccRCC.
Collapse
Affiliation(s)
- Hui Xin
- Department of Medical Genetics, Zunyi Medical University, Zunyi, Guizhou, China
- Key Laboratory of Gene Detection and Treatment in Guizhou Province, Zunyi, Guizhou, China
| | - Yu Tang
- Department of Medical Genetics, Zunyi Medical University, Zunyi, Guizhou, China
- Key Laboratory of Gene Detection and Treatment in Guizhou Province, Zunyi, Guizhou, China
| | - Yan-Hong Jin
- Department of Medical Genetics, Zunyi Medical University, Zunyi, Guizhou, China
- Key Laboratory of Gene Detection and Treatment in Guizhou Province, Zunyi, Guizhou, China
| | - Hu-Li Li
- Department of Medical Genetics, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yu Tian
- Department of Medical Genetics, Zunyi Medical University, Zunyi, Guizhou, China
| | - Cong Yu
- Department of Medical Genetics, Zunyi Medical University, Zunyi, Guizhou, China
| | - Ze-Ju Zhao
- Department of Urology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Ming-Song Wu
- Department of Medical Genetics, Zunyi Medical University, Zunyi, Guizhou, China
| | - You-Fu Pan
- Department of Medical Genetics, Zunyi Medical University, Zunyi, Guizhou, China
- Key Laboratory of Gene Detection and Treatment in Guizhou Province, Zunyi, Guizhou, China
| |
Collapse
|
12
|
Zheng Y, Zhong G, He C, Li M. Targeted splicing therapy: new strategies for colorectal cancer. Front Oncol 2023; 13:1222932. [PMID: 37664052 PMCID: PMC10470845 DOI: 10.3389/fonc.2023.1222932] [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: 05/15/2023] [Accepted: 08/07/2023] [Indexed: 09/05/2023] Open
Abstract
RNA splicing is the process of forming mature mRNA, which is an essential phase necessary for gene expression and controls many aspects of cell proliferation, survival, and differentiation. Abnormal gene-splicing events are closely related to the development of tumors, and the generation of oncogenic isoform in splicing can promote tumor progression. As a main process of tumor-specific splicing variants, alternative splicing (AS) can promote tumor progression by increasing the production of oncogenic splicing isoforms and/or reducing the production of normal splicing isoforms. This is the focus of current research on the regulation of aberrant tumor splicing. So far, AS has been found to be associated with various aspects of tumor biology, including cell proliferation and invasion, resistance to apoptosis, and sensitivity to different chemotherapeutic drugs. This article will review the abnormal splicing events in colorectal cancer (CRC), especially the tumor-associated splicing variants arising from AS, aiming to offer an insight into CRC-targeted splicing therapy.
Collapse
Affiliation(s)
| | | | - Chengcheng He
- Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | | |
Collapse
|
13
|
Schibler AC, Jevtic P, Pegoraro G, Levy DL, Misteli T. Identification of epigenetic modulators as determinants of nuclear size and shape. eLife 2023; 12:e80653. [PMID: 37219077 PMCID: PMC10259489 DOI: 10.7554/elife.80653] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 05/04/2023] [Indexed: 05/24/2023] Open
Abstract
The shape and size of the human cell nucleus is highly variable among cell types and tissues. Changes in nuclear morphology are associated with disease, including cancer, as well as with premature and normal aging. Despite the very fundamental nature of nuclear morphology, the cellular factors that determine nuclear shape and size are not well understood. To identify regulators of nuclear architecture in a systematic and unbiased fashion, we performed a high-throughput imaging-based siRNA screen targeting 867 nuclear proteins including chromatin-associated proteins, epigenetic regulators, and nuclear envelope components. Using multiple morphometric parameters, and eliminating cell cycle effectors, we identified a set of novel determinants of nuclear size and shape. Interestingly, most identified factors altered nuclear morphology without affecting the levels of lamin proteins, which are known prominent regulators of nuclear shape. In contrast, a major group of nuclear shape regulators were modifiers of repressive heterochromatin. Biochemical and molecular analysis uncovered a direct physical interaction of histone H3 with lamin A mediated via combinatorial histone modifications. Furthermore, disease-causing lamin A mutations that result in disruption of nuclear shape inhibited lamin A-histone H3 interactions. Oncogenic histone H3.3 mutants defective for H3K27 methylation resulted in nuclear morphology abnormalities. Altogether, our results represent a systematic exploration of cellular factors involved in determining nuclear morphology and they identify the interaction of lamin A with histone H3 as an important contributor to nuclear morphology in human cells.
Collapse
Affiliation(s)
| | - Predrag Jevtic
- Department of Molecular Biology, University of WyomingLaramieUnited States
| | - Gianluca Pegoraro
- High Throughput Imaging Facility (HiTIF), National Cancer Institute, NIHBethesdaUnited States
| | - Daniel L Levy
- Department of Molecular Biology, University of WyomingLaramieUnited States
| | - Tom Misteli
- National Cancer InstituteBethesdaUnited States
| |
Collapse
|
14
|
Kim HJ, Lee PCW, Hong JH. Overview of cellular homeostasis-associated nuclear envelope lamins and associated input signals. Front Cell Dev Biol 2023; 11:1173514. [PMID: 37250905 PMCID: PMC10213260 DOI: 10.3389/fcell.2023.1173514] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 05/03/2023] [Indexed: 05/31/2023] Open
Abstract
With the discovery of the role of the nuclear envelope protein lamin in human genetic diseases, further diverse roles of lamins have been elucidated. The roles of lamins have been addressed in cellular homeostasis including gene regulation, cell cycle, cellular senescence, adipogenesis, bone remodeling as well as modulation of cancer biology. Features of laminopathies line with oxidative stress-associated cellular senescence, differentiation, and longevity and share with downstream of aging-oxidative stress. Thus, in this review, we highlighted various roles of lamin as key molecule of nuclear maintenance, specially lamin-A/C, and mutated LMNA gene clearly reveal aging-related genetic phenotypes, such as enhanced differentiation, adipogenesis, and osteoporosis. The modulatory roles of lamin-A/C in stem cell differentiation, skin, cardiac regulation, and oncology have also been elucidated. In addition to recent advances in laminopathies, we highlighted for the first kinase-dependent nuclear lamin biology and recently developed modulatory mechanisms or effector signals of lamin regulation. Advanced knowledge of the lamin-A/C proteins as diverse signaling modulators might be biological key to unlocking the complex signaling of aging-related human diseases and homeostasis in cellular process.
Collapse
Affiliation(s)
- Hyeong Jae Kim
- Department of Physiology, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Peter C. W. Lee
- Lung Cancer Research Center, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, Republic of Korea
| | - Jeong Hee Hong
- Department of Physiology, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| |
Collapse
|
15
|
Liu S, Li Y, Hong Y, Wang M, Zhang H, Ma J, Qu K, Huang G, Lu TJ. Mechanotherapy in oncology: Targeting nuclear mechanics and mechanotransduction. Adv Drug Deliv Rev 2023; 194:114722. [PMID: 36738968 DOI: 10.1016/j.addr.2023.114722] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 12/23/2022] [Accepted: 01/28/2023] [Indexed: 02/05/2023]
Abstract
Mechanotherapy is proposed as a new option for cancer treatment. Increasing evidence suggests that characteristic differences are present in the nuclear mechanics and mechanotransduction of cancer cells compared with those of normal cells. Recent advances in understanding nuclear mechanics and mechanotransduction provide not only further insights into the process of malignant transformation but also useful references for developing new therapeutic approaches. Herein, we present an overview of the alterations of nuclear mechanics and mechanotransduction in cancer cells and highlight their implications in cancer mechanotherapy.
Collapse
Affiliation(s)
- Shaobao Liu
- State Key Laboratory of Mechanics and Control of Mechanical Structures, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, PR China; MIIT Key Laboratory of Multifunctional Lightweight Materials and Structures, Nanjing University of Aeronautics, Nanjing 210016, PR China
| | - Yuan Li
- MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Yuan Hong
- MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China; National Science Foundation Science and Technology Center for Engineering Mechanobiology, Washington University, St. Louis, MO 63130, USA
| | - Ming Wang
- MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Hao Zhang
- State Key Laboratory of Mechanics and Control of Mechanical Structures, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, PR China; MIIT Key Laboratory of Multifunctional Lightweight Materials and Structures, Nanjing University of Aeronautics, Nanjing 210016, PR China
| | - Jinlu Ma
- Department of Radiation Oncology, the First Affiliated Hospital, Xian Jiaotong University, Xi'an 710061, PR China
| | - Kai Qu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital, Xian Jiaotong University, Xi'an 710061, PR China
| | - Guoyou Huang
- Department of Engineering Mechanics, School of Civil Engineering, Wuhan University, Wuhan 430072, PR China.
| | - Tian Jian Lu
- State Key Laboratory of Mechanics and Control of Mechanical Structures, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, PR China; MIIT Key Laboratory of Multifunctional Lightweight Materials and Structures, Nanjing University of Aeronautics, Nanjing 210016, PR China.
| |
Collapse
|
16
|
Elevated Levels of Lamin A Promote HR and NHEJ-Mediated Repair Mechanisms in High-Grade Ovarian Serous Carcinoma Cell Line. Cells 2023; 12:cells12050757. [PMID: 36899893 PMCID: PMC10001195 DOI: 10.3390/cells12050757] [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: 01/05/2023] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 03/06/2023] Open
Abstract
Extensive research for the last two decades has significantly contributed to understanding the roles of lamins in the maintenance of nuclear architecture and genome organization which is drastically modified in neoplasia. It must be emphasized that alteration in lamin A/C expression and distribution is a consistent event during tumorigenesis of almost all tissues of human bodies. One of the important signatures of a cancer cell is its inability to repair DNA damage which befalls several genomic events that transform the cells to be sensitive to chemotherapeutic agents. This genomic and chromosomal instability is the most common feature found in cases of high-grade ovarian serous carcinoma. Here, we report elevated levels of lamins in OVCAR3 cells (high-grade ovarian serous carcinoma cell line) in comparison to IOSE (immortalised ovarian surface epithelial cells) and, consequently, altered damage repair machinery in OVCAR3. We have analysed the changes in global gene expression as a sequel to DNA damage induced by etoposide in ovarian carcinoma where lamin A is particularly elevated in expression and reported some differentially expressed genes associated with pathways conferring cellular proliferation and chemoresistance. We hereby establish the role of elevated lamin A in neoplastic transformation in the context of high-grade ovarian serous cancer through a combination of HR and NHEJ mechanisms.
Collapse
|
17
|
Xiang H, Shen X, Chen E, Chen W, Song Z. Construction and validation of a novel algorithm based on oncosis-related lncRNAs comprising the immune landscape and prediction of colorectal cancer prognosis. Oncol Lett 2023; 25:63. [PMID: 36644148 PMCID: PMC9827452 DOI: 10.3892/ol.2022.13650] [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: 07/20/2022] [Accepted: 12/01/2022] [Indexed: 12/25/2022] Open
Abstract
Colorectal cancer (CRC) has high morbidity and mortality, particularly if diagnosed at an advanced stage. Although there have been several studies on CRC, few have investigated the relationship between oncosis and CRC. Thus, the purpose of the present study was to identify oncosis-related long noncoding RNAs (lncRNAs) and to establish a clinical prognostic model. Original data were acquired from The Cancer Genome Atlas database and PubMed. Differentially expressed oncosis-related lncRNAs (DEorlncRNAs) were identified and were subsequently formed into pairs. Next, a series of tests and analyses, including both univariate and multivariate analyses, as well as Lasso and Cox regression analyses, were performed to establish a receiver operating characteristic curve. A cut-off point was subsequently used to divide the samples into groups labelled as high- or low-risk. Thus, a model was established and evaluated in several dimensions. Six pairs of DEorlncRNAs associated with prognosis according to the algorithm were screened out and the CRC cases were divided into high- and low-risk groups. Significant differences between patients in the different risk groups were observed for several traits, including survival outcomes, clinical pathology characteristics, immune cell infiltration status and drug sensitivity. In addition, PCR and flow cytometry were performed to further verify the model. In summary, a new risk model algorithm based on six pairs of DEorlncRNAs in CRC, which does not require specific data regarding the level of gene expression, was established and validated. This algorithm may be used to predict patient prognosis, immune cell infiltration and drug sensitivity.
Collapse
Affiliation(s)
- Haoyi Xiang
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital of Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
- Zhejiang University School of Medicine, Hangzhou, Zhejiang 310011, P.R. China
| | - Xuning Shen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital of Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
- Zhejiang University School of Medicine, Hangzhou, Zhejiang 310011, P.R. China
| | - Engeng Chen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital of Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | - Wei Chen
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| | - Zhangfa Song
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital of Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| |
Collapse
|
18
|
Scott NR, Parekh SH. A-type lamins involvement in transport and implications in cancer? Nucleus 2022; 13:221-235. [PMID: 36109835 PMCID: PMC9481127 DOI: 10.1080/19491034.2022.2118418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Nuclear lamins and transport are intrinsically linked, but their relationship is yet to be fully unraveled. A multitude of complex, coupled interactions between lamins and nucleoporins (Nups), which mediate active transport into and out of the nucleus, combined with well documented dysregulation of lamins in many cancers, suggests that lamins and nuclear transport may play a pivotal role in carcinogenesis and the preservation of cancer. Changes of function related to lamin/Nup activity can principally lead to DNA damage, further increasing the genetic diversity within a tumor, which could lead to the reduction the effectiveness of antineoplastic treatments. This review discusses and synthesizes different connections of lamins to nuclear transport and offers a number of outlook questions, the answers to which could reveal a new perspective on the connection of lamins to molecular transport of cancer therapeutics, in addition to their established role in nuclear mechanics.
Collapse
Affiliation(s)
- Nicholas R. Scott
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Sapun H. Parekh
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
19
|
Hiroe T, Moriya S, Kobayashi S, Nishijima Y, Watanabe A, Shirabe K, Ikota H, Yokoo H, Saio M. Negative correlation between the nuclear size and nuclear Lamina component Lamin A in intraductal papillary mucinous neoplasms of the pancreas. Pathol Oncol Res 2022; 28:1610684. [PMID: 36561231 PMCID: PMC9764245 DOI: 10.3389/pore.2022.1610684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022]
Abstract
Background: The nuclear laminar protein Lamin A and inner nuclear membrane protein Emerin plays important role in sustaining nuclear structure. However, They have not investigated the significance of these proteins for development of pancreatic intraductal papillary mucinous neoplasm (IPMN). Methods: We examined pancreatic IPMN specimens for nuclear morphology and nuclear protein expression pattern of Lamin A and Emerin. Forty-two IPMN specimens were included, with 30 classified as intraductal papillary mucinous adenoma (IPMA) and 12 as intraductal papillary mucinous carcinoma (IPMC). Results: Classification according to histological subtype revealed that 26 specimens were of the gastric subtype (1 IPMC case), 8 were pancreatobiliary (6 IPMC cases), 6 were intestinal (3 IPMC cases), and 2 were oncocytic (all cases were IPMC). The frequency of IPMN subtypes in this study seemed to agree with those in previous reports. We analyzed Feulgen staining sections for nuclear morphological analysis using computer-assisted image analysis. Nuclear area and perimeter were significantly larger in IPMC than in IPMA. Finally, we examined the positive ratios of Lamin A and Emerin in immunohistochemical staining sections by image analysis. We found a negative correlation between the nuclear size and Lamin A-positive ratio, which was significantly lower in IPMC than that in IPMA. However, no significant correlation was observed between nuclear size and Emerin expression was observed, and no differences were found in the Emerin-positive ratio between IPMA and IPMC. Conclusion: Our results suggest that a decreased Lamin A positive ratio induces nuclear enlargement in adenomas, which thereby induce promotion to carcinomas. Furthermore, Lamin A expression can be a reliable biomarker for distinguishing between IPMC and IPMA.
Collapse
Affiliation(s)
- Tamaki Hiroe
- Laboratory of Histopathology and Cytopathology, Department of Laboratory Sciences, Graduate School of Health Sciences, Gunma University, Maebashi, Gunma, Japan
| | - Shunichi Moriya
- Laboratory of Histopathology and Cytopathology, Department of Laboratory Sciences, Graduate School of Health Sciences, Gunma University, Maebashi, Gunma, Japan
| | - Sayaka Kobayashi
- Laboratory of Histopathology and Cytopathology, Department of Laboratory Sciences, Graduate School of Health Sciences, Gunma University, Maebashi, Gunma, Japan
| | - Yoshimi Nishijima
- Laboratory of Histopathology and Cytopathology, Department of Laboratory Sciences, Graduate School of Health Sciences, Gunma University, Maebashi, Gunma, Japan
| | - Akira Watanabe
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Gunma University, Maebashi, Gunma, Japan
| | - Ken Shirabe
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Gunma University, Maebashi, Gunma, Japan
| | - Hayato Ikota
- Clinical Department of Pathology, Gunma University Hospital, Maebashi, Gunma, Japan
| | - Hideaki Yokoo
- Department of Human Pathology, Graduate School of Medicine, Gunma University, Maebashi, Gunma, Japan
| | - Masanao Saio
- Laboratory of Histopathology and Cytopathology, Department of Laboratory Sciences, Graduate School of Health Sciences, Gunma University, Maebashi, Gunma, Japan,*Correspondence: Masanao Saio,
| |
Collapse
|
20
|
Schirmer EC, Latonen L, Tollis S. Nuclear size rectification: A potential new therapeutic approach to reduce metastasis in cancer. Front Cell Dev Biol 2022; 10:1022723. [PMID: 36299481 PMCID: PMC9589484 DOI: 10.3389/fcell.2022.1022723] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 09/12/2022] [Indexed: 03/07/2024] Open
Abstract
Research on metastasis has recently regained considerable interest with the hope that single cell technologies might reveal the most critical changes that support tumor spread. However, it is possible that part of the answer has been visible through the microscope for close to 200 years. Changes in nuclear size characteristically occur in many cancer types when the cells metastasize. This was initially discarded as contributing to the metastatic spread because, depending on tumor types, both increases and decreases in nuclear size could correlate with increased metastasis. However, recent work on nuclear mechanics and the connectivity between chromatin, the nucleoskeleton, and the cytoskeleton indicate that changes in this connectivity can have profound impacts on cell mobility and invasiveness. Critically, a recent study found that reversing tumor type-dependent nuclear size changes correlated with reduced cell migration and invasion. Accordingly, it seems appropriate to now revisit possible contributory roles of nuclear size changes to metastasis.
Collapse
Affiliation(s)
- Eric C. Schirmer
- Institute of Cell Biology, University of Edinburgh, Edinburgh, United Kingdom
| | - Leena Latonen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
- Foundation for the Finnish Cancer Institute, Helsinki, Finland
| | - Sylvain Tollis
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
21
|
Golloshi R, Playter C, Freeman TF, Das P, Raines TI, Garretson JH, Thurston D, McCord RP. Constricted migration is associated with stable 3D genome structure differences in cancer cells. EMBO Rep 2022; 23:e52149. [PMID: 35969179 PMCID: PMC9535800 DOI: 10.15252/embr.202052149] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 07/01/2022] [Accepted: 07/12/2022] [Indexed: 01/14/2023] Open
Abstract
To spread from a localized tumor, metastatic cancer cells must squeeze through constrictions that cause major nuclear deformations. Since chromosome structure affects nucleus stiffness, gene regulation, and DNA repair, here, we investigate the relationship between 3D genome structure and constricted migration in cancer cells. Using melanoma (A375) cells, we identify phenotypic differences in cells that have undergone multiple rounds of constricted migration. These cells display a stably higher migration efficiency, elongated morphology, and differences in the distribution of Lamin A/C and heterochromatin. Hi-C experiments reveal differences in chromosome spatial compartmentalization specific to cells that have passed through constrictions and related alterations in expression of genes associated with migration and metastasis. Certain features of the 3D genome structure changes, such as a loss of B compartment interaction strength, are consistently observed after constricted migration in clonal populations of A375 cells and in MDA-MB-231 breast cancer cells. Our observations suggest that consistent types of chromosome structure changes are induced or selected by passage through constrictions and that these may epigenetically encode stable differences in gene expression and cellular migration phenotype.
Collapse
Affiliation(s)
- Rosela Golloshi
- Biochemistry & Cellular and Molecular Biology DepartmentUniversity of TennesseeKnoxvilleTNUSA
| | - Christopher Playter
- Biochemistry & Cellular and Molecular Biology DepartmentUniversity of TennesseeKnoxvilleTNUSA
| | - Trevor F Freeman
- Biochemistry & Cellular and Molecular Biology DepartmentUniversity of TennesseeKnoxvilleTNUSA
| | - Priyojit Das
- UT‐ORNL Graduate School of Genome Science and TechnologyUniversity of TennesseeKnoxvilleTNUSA
| | - Thomas Isaac Raines
- Biochemistry & Cellular and Molecular Biology DepartmentUniversity of TennesseeKnoxvilleTNUSA
| | - Joshua H Garretson
- Biochemistry & Cellular and Molecular Biology DepartmentUniversity of TennesseeKnoxvilleTNUSA
| | - Delaney Thurston
- Biochemistry & Cellular and Molecular Biology DepartmentUniversity of TennesseeKnoxvilleTNUSA
| | - Rachel Patton McCord
- Biochemistry & Cellular and Molecular Biology DepartmentUniversity of TennesseeKnoxvilleTNUSA
| |
Collapse
|
22
|
La Torre M, Merigliano C, Maccaroni K, Chojnowski A, Goh WI, Giubettini M, Vernì F, Capanni C, Rhodes D, Wright G, Burke B, Soddu S, Burla R, Saggio I. Combined alteration of lamin and nuclear morphology influences the localization of the tumor-associated factor AKTIP. J Exp Clin Cancer Res 2022; 41:273. [PMID: 36096808 PMCID: PMC9469526 DOI: 10.1186/s13046-022-02480-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/30/2022] [Indexed: 11/10/2022] Open
Abstract
Background Lamins, key nuclear lamina components, have been proposed as candidate risk biomarkers in different types of cancer but their accuracy is still debated. AKTIP is a telomeric protein with the property of being enriched at the nuclear lamina. AKTIP has similarity with the tumor susceptibility gene TSG101. AKTIP deficiency generates genome instability and, in p53−/− mice, the reduction of the mouse counterpart of AKTIP induces the exacerbation of lymphomas. Here, we asked whether the distribution of AKTIP is altered in cancer cells and whether this is associated with alterations of lamins. Methods We performed super-resolution imaging, quantification of lamin expression and nuclear morphology on HeLa, MCF7, and A549 tumor cells, and on non-transformed fibroblasts from healthy donor and HGPS (LMNA c.1824C > T p.Gly608Gly) and EDMD2 (LMNA c.775 T > G) patients. As proof of principle model combining a defined lamin alteration with a tumor cell setting, we produced HeLa cells exogenously expressing the HGPS lamin mutant progerin that alters nuclear morphology. Results In HeLa cells, AKTIP locates at less than 0.5 µm from the nuclear rim and co-localizes with lamin A/C. As compared to HeLa, there is a reduced co-localization of AKTIP with lamin A/C in both MCF7 and A549. Additionally, MCF7 display lower amounts of AKTIP at the rim. The analyses in non-transformed fibroblasts show that AKTIP mislocalizes in HGPS cells but not in EDMD2. The integrated analysis of lamin expression, nuclear morphology, and AKTIP topology shows that positioning of AKTIP is influenced not only by lamin expression, but also by nuclear morphology. This conclusion is validated by progerin-expressing HeLa cells in which nuclei are morphologically altered and AKTIP is mislocalized. Conclusions Our data show that the combined alteration of lamin and nuclear morphology influences the localization of the tumor-associated factor AKTIP. The results also point to the fact that lamin alterations per se are not predictive of AKTIP mislocalization, in both non-transformed and tumor cells. In more general terms, this study supports the thesis that a combined analytical approach should be preferred to predict lamin-associated changes in tumor cells. This paves the way of next translational evaluation to validate the use of this combined analytical approach as risk biomarker. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02480-5.
Collapse
|
23
|
Borah S, Dhanasekaran K, Kumar S. The LEM-ESCRT toolkit: Repair and maintenance of the nucleus. Front Cell Dev Biol 2022; 10:989217. [PMID: 36172278 PMCID: PMC9512039 DOI: 10.3389/fcell.2022.989217] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 08/24/2022] [Indexed: 12/04/2022] Open
Abstract
The eukaryotic genome is enclosed in a nuclear envelope that protects it from potentially damaging cellular activities and physically segregates transcription and translation.Transport across the NE is highly regulated and occurs primarily via the macromolecular nuclear pore complexes.Loss of nuclear compartmentalization due to defects in NPC function and NE integrity are tied to neurological and ageing disorders like Alzheimer’s, viral pathogenesis, immune disorders, and cancer progression.Recent work implicates inner-nuclear membrane proteins of the conserved LEM domain family and the ESCRT machinery in NE reformation during cell division and NE repair upon rupture in migrating cancer cells, and generating seals over defective NPCs. In this review, we discuss the recent in-roads made into defining the molecular mechanisms and biochemical networks engaged by LEM and many other integral inner nuclear membrane proteins to preserve the nuclear barrier.
Collapse
Affiliation(s)
- Sapan Borah
- National Institute of Immunohaematology, Mumbai, Maharashtra, India
- *Correspondence: Sapan Borah, ; Karthigeyan Dhanasekaran, ; Santosh Kumar,
| | - Karthigeyan Dhanasekaran
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad, Haryana, India
- *Correspondence: Sapan Borah, ; Karthigeyan Dhanasekaran, ; Santosh Kumar,
| | - Santosh Kumar
- National Centre for Cell Science, Pune, Maharashtra, India
- *Correspondence: Sapan Borah, ; Karthigeyan Dhanasekaran, ; Santosh Kumar,
| |
Collapse
|
24
|
Bell ES, Shah P, Zuela-Sopilniak N, Kim D, Varlet AA, Morival JL, McGregor AL, Isermann P, Davidson PM, Elacqua JJ, Lakins JN, Vahdat L, Weaver VM, Smolka MB, Span PN, Lammerding J. Low lamin A levels enhance confined cell migration and metastatic capacity in breast cancer. Oncogene 2022; 41:4211-4230. [PMID: 35896617 PMCID: PMC9925375 DOI: 10.1038/s41388-022-02420-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 07/12/2022] [Accepted: 07/14/2022] [Indexed: 02/07/2023]
Abstract
Aberrations in nuclear size and shape are commonly used to identify cancerous tissue. However, it remains unclear whether the disturbed nuclear structure directly contributes to the cancer pathology or is merely a consequence of other events occurring during tumorigenesis. Here, we show that highly invasive and proliferative breast cancer cells frequently exhibit Akt-driven lower expression of the nuclear envelope proteins lamin A/C, leading to increased nuclear deformability that permits enhanced cell migration through confined environments that mimic interstitial spaces encountered during metastasis. Importantly, increasing lamin A/C expression in highly invasive breast cancer cells reflected gene expression changes characteristic of human breast tumors with higher LMNA expression, and specifically affected pathways related to cell-ECM interactions, cell metabolism, and PI3K/Akt signaling. Further supporting an important role of lamins in breast cancer metastasis, analysis of lamin levels in human breast tumors revealed a significant association between lower lamin A levels, Akt signaling, and decreased disease-free survival. These findings suggest that downregulation of lamin A/C in breast cancer cells may influence both cellular physical properties and biochemical signaling to promote metastatic progression.
Collapse
Affiliation(s)
- Emily S. Bell
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY,Current address: Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA
| | - Pragya Shah
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | | | - Dongsung Kim
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - Alice-Anais Varlet
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - Julien L.P. Morival
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - Alexandra L. McGregor
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY,Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY
| | - Philipp Isermann
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | | | - Joshua J. Elacqua
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - Jonathan N. Lakins
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA
| | - Linda Vahdat
- Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Valerie M. Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA,Helen Diller Cancer Center, Department of Bioengineering and Therapeutic Sciences, and Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA
| | - Marcus B. Smolka
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - Paul N. Span
- Department of Radiation Oncology, Radiotherapy & OncoImmunology laboratory, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands
| | - Jan Lammerding
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA. .,Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
25
|
Urciuoli E, Peruzzi B. The Paradox of Nuclear Lamins in Pathologies: Apparently Controversial Roles Explained by Tissue-Specific Mechanobiology. Cells 2022; 11:cells11142194. [PMID: 35883635 PMCID: PMC9318957 DOI: 10.3390/cells11142194] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/06/2022] [Accepted: 07/08/2022] [Indexed: 11/24/2022] Open
Abstract
The nuclear lamina is a complex meshwork of intermediate filaments (lamins) that is located beneath the inner nuclear membrane and the surrounding nucleoplasm. The lamins exert both structural and functional roles in the nucleus and, by interacting with several nuclear proteins, are involved in a wide range of nuclear and cellular activities. Due their pivotal roles in basic cellular processes, lamin gene mutations, or modulations in lamin expression, are often associated with pathological conditions, ranging from rare genetic diseases, such as laminopathies, to cancer. Although a substantial amount of literature describes the effects that are mediated by the deregulation of nuclear lamins, some apparently controversial results have been reported, which may appear to conflict with each other. In this context, we herein provide our explanation of such “controversy”, which, in our opinion, derives from the tissue-specific expression of nuclear lamins and their close correlation with mechanotransduction processes, which could be very different, or even opposite, depending on the specific mechanical conditions that should not be compared (a tissue vs. another tissue, in vivo studies vs. cell cultures on glass/plastic supports, etc.). Moreover, we have stressed the relevance of considering and reproducing the “mechano-environment” in in vitro experimentation. Indeed, when primary cells that are collected from patients or donors are maintained in a culture, the mechanical signals deriving from canonical experimental procedures of cell culturing could alter the lamin expression, thereby profoundly modifying the assessed cell type, in some cases even too much, compared to the cell of origin.
Collapse
|
26
|
Akkour K, Alanazi IO, Alfadda AA, Alhalal H, Masood A, Musambil M, Rahman AMA, Alwehaibi MA, Arafah M, Bassi A, Benabdelkamel H. Tissue-Based Proteomic Profiling in Patients with Hyperplasia and Endometrial Cancer. Cells 2022; 11:cells11132119. [PMID: 35805203 PMCID: PMC9265283 DOI: 10.3390/cells11132119] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 12/24/2022] Open
Abstract
Uterine cancers are among the most prevalent gynecological malignancies, and endometrial cancer (EC) is the most common in this group. This study used tissue-based proteomic profiling analysis in patients with endometrial cancer and hyperplasia, and control patients. Conventional 2D gel electrophoresis, followed by a mass spectrometry approach with bioinformatics, including a network pathway analysis pipeline, was used to identify differentially expressed proteins and associated metabolic pathways between the study groups. Thirty-six patients (twelve with endometrial cancer, twelve with hyperplasia, and twelve controls) were enrolled in this study. The mean age of the participants was 46–75 years. Eighty-seven proteins were significantly differentially expressed between the study groups, of which fifty-three were significantly differentially regulated (twenty-eight upregulated and twenty-five downregulated) in the tissue samples of EC patients compared to the control (Ctrl). Furthermore, 26 proteins were significantly dysregulated (8 upregulated and 18 downregulated) in tissue samples of hyperplasia (HY) patients compared to Ctrl. Thirty-two proteins (nineteen upregulated and thirteen downregulated) including desmin, peptidyl prolyl cis-trans isomerase A, and zinc finger protein 844 were downregulated in the EC group compared to the HY group. Additionally, fructose bisphosphate aldolase A, alpha enolase, and keratin type 1 cytoskeletal 10 were upregulated in the EC group compared to those in the HY group. The proteins identified in this study were known to regulate cellular processes (36%), followed by biological regulation (16%). Ingenuity pathway analysis found that proteins that are differentially expressed between EC and HY are linked to AKT, ACTA2, and other signaling pathways. The panels of protein markers identified in this study could be used as potential biomarkers for distinguishing between EC and HY and early diagnosis and progression of EC from hyperplasia and normal patients.
Collapse
Affiliation(s)
- Khalid Akkour
- Obstetrics and Gynecology Department, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (K.A.); (H.A.); (A.B.)
| | - Ibrahim O. Alanazi
- The National Center for Biotechnology (NCB), Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia;
| | - Assim A. Alfadda
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (A.A.A.); (A.M.); (M.M.); (M.A.A.)
- Department of Medicine, College of Medicine and King Saud Medical City, King Saud University, Riyadh 11461, Saudi Arabia
| | - Hani Alhalal
- Obstetrics and Gynecology Department, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (K.A.); (H.A.); (A.B.)
| | - Afshan Masood
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (A.A.A.); (A.M.); (M.M.); (M.A.A.)
| | - Mohthash Musambil
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (A.A.A.); (A.M.); (M.M.); (M.A.A.)
| | - Anas M. Abdel Rahman
- Metabolomics Section, Department of Clinical Genomics, Center for Genome Medicine, King Faisal Specialist Hospital and Research Centre (KFSHRC), Riyadh 11211, Saudi Arabia;
| | - Moudi A. Alwehaibi
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (A.A.A.); (A.M.); (M.M.); (M.A.A.)
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11461, Saudi Arabia
| | - Maria Arafah
- Department of Pathology, College of Medicine, King Saud University, King Saud University Medical City, Riyadh 11461, Saudi Arabia;
| | - Ali Bassi
- Obstetrics and Gynecology Department, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (K.A.); (H.A.); (A.B.)
| | - Hicham Benabdelkamel
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (A.A.A.); (A.M.); (M.M.); (M.A.A.)
- Correspondence:
| |
Collapse
|
27
|
Nanou A, Lorenzo-Moldero I, Gazouleas KD, Cortese B, Moroni L. 3D Culture Modeling of Metastatic Breast Cancer Cells in Additive Manufactured Scaffolds. ACS APPLIED MATERIALS & INTERFACES 2022; 14:28389-28402. [PMID: 35687666 PMCID: PMC9227707 DOI: 10.1021/acsami.2c07492] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Cancer biology research is increasingly moving toward innovative in vitro 3D culture models, as conventional and current 2D cell cultures fail to resemble in vivo cancer biology. In the current study, porous 3D scaffolds, designed with two different porosities along with 2D tissue culture polystyrene (TCP) plates were used with a model breast cancer human cell line. The 3D engineered system was evaluated for the optimal seeding method (dynamic versus static), adhesion, and proliferation rate of MDA-MB-231 breast cancer cells. The expression profiles of proliferation-, stemness-, and dormancy-associated cancer markers, namely, ki67, lamin A/C, SOX2, Oct3/4, stanniocalcin 1 (STC1), and stanniocalcin 2 (STC2), were evaluated in the 3D cultured cells and compared to the respective profiles of the cells cultured in the conventional 2D TCP. Our data suggested that static seeding was the optimal seeding method with porosity-dependent efficiency. Moreover, cells cultured in 3D scaffolds displayed a more dormant phenotype in comparison to 2D, which was manifested by the lower proliferation rate, reduced ki67 expression, increased lamin A/C expression, and overexpression of STCs. The possible relationship between the cell affinity to different extracellular matrix (ECM) proteins and the RANK expression levels was also addressed after deriving collagen type I (COL-I) and fibronectin (FN) MDA-MB-231 filial cell lines with enhanced capacity to attach to the respective ECM proteins. The new derivatives exhibited a more mesenchymal like phenotype and higher RANK levels in relation to the parental cells, suggesting a relationship between ECM cell affinity and RANK expression. Therefore, the present 3D cell culture model shows that cancer cells on printed scaffolds can work as better representatives in cancer biology and drug screening related studies.
Collapse
Affiliation(s)
- Afroditi Nanou
- Tissue
Regeneration Department, MIRA Institute for Biomedical Technology, University of Twente, Drienerlolaan 5, 7522 ND Enschede, The Netherlands
- Medical
Cell BioPhysics Department, Faculty of Science and Technology, University of Twente, Dienstweg 1, 7522 ND Enschede, The Netherlands
| | - Ivan Lorenzo-Moldero
- Tissue
Regeneration Department, MIRA Institute for Biomedical Technology, University of Twente, Drienerlolaan 5, 7522 ND Enschede, The Netherlands
- Complex
Tissue Regeneration Department, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Kyriakos D. Gazouleas
- Tissue
Regeneration Department, MIRA Institute for Biomedical Technology, University of Twente, Drienerlolaan 5, 7522 ND Enschede, The Netherlands
| | - Barbara Cortese
- National
Research Council-Nanotechnology Institute (CNR Nanotec), 00185 Rome, Italy
- Email for B.C.:
| | - Lorenzo Moroni
- Tissue
Regeneration Department, MIRA Institute for Biomedical Technology, University of Twente, Drienerlolaan 5, 7522 ND Enschede, The Netherlands
- Complex
Tissue Regeneration Department, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
- National
Research Council-Nanotechnology Institute (CNR Nanotec), 00185 Rome, Italy
- Email for L.M.:
| |
Collapse
|
28
|
Bellanger A, Madsen-Østerbye J, Galigniana NM, Collas P. Restructuring of Lamina-Associated Domains in Senescence and Cancer. Cells 2022; 11:1846. [PMID: 35681541 PMCID: PMC9180887 DOI: 10.3390/cells11111846] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/02/2022] [Accepted: 06/04/2022] [Indexed: 01/01/2023] Open
Abstract
Induction of cellular senescence or cancer is associated with a reshaping of the nuclear envelope and a broad reorganization of heterochromatin. At the periphery of mammalian nuclei, heterochromatin is stabilized at the nuclear lamina via lamina-associated domains (LADs). Alterations in the composition of the nuclear lamina during senescence lead to a loss of peripheral heterochromatin, repositioning of LADs, and changes in epigenetic states of LADs. Cancer initiation and progression are also accompanied by a massive reprogramming of the epigenome, particularly in domains coinciding with LADs. Here, we review recent knowledge on alterations in chromatin organization and in the epigenome that affect LADs and related genomic domains in senescence and cancer.
Collapse
Affiliation(s)
- Aurélie Bellanger
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway; (A.B.); (J.M.-Ø.); (N.M.G.)
| | - Julia Madsen-Østerbye
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway; (A.B.); (J.M.-Ø.); (N.M.G.)
| | - Natalia M. Galigniana
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway; (A.B.); (J.M.-Ø.); (N.M.G.)
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, 0372 Oslo, Norway
| | - Philippe Collas
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway; (A.B.); (J.M.-Ø.); (N.M.G.)
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, 0372 Oslo, Norway
| |
Collapse
|
29
|
Basu A, Patel NG, Nicholson ED, Weiss RJ. Spatiotemporal diversity and regulation of glycosaminoglycans in cell homeostasis and human disease. Am J Physiol Cell Physiol 2022; 322:C849-C864. [PMID: 35294848 PMCID: PMC9037703 DOI: 10.1152/ajpcell.00085.2022] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Glycosaminoglycans (GAGs) are long, linear polysaccharides that are ubiquitously expressed on the cell surface and in the extracellular matrix of all animal cells. These complex carbohydrates play important roles in many cellular processes and have been implicated in many disease states, including cancer, inflammation, and genetic disorders. GAGs are among the most complex molecules in biology with enormous information content and extensive structural and functional heterogeneity. GAG biosynthesis is a nontemplate-driven process facilitated by a large group of biosynthetic enzymes that have been extensively characterized over the past few decades. Interestingly, the expression of the enzymes and the consequent structure and function of the polysaccharide chains can vary temporally and spatially during development and under certain pathophysiological conditions, suggesting their assembly is tightly regulated in cells. Due to their many key roles in cell homeostasis and disease, there is much interest in targeting the assembly and function of GAGs as a therapeutic approach. Recent advances in genomics and GAG analytical techniques have pushed the field and generated new perspectives on the regulation of mammalian glycosylation. This review highlights the spatiotemporal diversity of GAGs and the mechanisms guiding their assembly and function in human biology and disease.
Collapse
Affiliation(s)
- Amrita Basu
- 1Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia
| | - Neil G. Patel
- 1Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia,2Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia
| | - Elijah D. Nicholson
- 2Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia
| | - Ryan J. Weiss
- 1Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia,2Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia
| |
Collapse
|
30
|
Rose M, Burgess JT, O’Byrne K, Richard DJ, Bolderson E. The role of inner nuclear membrane proteins in tumourigenesis and as potential targets for cancer therapy. Cancer Metastasis Rev 2022; 41:953-963. [PMID: 36205821 PMCID: PMC9758098 DOI: 10.1007/s10555-022-10065-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 09/18/2022] [Indexed: 01/25/2023]
Abstract
Despite significant advances in our understanding of tumourigenesis and cancer therapeutics, cancer continues to account for 30% of worldwide deaths. Therefore, there remains an unmet need for the development of cancer therapies to improve patient quality of life and survival outcomes. The inner nuclear membrane has an essential role in cell division, cell signalling, transcription, cell cycle progression, chromosome tethering, cell migration and mitosis. Furthermore, expression of several inner nuclear membrane proteins has been shown to be frequently altered in tumour cells, resulting in the dysregulation of cellular pathways to promote tumourigenesis. However, to date, minimal research has been conducted to investigate how targeting these dysregulated and variably expressed proteins may provide a novel avenue for cancer therapies. In this review, we present an overview of the involvement of the inner nuclear membrane proteins within the hallmarks of cancer and how they may be exploited as potent anti-cancer therapeutics.
Collapse
Affiliation(s)
- Maddison Rose
- grid.1024.70000000089150953Cancer & Ageing Research Program (CARP), Centre for Genomics and Personalised Health (CGPH), School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD Australia
| | - Joshua T. Burgess
- grid.1024.70000000089150953Cancer & Ageing Research Program (CARP), Centre for Genomics and Personalised Health (CGPH), School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD Australia
| | - Kenneth O’Byrne
- grid.1024.70000000089150953Cancer & Ageing Research Program (CARP), Centre for Genomics and Personalised Health (CGPH), School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD Australia ,grid.412744.00000 0004 0380 2017Princess Alexandra Hospital, Ipswich Road, Woolloongabba, Brisbane, QLD 4102 Australia
| | - Derek J. Richard
- grid.1024.70000000089150953Cancer & Ageing Research Program (CARP), Centre for Genomics and Personalised Health (CGPH), School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD Australia
| | - Emma Bolderson
- grid.1024.70000000089150953Cancer & Ageing Research Program (CARP), Centre for Genomics and Personalised Health (CGPH), School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD Australia
| |
Collapse
|
31
|
Stenvall CGA, Tayyab M, Grönroos TJ, Ilomäki MA, Viiri K, Ridge KM, Polari L, Toivola DM. Targeted deletion of keratin 8 in intestinal epithelial cells disrupts tissue integrity and predisposes to tumorigenesis in the colon. Cell Mol Life Sci 2021; 79:10. [PMID: 34951664 PMCID: PMC8709826 DOI: 10.1007/s00018-021-04081-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/24/2021] [Accepted: 12/04/2021] [Indexed: 01/08/2023]
Abstract
Keratin 8 (K8) is the main intestinal epithelial intermediate filament protein with proposed roles for colonic epithelial cell integrity. Here, we used mice lacking K8 in intestinal epithelial cells (floxed K8 and Villin-Cre1000 and Villin-CreERt2) to investigate the cell-specific roles of intestinal epithelial K8 for colonocyte function and pathologies. Intestinal epithelial K8 deletion decreased K8 partner proteins, K18-K20, 75-95%, and the remaining keratin filaments were located at the colonocyte apical regions with type II K7, which decreased 30%. 2-Deoxy-2-[18F]-fluoroglucose positron emission tomography in vivo imaging identified a metabolic phenotype in the lower gut of the conditional K8 knockouts. These mice developed intestinal barrier leakiness, mild diarrhea, and epithelial damage, especially in the proximal colon. Mice exhibited shifted differentiation from enterocytes to goblet cells, displayed longer crypts and an increased number of Ki67 + transit-amplifying cells in the colon. Significant proproliferative and regenerative signaling occurred in the IL-22, STAT3, and pRb pathways, with minor effects on inflammatory parameters, which, however, increased in aging mice. Importantly, colonocyte K8 deletion induced a dramatically increased sensitivity to azoxymethane-induced tumorigenesis. In conclusion, intestinal epithelial K8 plays a significant role in colonocyte epithelial integrity maintenance, proliferation regulation and tumor suppression.
Collapse
Affiliation(s)
- Carl-Gustaf A Stenvall
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, BioCity, Tykistökatu 6A, N20520, Turku, Finland
| | - Mina Tayyab
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, BioCity, Tykistökatu 6A, N20520, Turku, Finland
| | - Tove J Grönroos
- Turku PET Centre, University of Turku, Turku, Finland
- Medicity Research Laboratories, University of Turku, Turku, Finland
| | - Maria A Ilomäki
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, BioCity, Tykistökatu 6A, N20520, Turku, Finland
| | - Keijo Viiri
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere University Hospital, Tampere, Finland
| | - Karen M Ridge
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Lauri Polari
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, BioCity, Tykistökatu 6A, N20520, Turku, Finland
| | - Diana M Toivola
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, BioCity, Tykistökatu 6A, N20520, Turku, Finland.
- Turku Center for Disease Modeling, University of Turku, Turku, Finland.
| |
Collapse
|
32
|
Stefanello ST, Luchtefeld I, Liashkovich I, Pethö Z, Azzam I, Bulk E, Rosso G, Döhlinger L, Hesse B, Oeckinghaus A, Shahin V. Impact of the Nuclear Envelope on Malignant Transformation, Motility, and Survival of Lung Cancer Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2102757. [PMID: 34658143 PMCID: PMC8596107 DOI: 10.1002/advs.202102757] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/11/2021] [Indexed: 05/08/2023]
Abstract
Nuclear pore complexes (NPCs) selectively mediate all nucleocytoplasmic transport and engage in fundamental cell-physiological processes. It is hypothesized that NPCs are critical for malignant transformation and survival of lung cancer cells, and test the hypothesis in lowly and highly metastatic non-small human lung cancer cells (NSCLCs). It is shown that malignant transformation is paralleled by an increased NPCs density, and a balanced pathological weakening of the physiological stringency of the NPC barrier. Pharmacological interference using barrier-breaking compounds collapses the stringency. Concomitantly, it induces drastic overall structural changes of NSCLCs, terminating their migration. Moreover, the degree of malignancy is found to be paralleled by substantially decreased lamin A/C levels. The latter provides crucial structural and mechanical stability to the nucleus, and interacts with NPCs, cytoskeleton, and nucleoskeleton for cell maintenance, survival, and motility. The recent study reveals the physiological importance of the NPC barrier stringency for mechanical and structural resilience of normal cell nuclei. Hence, reduced lamin A/C levels in conjunction with controlled pathological weakening of the NPC barrier stringency may facilitate deformability of NSCLCs during the metastasis steps. Modulation of the NPC barrier presents a potential strategy for suppressing the malignant phenotype or enhancing the effectiveness of currently existing chemotherapeutics.
Collapse
Affiliation(s)
- Sílvio Terra Stefanello
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, Münster, 48149, Germany
| | - Isabelle Luchtefeld
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, Münster, 48149, Germany
| | - Ivan Liashkovich
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, Münster, 48149, Germany
| | - Zoltan Pethö
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, Münster, 48149, Germany
| | - Ihab Azzam
- Institute of Immunology, University of Münster, Röntgen-Str. 21, Münster, 48149, Germany
| | - Etmar Bulk
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, Münster, 48149, Germany
| | - Gonzalo Rosso
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, Münster, 48149, Germany
| | - Lilly Döhlinger
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, Münster, 48149, Germany
| | - Bettina Hesse
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, Münster, 48149, Germany
| | - Andrea Oeckinghaus
- Institute of Molecular Tumor Biology, University of Münster, Robert-Koch-Str. 43, Münster, 48149, Germany
| | - Victor Shahin
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, Münster, 48149, Germany
| |
Collapse
|
33
|
The Role of Emerin in Cancer Progression and Metastasis. Int J Mol Sci 2021; 22:ijms222011289. [PMID: 34681951 PMCID: PMC8537873 DOI: 10.3390/ijms222011289] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 12/27/2022] Open
Abstract
It is commonly recognized in the field that cancer cells exhibit changes in the size and shape of their nuclei. These features often serve as important biomarkers in the diagnosis and prognosis of cancer patients. Nuclear size can significantly impact cell migration due to its incredibly large size. Nuclear structural changes are predicted to regulate cancer cell migration. Nuclear abnormalities are common across a vast spectrum of cancer types, regardless of tissue source, mutational spectrum, and signaling dependencies. The pervasiveness of nuclear alterations suggests that changes in nuclear structure may be crucially linked to the transformation process. The factors driving these nuclear abnormalities, and the functional consequences, are not completely understood. Nuclear envelope proteins play an important role in regulating nuclear size and structure in cancer. Altered expression of nuclear lamina proteins, including emerin, is found in many cancers and this expression is correlated with better clinical outcomes. A model is emerging whereby emerin, as well as other nuclear lamina proteins, binding to the nucleoskeleton regulates the nuclear structure to impact metastasis. In this model, emerin and lamins play a central role in metastatic transformation, since decreased emerin expression during transformation causes the nuclear structural defects required for increased cell migration, intravasation, and extravasation. Herein, we discuss the cellular functions of nuclear lamina proteins, with a particular focus on emerin, and how these functions impact cancer progression and metastasis.
Collapse
|
34
|
Urciuoli E, D'Oria V, Petrini S, Peruzzi B. Lamin A/C Mechanosensor Drives Tumor Cell Aggressiveness and Adhesion on Substrates With Tissue-Specific Elasticity. Front Cell Dev Biol 2021; 9:712377. [PMID: 34595168 PMCID: PMC8476891 DOI: 10.3389/fcell.2021.712377] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 08/24/2021] [Indexed: 12/03/2022] Open
Abstract
Besides its structural properties in the nucleoskeleton, Lamin A/C is a mechanosensor protein involved in perceiving the elasticity of the extracellular matrix. In this study we provide evidence about Lamin A/C-mediated regulation of osteosarcoma cell adhesion and spreading on substrates with tissue-specific elasticities. Our working hypothesis is based on the observation that low-aggressive and bone-resident SaOS-2 osteosarcoma cells express high level of Lamin A/C in comparison to highly metastatic, preferentially to the lung, osteosarcoma 143B cells, thereby suggesting a role for Lamin A/C in tumor cell tropism. Specifically, LMNA gene over-expression in 143B cells induced a reduction in tumor cell aggressiveness in comparison to parental cells, with decreased proliferation rate and reduced migration capability. Furthermore, LMNA reintegration into 143B cells changed the adhesion properties of tumor cells, from a preferential tropism toward the 1.5 kPa PDMS substrate (resembling normal lung parenchyma) to the 28 kPa (resembling pre-mineralized bone osteoid matrix). Our study suggests that Lamin A/C expression could be involved in the organ tropism of tumor cells, thereby providing a rationale for further studies focused on the definition of cancer mechanism of metastatization.
Collapse
Affiliation(s)
- Enrica Urciuoli
- Multifactorial Disease and Complex Phenotype Research Area, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Valentina D'Oria
- Confocal Microscopy Core Facility, Research Center, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Stefania Petrini
- Confocal Microscopy Core Facility, Research Center, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Barbara Peruzzi
- Multifactorial Disease and Complex Phenotype Research Area, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| |
Collapse
|
35
|
Ovsiannikova NL, Lavrushkina SV, Ivanova AV, Mazina LM, Zhironkina OA, Kireev II. Lamin A as a Determinant of Mechanical Properties of the Cell Nucleus in Health and Disease. BIOCHEMISTRY. BIOKHIMIIA 2021; 86:1288-1300. [PMID: 34903160 DOI: 10.1134/s0006297921100102] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/31/2021] [Accepted: 08/02/2021] [Indexed: 06/14/2023]
Abstract
One of the main factors associated with worse prognosis in oncology is metastasis, which is based on the ability of tumor cells to migrate from the primary source and to form secondary tumors. The search for new strategies to control migration of metastatic cells is one of the urgent issues in biomedicine. One of the strategies to stop spread of cancer cells could be regulation of the nuclear elasticity. Nucleus, as the biggest and stiffest cellular compartment, determines mechanical properties of the cell as a whole, and, hence, could prevent cell migration through the three-dimensional extracellular matrix. Nuclear rigidity is maintained by the nuclear lamina, two-dimensional network of intermediate filaments in the inner nuclear membrane (INM). Here we present the most significant factors defining nucleus rigidity, discuss the role of nuclear envelope composition in the cell migration, as well consider possible approaches to control lamina composition in order to change plasticity of the cell nucleus and ability of the tumor cells to metastasize.
Collapse
Affiliation(s)
- Natalia L Ovsiannikova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992, Russia.
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Svetlana V Lavrushkina
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Anastasia V Ivanova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Ludmila M Mazina
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Oxana A Zhironkina
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992, Russia
| | - Igor I Kireev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
- Kulakov National Medical Research Center for Obstetrics, Gynecology, and Perinatology, Moscow, 117198, Russia
| |
Collapse
|
36
|
Gatti G, Vilardo L, Musa C, Di Pietro C, Bonaventura F, Scavizzi F, Torcinaro A, Bucci B, Saporito R, Arisi I, De Santa F, Raspa M, Guglielmi L, D’Agnano I. Role of Lamin A/C as Candidate Biomarker of Aggressiveness and Tumorigenicity in Glioblastoma Multiforme. Biomedicines 2021; 9:biomedicines9101343. [PMID: 34680461 PMCID: PMC8533312 DOI: 10.3390/biomedicines9101343] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/21/2021] [Accepted: 09/24/2021] [Indexed: 12/11/2022] Open
Abstract
Nuclear lamina components have long been regarded as scaffolding proteins, forming a dense fibrillar structure necessary for the maintenance of the nucleus shape in all the animal kingdom. More recently, mutations, aberrant localisation and deregulation of these proteins have been linked to several diseases, including cancer. Using publicly available data we found that the increased expression levels of the nuclear protein Lamin A/C correlate with a reduced overall survival in The Cancer Genome Atlas Research Network (TCGA) patients affected by glioblastoma multiforme (GBM). We show that the expression of the LMNA gene is linked to the enrichment of cancer-related pathways, particularly pathways related to cell adhesion and cell migration. Mimicking the modulation of LMNA in a GBM preclinical cancer model, we confirmed both in vitro and in vivo that the increased expression of LMNA is associated with an increased aggressiveness and tumorigenicity. In addition, delving into the possible mechanism behind LMNA-induced GBM aggressiveness and tumorigenicity, we found that the mTORC2 component, Rictor, plays a central role in mediating these effects.
Collapse
Affiliation(s)
- Giuliana Gatti
- Department of Biotechnology and Translational Medicine, University of Milan, 20129 Milan, Italy;
| | - Laura Vilardo
- Institute for Biomedical Technologies (ITB), CNR, 20054 Segrate, Italy; (L.V.); (C.M.)
| | - Carla Musa
- Institute for Biomedical Technologies (ITB), CNR, 20054 Segrate, Italy; (L.V.); (C.M.)
| | - Chiara Di Pietro
- Institute of Biochemistry and Cell Biology (IBBC), CNR, 00015 Monterotondo, Italy; (C.D.P.); (F.B.); (F.S.); (A.T.); (F.D.S.); (M.R.)
| | - Fabrizio Bonaventura
- Institute of Biochemistry and Cell Biology (IBBC), CNR, 00015 Monterotondo, Italy; (C.D.P.); (F.B.); (F.S.); (A.T.); (F.D.S.); (M.R.)
| | - Ferdinando Scavizzi
- Institute of Biochemistry and Cell Biology (IBBC), CNR, 00015 Monterotondo, Italy; (C.D.P.); (F.B.); (F.S.); (A.T.); (F.D.S.); (M.R.)
| | - Alessio Torcinaro
- Institute of Biochemistry and Cell Biology (IBBC), CNR, 00015 Monterotondo, Italy; (C.D.P.); (F.B.); (F.S.); (A.T.); (F.D.S.); (M.R.)
| | - Barbara Bucci
- UOC Clinical Pathology, San Pietro Hospital FBF, 00189 Rome, Italy; (B.B.); (R.S.)
| | - Raffaele Saporito
- UOC Clinical Pathology, San Pietro Hospital FBF, 00189 Rome, Italy; (B.B.); (R.S.)
| | - Ivan Arisi
- Bioinformatics Facility, European Brain Research Institute (EBRI) “Rita Levi Montalcini”, 00161 Rome, Italy;
| | - Francesca De Santa
- Institute of Biochemistry and Cell Biology (IBBC), CNR, 00015 Monterotondo, Italy; (C.D.P.); (F.B.); (F.S.); (A.T.); (F.D.S.); (M.R.)
| | - Marcello Raspa
- Institute of Biochemistry and Cell Biology (IBBC), CNR, 00015 Monterotondo, Italy; (C.D.P.); (F.B.); (F.S.); (A.T.); (F.D.S.); (M.R.)
| | - Loredana Guglielmi
- Institute for Biomedical Technologies (ITB), CNR, 20054 Segrate, Italy; (L.V.); (C.M.)
- Correspondence: (L.G.); (I.D.)
| | - Igea D’Agnano
- Institute for Biomedical Technologies (ITB), CNR, 20054 Segrate, Italy; (L.V.); (C.M.)
- Correspondence: (L.G.); (I.D.)
| |
Collapse
|
37
|
Setti Boubaker N, Gurtner A, Trabelsi N, Manni I, Ayed H, Saadi A, Zaghbib S, Naimi Z, Sahraoui G, Zouari S, Meddeb K, Mrad K, Chebil M, Piaggio G, Ouerhani S. The diagnostic applicability of A-type Lamin in non-muscle invasive bladder cancer. Ann Diagn Pathol 2021; 54:151808. [PMID: 34438192 DOI: 10.1016/j.anndiagpath.2021.151808] [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: 06/09/2021] [Revised: 07/04/2021] [Accepted: 08/10/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE Lamin A is a major component of the nuclear lamina maintaining nuclear integrity, regulation of gene expression, cell proliferation, and apoptosis. Its deregulation in cancer has been recently reported to be associated with its prognosis. However, its clinical significance in non-muscle invasive bladder cancer (NMIBC) remains to be defined. MATERIAL/METHODS Immunohistochemical staining and RT-qPCR were performed to screen the expression patterns of Lamin A/C protein and Lamin A mRNA respectively in 58 high and low grade NMIBC specimens. RESULTS Lamin A/C protein was expressed only in the nucleus and less exhibited in NMIBC tissues compared to non-tumoral ones. On the other side, Lamin A mRNA was up-regulated in NMIBC compared to controls. Nevertheless, both expression patterns (protein and mRNA) were not correlated to clinical prognosis factors and were not able to predict the overall survival of patients with high-grade NMIBC. CONCLUSIONS The deregulation of A-type Lamin is not associated with the prognosis of NMIBC, but it could serve as a diagnostic biomarker distinguishing NMIBC patients from healthy subjects suggesting its involvement as an initiator event of tumorigenesis in our cohort.
Collapse
Affiliation(s)
- Nouha Setti Boubaker
- Laboratory of Proteins Engineering and Bioactive Molecules (LIP-MB), INSAT, University of Tunis Carthage, Tunis, Tunisia; UOSD SAFU, Department of Research, Diagnosis and Innovative Technologies, IRCCS-Regina Elena National Cancer Institute, Rome, Italy.
| | - Aymone Gurtner
- UOSD SAFU, Department of Research, Diagnosis and Innovative Technologies, IRCCS-Regina Elena National Cancer Institute, Rome, Italy; Institute of Translational Pharmacology, National Research Council, Rome, Italy.
| | - Nesrine Trabelsi
- Laboratory of Proteins Engineering and Bioactive Molecules (LIP-MB), INSAT, University of Tunis Carthage, Tunis, Tunisia.
| | - Isabella Manni
- UOSD SAFU, Department of Research, Diagnosis and Innovative Technologies, IRCCS-Regina Elena National Cancer Institute, Rome, Italy.
| | - Haroun Ayed
- Urology Department, Charles Nicolle Hospital, Faculty of Medicine of Tunis, University of Tunis-El Manar, Tunis, Tunisia.
| | - Ahmed Saadi
- Urology Department, Charles Nicolle Hospital, Faculty of Medicine of Tunis, University of Tunis-El Manar, Tunis, Tunisia.
| | - Selim Zaghbib
- Urology Department, Charles Nicolle Hospital, Faculty of Medicine of Tunis, University of Tunis-El Manar, Tunis, Tunisia.
| | - Zeineb Naimi
- Medical Oncology Department, Salah Azaiez Institute, Faculty of Medicine of Tunis, University of Tunis-El Manar, Tunis, Tunisia.
| | - Ghada Sahraoui
- Pathology Department, Salah Azaiez Institute, Faculty of Medicine of Tunis, University of Tunis-El Manar, Tunis, Tunisia.
| | - Skander Zouari
- Urology Department, Charles Nicolle Hospital, Faculty of Medicine of Tunis, University of Tunis-El Manar, Tunis, Tunisia.
| | - Khedija Meddeb
- Medical Oncology Department, Salah Azaiez Institute, Faculty of Medicine of Tunis, University of Tunis-El Manar, Tunis, Tunisia.
| | - Karima Mrad
- Pathology Department, Salah Azaiez Institute, Faculty of Medicine of Tunis, University of Tunis-El Manar, Tunis, Tunisia.
| | - Mohamed Chebil
- Urology Department, Charles Nicolle Hospital, Faculty of Medicine of Tunis, University of Tunis-El Manar, Tunis, Tunisia.
| | - Giulia Piaggio
- UOSD SAFU, Department of Research, Diagnosis and Innovative Technologies, IRCCS-Regina Elena National Cancer Institute, Rome, Italy.
| | - Slah Ouerhani
- Laboratory of Proteins Engineering and Bioactive Molecules (LIP-MB), INSAT, University of Tunis Carthage, Tunis, Tunisia.
| |
Collapse
|
38
|
Nishikawa T, Kuwano Y, Nakata M, Rokutan K, Nishida K. Multiple G-quadruplexes in the LMNA promoter regulate LMNA variant 6 transcription and promote colon cancer cell growth. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2021; 1864:194746. [PMID: 34419630 DOI: 10.1016/j.bbagrm.2021.194746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/22/2021] [Accepted: 08/11/2021] [Indexed: 10/20/2022]
Abstract
Lamin A/C proteins, major components of the nuclear lamina, are encoded by the LMNA gene. These proteins have multiple cellular functions, including DNA transcription and replication, chromatin organization, regulation of the cell cycle, and apoptosis. Mutations in LMNA are associated with a variety of diseases called laminopathies. LMNA has implications in cancer; however, its mechanisms of dysregulation in cancer cells are not yet fully understood. In this study, among the LMNA transcript variants, we focused on a transcriptional variant 6 (termed LMNA-V6), which contains unique 3 exons upstream of exon 1 of LMNA. The promoter region of LMNA-V6 formed multiple G-quadruplexes and increased its transcriptional activity. Moreover, LMNA-V6 negatively regulated other LMNA mRNA variants, lamin A and lamin C, via direct interaction with their promoter. Knockdown of LMNA-V6 decreased the proliferation of colon cancer cells, whereas overexpression of the unique 3 exons of LMNA-V6 increased cell growth. Furthermore, microarray gene expression profiling showed that alteration of LMNA-V6 levels influenced the expression of p53 in colon cancer cells. Taken together, the results suggest that LMNA-V6 may be a novel functional RNA whose expression is regulated through multiple G-quadruplexes in colon cancer cells.
Collapse
Affiliation(s)
- Tatsuya Nishikawa
- Department of Pathophysiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan; Department of Onco-cardiology, Osaka International Cancer Institute, Osaka, Japan
| | - Yuki Kuwano
- Department of Pathophysiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan.
| | - Mayu Nakata
- Student Lab, Tokushima University Faculty of Medicine, Tokushima, Japan
| | - Kazuhito Rokutan
- Department of Pathophysiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Kensei Nishida
- Department of Pathophysiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| |
Collapse
|
39
|
Plastin 3 in health and disease: a matter of balance. Cell Mol Life Sci 2021; 78:5275-5301. [PMID: 34023917 PMCID: PMC8257523 DOI: 10.1007/s00018-021-03843-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/06/2021] [Accepted: 04/20/2021] [Indexed: 02/06/2023]
Abstract
For a long time, PLS3 (plastin 3, also known as T-plastin or fimbrin) has been considered a rather inconspicuous protein, involved in F-actin-binding and -bundling. However, in recent years, a plethora of discoveries have turned PLS3 into a highly interesting protein involved in many cellular processes, signaling pathways, and diseases. PLS3 is localized on the X-chromosome, but shows sex-specific, inter-individual and tissue-specific expression variability pointing towards skewed X-inactivation. PLS3 is expressed in all solid tissues but usually not in hematopoietic cells. When escaping X-inactivation, PLS3 triggers a plethora of different types of cancers. Elevated PLS3 levels are considered a prognostic biomarker for cancer and refractory response to therapies. When it is knocked out or mutated in humans and mice, it causes osteoporosis with bone fractures; it is the only protein involved in actin dynamics responsible for osteoporosis. Instead, when PLS3 is upregulated, it acts as a highly protective SMN-independent modifier in spinal muscular atrophy (SMA). Here, it seems to counteract reduced F-actin levels by restoring impaired endocytosis and disturbed calcium homeostasis caused by reduced SMN levels. In contrast, an upregulation of PLS3 on wild-type level might cause osteoarthritis. This emphasizes that the amount of PLS3 in our cells must be precisely balanced; both too much and too little can be detrimental. Actin-dynamics, regulated by PLS3 among others, are crucial in a lot of cellular processes including endocytosis, cell migration, axonal growth, neurotransmission, translation, and others. Also, PLS3 levels influence the infection with different bacteria, mycosis, and other pathogens.
Collapse
|
40
|
Dubik N, Mai S. Lamin A/C: Function in Normal and Tumor Cells. Cancers (Basel) 2020; 12:cancers12123688. [PMID: 33316938 PMCID: PMC7764147 DOI: 10.3390/cancers12123688] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/07/2020] [Accepted: 12/08/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary The aim of this review is to summarize lamin A/C’s currently known functions in both normal and diseased cells. Lamin A/C is a nuclear protein with many functions in cells, such as maintaining a cell’s structural stability, cell motility, mechanosensing, chromosome organization, gene regulation, cell differentiation, DNA damage repair, and telomere protection. Mutations of the lamin A/C gene, incorrect processing of the protein, and lamin A/C deregulation can lead to various diseases and cancer. This review touches on diseases caused by mutation and incorrect processing of lamin A/C, called laminopathies. The effect of lamin A/C deregulation in cancer is also reviewed, and lamin A/C’s potential in helping to diagnose prostate cancers more accurately is discussed. Abstract This review is focused on lamin A/C, a nuclear protein with multiple functions in normal and diseased cells. Its functions, as known to date, are summarized. This summary includes its role in maintaining a cell’s structural stability, cell motility, mechanosensing, chromosome organization, gene regulation, cell differentiation, DNA damage repair, and telomere protection. As lamin A/C has a variety of critical roles within the cell, mutations of the lamin A/C gene and incorrect processing of the protein results in a wide variety of diseases, ranging from striated muscle disorders to accelerated aging diseases. These diseases, collectively termed laminopathies, are also touched upon. Finally, we review the existing evidence of lamin A/C’s deregulation in cancer. Lamin A/C deregulation leads to various traits, including genomic instability and increased tolerance to mechanical insult, which can lead to more aggressive cancer and poorer prognosis. As lamin A/C’s expression in specific cancers varies widely, currently known lamin A/C expression in various cancers is reviewed. Additionally, Lamin A/C’s potential as a biomarker in various cancers and as an aid in more accurately diagnosing intermediate Gleason score prostate cancers is also discussed.
Collapse
|
41
|
Emerging roles of lamins and DNA damage repair mechanisms in ovarian cancer. Biochem Soc Trans 2020; 48:2317-2333. [DOI: 10.1042/bst20200713] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/14/2020] [Accepted: 09/21/2020] [Indexed: 02/06/2023]
Abstract
Lamins are type V intermediate filament proteins which are ubiquitously present in all metazoan cells providing a platform for binding of chromatin and related proteins, thereby serving a wide range of nuclear functions including DNA damage repair. Altered expression of lamins in different subtypes of cancer is evident from researches worldwide. But whether cancer is a consequence of this change or this change is a consequence of cancer is a matter of future investigation. However changes in the expression levels of lamins is reported to have direct or indirect association with cancer progression or have regulatory roles in common neoplastic symptoms like higher nuclear deformability, increased genomic instability and reduced susceptibility to DNA damaging agents. It has already been proved that loss of A type lamin positively regulates cathepsin L, eventually leading to degradation of several DNA damage repair proteins, hence impairing DNA damage repair pathways and increasing genomic instability. It is established in ovarian cancer, that the extent of alteration in nuclear morphology can determine the degree of genetic changes and thus can be utilized to detect low to high form of serous carcinoma. In this review, we have focused on ovarian cancer which is largely caused by genomic alterations in the DNA damage response pathways utilizing proteins like RAD51, BRCA1, 53BP1 which are regulated by lamins. We have elucidated the current understanding of lamin expression in ovarian cancer and its implications in the regulation of DNA damage response pathways that ultimately result in telomere deformation and genomic instability.
Collapse
|
42
|
Liu H, Li D, Zhou L, Kan S, He G, Zhou K, Wang L, Chen M, Shu W. LMNA functions as an oncogene in hepatocellular carcinoma by regulating the proliferation and migration ability. J Cell Mol Med 2020; 24:12008-12019. [PMID: 32896989 PMCID: PMC7578910 DOI: 10.1111/jcmm.15829] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/21/2020] [Accepted: 08/17/2020] [Indexed: 12/13/2022] Open
Abstract
The role of the LMNA gene in the development and progression of hepatocellular carcinoma (HCC) and the associated molecular mechanism is not yet clear. Therefore, the purpose of this study was to evaluate the relationship between LMNA and HCC. LMNA gene expression in normal tissues and corresponding tumours was evaluated and the Kaplan-Meier survival analysis was performed. Next, the LMNA gene was knocked out in the 293T and HepG2 cell lines using the CRISPR/Cas9 technique. Subsequently, the proliferation, migration and colony formation rate of the two LMNA knockout cell lines were analysed. Finally, the molecular mechanism affecting the tumorigenesis due to the loss of the LMNA gene was evaluated. The results showed that the LMNA gene was abnormally expressed in many tumours, and the survival rate of the HCC patients with a high expression of the LMNA gene was significantly reduced compared with the rate in patients with a low LMNA expression. The knockout of the LMNA gene in the HCC cell line HepG2 resulted in a decreased tumorigenicity, up-regulation of the P16 expression and down-regulation of the CDK1 expression. These findings suggested that LMNA might function as an oncogene in HCC and provided a potential new target for the diagnosis and treatment of HCC.
Collapse
Affiliation(s)
- Heng Liu
- College of Biotechnology, Guilin Medical University, Guilin, China.,College of Stomatology, Guangxi Medical University, Nanning, China
| | - Dongming Li
- Department of Cell Biology and Genetics, Guangxi Medical University, Nanning, China
| | - Ling Zhou
- Department of Cell Biology and Genetics, Guangxi Medical University, Nanning, China
| | - Shuang Kan
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin, China
| | - Guozhang He
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin, China
| | - Kun Zhou
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Liping Wang
- College of Biotechnology, Guilin Medical University, Guilin, China
| | - Ming Chen
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin, China
| | - Wei Shu
- College of Biotechnology, Guilin Medical University, Guilin, China
| |
Collapse
|
43
|
Structural and Mechanical Aberrations of the Nuclear Lamina in Disease. Cells 2020; 9:cells9081884. [PMID: 32796718 PMCID: PMC7464082 DOI: 10.3390/cells9081884] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/02/2020] [Accepted: 08/10/2020] [Indexed: 12/13/2022] Open
Abstract
The nuclear lamins are the major components of the nuclear lamina in the nuclear envelope. Lamins are involved in numerous functions, including a role in providing structural support to the cell and the mechanosensing of the cell. Mutations in the genes encoding for lamins lead to the rare diseases termed laminopathies. However, not only laminopathies show alterations in the nuclear lamina. Deregulation of lamin expression is reported in multiple cancers and several viral infections lead to a disrupted nuclear lamina. The structural and mechanical effects of alterations in the nuclear lamina can partly explain the phenotypes seen in disease, such as muscular weakness in certain laminopathies and transmigration of cancer cells. However, a lot of answers to questions about the relation between changes in the nuclear lamina and disease development remain elusive. Here, we review the current understandings of the contribution of the nuclear lamina in the structural support and mechanosensing of healthy and diseased cells.
Collapse
|
44
|
Altinoz MA, Ucal Y, Yilmaz MC, Kiris İ, Ozisik O, Sezerman U, Ozpinar A, Elmaci İ. Progesterone at high doses reduces the growth of U87 and A172 glioblastoma cells: Proteomic changes regarding metabolism and immunity. Cancer Med 2020; 9:5767-5780. [PMID: 32590878 PMCID: PMC7433824 DOI: 10.1002/cam4.3223] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 04/24/2020] [Accepted: 05/15/2020] [Indexed: 12/16/2022] Open
Abstract
While pregnancy may accelerate glioblastoma multiforme (GBM) growth, parity and progesterone (P4) containing treatments (ie, hormone replacement therapy) reduce the risk of GBM development. In parallel, low and high doses of P4 exert stimulating and inhibitory actions on GBM growth, respectively. The mechanisms behind the high‐dose P4‐suppression of GBM growth is unknown. In the present study, we assessed the changes in growth and proteomic profiles when high‐dose P4 (100 and 300 µM) was administered in human U87 and A172 GBM cell lines. The xCELLigence system was used to examine cell growth when different concentrations of P4 (20, 50, 100, and 300 µM) was administered. The protein profiles were determined by two‐dimensional gel electrophoresis in both cell lines when 100 and 300 µM P4 were administered. Finally, the pathways enriched by the differentially expressed proteins were assessed using bioinformatic tools. Increasing doses of P4 blocked the growth of both GBM cells. We identified 26 and 51 differentially expressed proteins (fc > 2) in A172 and U87 cell lines treated with P4, respectively. Only the pro‐tumorigenic mitochondrial ornithine aminotransferase and anti‐apoptotic mitochondrial 60 kDa heat shock protein were downregulated in A172 cell line and U87 cell line when treated with P4, respectively. Detoxification of reactive oxygen species, cellular response to stress, glucose metabolism, and immunity‐related proteins were altered in P4‐treated GBM cell lines. The paradox on the effect of low and high doses of P4 on GBM growth is gaining attention. The mechanism related to the high dose of P4 on GBM growth can be explained by the alterations in detoxification mechanisms, stress, and immune response and glucose metabolism. P4 suppresses GBM growth and as it is nontoxic in comparison to classical chemotherapeutics, it can be used as a new strategy in GBM treatment in the future.
Collapse
Affiliation(s)
- Meric A Altinoz
- Department of Medical Biochemistry, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Yasemin Ucal
- Department of Medical Biochemistry, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Muazzez C Yilmaz
- Department of Medical Biochemistry, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - İrem Kiris
- Department of Medical Biochemistry, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Ozan Ozisik
- Medical Genetics, Aix Marseille University, Inserm, MMG, Marseille, France
| | - Ugur Sezerman
- Department of Biostatistics and Medical Informatics, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Aysel Ozpinar
- Department of Medical Biochemistry, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - İlhan Elmaci
- Department of Neurosurgery, Acibadem Maslak Hospital and School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| |
Collapse
|
45
|
Oncogene PRR14 promotes breast cancer through activation of PI3K signal pathway and inhibition of CHEK2 pathway. Cell Death Dis 2020; 11:464. [PMID: 32541902 PMCID: PMC7296039 DOI: 10.1038/s41419-020-2640-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 12/28/2022]
Abstract
Nuclear envelope component PRR14 has been detected to be upregulated in varieties of cancers, especially in breast cancer. But its role in breast carcinogenesis is poorly understood. In this study, we show PRR14 contributes to breast carcinogenesis mainly through overexpression, which derives from elevated transcription and gene amplification. Increased PRR14 expression promotes breast cancer cell proliferation and tumor formation. Biochemical analysis reveals, in addition to previously reported activation of PI3-kinase/Akt/mTOR pathway, PRR14 overexpression regulates cell cycle in breast cancer by inhibiting CHEK2’s activation, followed with the deregulation of DNA damage pathway. In correspondence, CHEK2 and PRR14 show opposite impact on breast cancer patients receiving chemotherapy. Collectively, our study is the first to document the oncogenetic role of PRR14 in breast cancer, which protects cells from apoptosis and stimulates proliferation by activating the PI3-kinase/Akt/mTOR pathway and inhibiting the CHEK2 pathway. Both of these pathways are of great influence in breast cancer and PRR14 appears to be their novel interacting node, which renders patients more resistance to chemotherapy and provides a potential therapeutic target in breast cancer.
Collapse
|
46
|
Blank M. Targeted Regulation of Nuclear Lamins by Ubiquitin and Ubiquitin-Like Modifiers. Cells 2020; 9:cells9061340. [PMID: 32471220 PMCID: PMC7348791 DOI: 10.3390/cells9061340] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/25/2020] [Accepted: 05/26/2020] [Indexed: 02/06/2023] Open
Abstract
Nuclear lamins (NLs) are essential components of the animal cell nucleus involved in the regulation of a plethora of molecular and cellular processes. These include the nuclear envelope assembly and stability, mechanotransduction and chromatin organization, transcription, DNA replication, damage repair, and genomic integrity maintenance. Mutations in NLs can lead to the development of a wide range of distinct disease phenotypes, laminopathies, consisting of cardiac, neuromuscular, metabolic and premature aging syndromes. In addition, alterations in the expression of nuclear lamins were associated with different types of neoplastic diseases. Despite the importance and critical roles that NLs play in the diverse cellular activities, we only recently started to uncover the complexity of regulatory mechanisms governing their expression, localization and functions. This integrative review summarizes and discusses the recent findings on the emerging roles of ubiquitin and ubiquitin-like modifiers (ULMs) in the regulation of NLs, highlighting the intriguing molecular associations and cross-talks occurring between NLs and these regulatory molecules under physiological conditions and in the disease states.
Collapse
Affiliation(s)
- Michael Blank
- Laboratory of Molecular and Cellular Cancer Biology, Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| |
Collapse
|
47
|
Cancer associated fibroblast: Mediators of tumorigenesis. Matrix Biol 2020; 91-92:19-34. [PMID: 32450219 DOI: 10.1016/j.matbio.2020.05.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 05/08/2020] [Accepted: 05/12/2020] [Indexed: 02/07/2023]
Abstract
It is well accepted that the tumor microenvironment plays a pivotal role in cancer onset, development, and progression. The majority of clinical interventions are designed to target either cancer or stroma cells. These emphases have been directed by one of two prevailing theories in the field, the Somatic Mutation Theory and the Tissue Organization Field Theory, which represent two seemingly opposing concepts. This review proposes that the two theories are mutually inclusive and should be concurrently considered for cancer treatments. Specifically, this review discusses the dynamic and reciprocal processes between stromal cells and extracellular matrices, using pancreatic cancer as an example, to demonstrate the inclusivity of the theories. Furthermore, this review highlights the functions of cancer associated fibroblasts, which represent the major stromal cell type, as important mediators of the known cancer hallmarks that the two theories attempt to explain.
Collapse
|
48
|
Zhang F, Li X, Ni Y, Shan G, Gao Y. Preliminary study of the urinary proteome in Li and Han ethnic individuals from Hainan. SCIENCE CHINA. LIFE SCIENCES 2020; 63:125-137. [PMID: 31102176 DOI: 10.1007/s11427-018-9485-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 01/23/2019] [Indexed: 01/05/2023]
Abstract
Biomarkers indicate changes associated with disease. Blood is relatively stable due to the homeostatic mechanisms of the body; however, urine accumulates metabolites from changes in the body, making it a better source for early biomarker discovery. The Li ethnic group is a unique minority ethnic group that has only lived on Hainan Island for approximately 5,000 years. Studies have shown that various specific genetic variations are different between the Li and Han ethnic groups. However, whether the urinary proteome between these two ethnic groups is significantly different remains unknown. In this study, differential urinary proteins were identified in the Li and Han ethnic groups using liquid chromatography tandem mass spectrometry (LC-MS/MS). In total, 1,555 urinary proteins were identified. Twenty-five of the urinary proteins were statistically significantly different, 16 of which have been previously reported to be biomarkers of many diseases, and that these significantly different proteins were caused by ethnic differences rather than random differences. Ethnic group differences may be an influencing factor in urine proteome studies and should be considered when human urine samples are used for biomarker discovery.
Collapse
Affiliation(s)
- Fanshuang Zhang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.,Department of Pathophysiology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Xundou Li
- Department of Pathophysiology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Yanying Ni
- Department of Pathophysiology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Guangliang Shan
- Department of Epidemiology and Statistics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.
| | - Youhe Gao
- Department of Biochemistry and Molecular Biology, Beijing Normal University, Gene Engineering Drug and Biotechnology Beijing Key Laboratory, Beijing, 100875, China.
| |
Collapse
|
49
|
Klingler-Hoffmann M, Mittal P, Hoffmann P. The Emerging Role of Cytoskeletal Proteins as Reliable Biomarkers. Proteomics 2019; 19:e1800483. [PMID: 31525818 DOI: 10.1002/pmic.201800483] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 07/12/2019] [Indexed: 12/26/2022]
Abstract
Cytoskeletal proteins are essential building blocks of cells. More than 100 cytoskeletal and cytoskeleton-associated proteins are known and for some, their function and regulation are understood in great detail. Apart from cell shape and support, they facilitate many processes such as intracellular signaling and transport, and cancer related processes such as proliferation, migration, and invasion. During the last decade, comparative proteomic studies have identified cytoskeletal proteins as in vitro markers for tumor progression and metastasis. Here, these results are summarized and a number of unrelated studies are highlighted, identifying the same cytoskeletal proteins as potential biomarkers. These findings might indicate that the abundance of these potential markers of tumor progression is associated with the biological outcome and are independent of the cancer origin. This correlates well with recently published results from the Cancer Genome Atlas, indicating that cancers show remarkable similarities in their analyzed molecular information, independent of their organ of origin. It is postulated that the quantification of cytoskeletal proteins in healthy tissues, tumors, in adjacent tissues, and in stroma, is a great source of molecular information, which might not only be used to classify tumors, but more importantly to predict patients' outcome or even best treatment choices.
Collapse
Affiliation(s)
- Manuela Klingler-Hoffmann
- Future Industries Institute, University of South Australia, Mawson Lakes Campus, Mawson Lakes, 5095, Australia
| | - Parul Mittal
- Adelaide Proteomics Centre, School of Biological Sciences, University of Adelaide, Adelaide, 5005, Australia
| | - Peter Hoffmann
- Future Industries Institute, University of South Australia, Mawson Lakes Campus, Mawson Lakes, 5095, Australia
| |
Collapse
|
50
|
Qiu C, Huang F, Zhang Q, Chen W, Zhang H. miR-205-3p promotes proliferation and reduces apoptosis of breast cancer MCF-7 cells and is associated with poor prognosis of breast cancer patients. J Clin Lab Anal 2019; 33:e22966. [PMID: 31578772 PMCID: PMC6805278 DOI: 10.1002/jcla.22966] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 06/03/2019] [Accepted: 06/05/2019] [Indexed: 12/24/2022] Open
Abstract
Background To study the expression of microribonucleic acid (miR)‐205 in breast cancer and its effects on the proliferation and apoptosis of breast cancer cells. Methods Breast cancer cell line MCF‐7 cells with stable expression of miR‐205‐3p were constructed. Cell proliferation, invasion, and apoptosis were detected via MTT assay, transwell assay, and flow cytometry, respectively. The expressions of Ezrin, LaminA/C, cleaved caspase‐3, Bcl‐2, and Bax were detected via Western blotting. The expressions of miR‐205‐3p in breast cancer tissues and para‐carcinoma tissues were detected via quantitative PCR (qPCR). Results In transfection group, cell proliferation and invasion capacities were increased significantly (P < 0.01), but apoptotic cells were significantly reduced (P < 0.01). In addition, the expressions of Ezrin, LaminA/C, and cleaved caspase‐3 in the transfection group were significantly decreased (P < 0.01), but the Bcl‐2/Bax ratio was significantly increased (P < 0.01). The miR‐205‐3p expression in tumor tissues of breast cancer patients was significantly higher than that in para‐carcinoma tissue, but Ezrin, LaminA/C, and cleaved caspase‐3 expressions in tumor tissues were remarkably declined (P < 0.01), while the Bcl‐2/Bax ratio was remarkably increased (P < 0.01). Moreover, the 5‐year survival of patients with high expression of miR‐205‐3p was significantly shorter than patients with normal or low expression (P < 0.01). Conclusion Highly expressed miR‐205‐3p can promote the proliferation and invasion and reduce the apoptosis of breast cancer cells, and the high expression of miR‐205‐3p can significantly reduce the survival time of patients.
Collapse
Affiliation(s)
- Changhong Qiu
- Department of General Surgery, The First People's Hospital of ZhaoQing, ZhaoQing, China
| | - Fei Huang
- Department of General Surgery, The Seventh Affiliated Hospital of Sun Yat-Sen University (Shen Zhen), Shen Zhen, China
| | - Qing Zhang
- Department of General Surgery, The First People's Hospital of ZhaoQing, ZhaoQing, China
| | - Wei Chen
- Department of General Surgery, The First People's Hospital of ZhaoQing, ZhaoQing, China
| | - Huiting Zhang
- Department of General Surgery, The First People's Hospital of ZhaoQing, ZhaoQing, China
| |
Collapse
|