1
|
Samanta P, Ghosh R, Pakhira S, Mondal M, Biswas S, Sarkar R, Bhowmik A, Saha P, Hajra S. Ribosome biogenesis and ribosomal proteins in cancer stem cells: a new therapeutic prospect. Mol Biol Rep 2024; 51:1016. [PMID: 39325314 DOI: 10.1007/s11033-024-09963-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 09/20/2024] [Indexed: 09/27/2024]
Abstract
Ribosome has been considered as the fundamental macromolecular machine involved in protein synthesis in both prokaryotic and eukaryotic cells. This protein synthesis machinery consists of several rRNAs and numerous proteins. All of these factors are synthesized, translocated and assembled in a tightly regulated process known as ribosome biogenesis. Any impairment in this process causes development of several diseases like cancer. According to growing evidences, cancer cells display alteration of several ribosomal proteins. Besides, most of them are considered as key molecules involved in ribosome biogenesis, suggesting a correlation between those proteins and formation of ribosomes. Albeit, defective ribosome biogenesis in several cancers has gained prime importance, regulation of this process in cancer stem cells (CSCs) are still unrecognized. In this article, we aim to summarize the alteration of ribosome biogenesis and ribosomal proteins in CSCs. Moreover, we want to highlight the relation of ribosome biogenesis with hypoxia and drug resistance in CSCs based on the existing evidences. Lastly, this review wants to pay attention about the promising anti-cancer drugs which have potential to inhibit ribosome biogenesis in cancer cells as well as CSCs.
Collapse
Affiliation(s)
- Priya Samanta
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, S.P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Rituparna Ghosh
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, S.P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Shampa Pakhira
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, S.P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Mrinmoyee Mondal
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, S.P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Souradeep Biswas
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, S.P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Rupali Sarkar
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, S.P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Arijit Bhowmik
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, S.P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Prosenjit Saha
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, S.P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Subhadip Hajra
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, S.P. Mukherjee Road, Kolkata, West Bengal, 700026, India.
| |
Collapse
|
2
|
Lynch SM, Heeran AB, Burke C, Lynam-Lennon N, Eustace AJ, Dean K, Robson T, Rahman A, Marcone S. Precision Oncology, Artificial Intelligence, and Novel Therapeutic Advancements in the Diagnosis, Prevention, and Treatment of Cancer: Highlights from the 59th Irish Association for Cancer Research (IACR) Annual Conference. Cancers (Basel) 2024; 16:1989. [PMID: 38893110 PMCID: PMC11171401 DOI: 10.3390/cancers16111989] [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: 04/19/2024] [Revised: 05/21/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Advancements in oncology, especially with the era of precision oncology, is resulting in a paradigm shift in cancer care. Indeed, innovative technologies, such as artificial intelligence, are paving the way towards enhanced diagnosis, prevention, and personalised treatments as well as novel drug discoveries. Despite excellent progress, the emergence of resistant cancers has curtailed both the pace and extent to which we can advance. By combining both their understanding of the fundamental biological mechanisms and technological advancements such as artificial intelligence and data science, cancer researchers are now beginning to address this. Together, this will revolutionise cancer care, by enhancing molecular interventions that may aid cancer prevention, inform clinical decision making, and accelerate the development of novel therapeutic drugs. Here, we will discuss the advances and approaches in both artificial intelligence and precision oncology, presented at the 59th Irish Association for Cancer Research annual conference.
Collapse
Affiliation(s)
- Seodhna M. Lynch
- Personalised Medicine Centre, School of Medicine, Ulster University, C-TRIC Building, Altnagelvin Area Hospital, Glenshane Road, Londonderry BT47 6SB, UK;
| | - Aisling B. Heeran
- Department of Surgery, Trinity Translational Medicine Institute, Trinity St. James’s Cancer Institute, Trinity College Dublin, D02 PN40 Dublin, Ireland; (A.B.H.); (N.L.-L.); (S.M.)
| | - Caoimbhe Burke
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Belfield, D04 C1P1 Dublin, Ireland;
| | - Niamh Lynam-Lennon
- Department of Surgery, Trinity Translational Medicine Institute, Trinity St. James’s Cancer Institute, Trinity College Dublin, D02 PN40 Dublin, Ireland; (A.B.H.); (N.L.-L.); (S.M.)
| | - Alex J. Eustace
- Life Sciences Institute, Dublin City University, D09 NR58 Dublin, Ireland;
| | - Kellie Dean
- School of Biochemistry and Cell Biology, Western Gateway Building, University College Cork, T12 XF62 Cork, Ireland;
| | - Tracy Robson
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland
| | - Arman Rahman
- UCD School of Medicine, UCD Conway Institute, University College Dublin, Belfield, D04 C1P1 Dublin, Ireland;
| | - Simone Marcone
- Department of Surgery, Trinity Translational Medicine Institute, Trinity St. James’s Cancer Institute, Trinity College Dublin, D02 PN40 Dublin, Ireland; (A.B.H.); (N.L.-L.); (S.M.)
| |
Collapse
|
3
|
Cheng IH, Pi WC, Hsu CH, Guo Y, Lai JL, Wang GG, Chung BC, Roeder RG, Chen WY. TAF2, within the TFIID complex, regulates the expression of a subset of protein-coding genes. Cell Death Discov 2024; 10:244. [PMID: 38773077 PMCID: PMC11109217 DOI: 10.1038/s41420-024-02017-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/03/2024] [Accepted: 05/08/2024] [Indexed: 05/23/2024] Open
Abstract
TFIID, one of the general transcription factor (GTF), regulates transcriptional initiation of protein-coding genes through direct binding to promoter elements and subsequent recruitment of other GTFs and RNA polymerase II. Although generally required for most protein-coding genes, accumulated studies have also demonstrated promoter-specific functions for several TFIID subunits in gene activation. Here, we report that TBP-associated factor 2 (TAF2) specifically regulates TFIID binding to a small subset of protein-coding genes and is essential for cell growth of multiple cancer lines. Co-immunoprecipitation assays revealed that TAF2 may be sub-stoichiometrically associated with the TFIID complex, thus indicating a minor fraction of TAF2-containing TFIID in cells. Consistently, integrated genome-wide profiles show that TAF2 binds to and regulates only a small subset of protein-coding genes. Furthermore, through the use of an inducible TAF2 degradation system, our results reveal a reduction of TBP/TFIID binding to several ribosomal genes upon selective ablation of TAF2. In addition, depletion of TAF2, as well as the TAF2-regulated ribosomal protein genes RPL30 and RPL39, decreases ribosome assembly and global protein translation. Collectively, this study suggests that TAF2 within the TFIID complex is of functional importance for TBP/TFIID binding to and expression of a small subset of protein-coding genes, thus establishing a previously unappreciated promoter-selective function for TAF2.
Collapse
Affiliation(s)
- I-Hsin Cheng
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wen-Chieh Pi
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chung-Hao Hsu
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yiran Guo
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Cancer Institute, Durham, NC, 27710, USA
| | - Jun-Lin Lai
- Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Gang G Wang
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Cancer Institute, Durham, NC, 27710, USA
| | - Bon-Chu Chung
- Insitute of Molecular Biology, Academia Sinica, Taipei, Taiwan
- Graduate Institute of Biomedical Sciences, Neuroscience and Brain Disease Center, China Medical University, Taichung, Taiwan
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan
| | - Robert G Roeder
- Laboratory of Biochemistry and Molecular Biology, The Rockefeller University, New York, NY, 10065, USA
| | - Wei-Yi Chen
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan.
- Cancer and Immunology Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
4
|
Peng C, Ye H, Yi Z. GAPDH: unveiling its impact as a key hypoxia-related player in head and neck squamous cell carcinoma tumor progression, prognosis, and therapeutic potential. Am J Cancer Res 2023; 13:5846-5867. [PMID: 38187058 PMCID: PMC10767337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 11/01/2023] [Indexed: 01/09/2024] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC), characterized by hypoxia patterns, ranks as the sixth most prevalent malignant tumor worldwide. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) plays a role in oncogenesis under hypoxic conditions in various cancers. However, its precise function in HNSCC, especially under varied hypoxic conditions, including at high altitudes, remains unclear. Elevated GAPDH mRNA and protein levels in HNSCC relative to normal tissues have been demonstrated through data from The Cancer Genome Atlas (TCGA), GSE29330, and the Human Protein Atlas (P<0.05). This elevation was further confirmed through in vitro experiments utilizing two HNSCC cell lines and a normal oral mucosal epithelial cell line. Additionally, data from TCGA and GSE41613 reveal a correlation between elevated GAPDH expression and diminished overall and progression-free survival in patients (P<0.05). Subsequent analysis identifies GAPDH as an independent risk factor for HNSCC (P<0.05). Using the ESTIMATE and single-sample gene set enrichment analysis (ssGSEA) algorithms, high GAPDH expression was found to be associated with reduced immune scores and diminished anti-tumor cell infiltration, such as CD8+ T cells, in TCGA and GSE41613 datasets (P<0.05). Analysis of single-cell RNA sequencing data from GSE139324 suggests that elevated GAPDH expression hinders communication between plasmacytoid dendritic cells and mast cells (P<0.05). Furthermore, in the TCGA and GSE41613 datasets, GAPDH's biological function is closely tied to hypoxia through Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Variation Analysis (GSVA) analyses. Moreover, its expression is linked to one cuproptosis-related gene, five N6-methyladenosine-related genes, six immune checkpoint genes, and pivotal pathways such as MYC and E2F (P<0.05). GAPDH showed excellent predictive value in estimating the efficacy of immunotherapy and 11 anti-tumor drugs (e.g., cisplatin) (P<0.05), using TIDE and pRRophetic algorithms on the TCGA and GSE41613 datasets. Under 1% O2 in vitro, HNSCC cells show elevated GAPDH expression, leading to decreased apoptosis and increased migration, clonogenicity, invasiveness, and resistance to cisplatin (P<0.05). At 5% O2, these effects persisted, albeit less pronouncedly. Inhibiting GAPDH reversed these effects under all oxygen concentrations (P<0.05). Overall, our findings reveal GAPDH as a key hypoxia-related player influencing tumor progression, prognosis, and therapeutic potential in HNSCC.
Collapse
Affiliation(s)
- Cong Peng
- Department of Otolaryngology, Guizhou Provincial People's Hospital Guiyang, Guizhou, China
| | - Huiping Ye
- Department of Otolaryngology, Guizhou Provincial People's Hospital Guiyang, Guizhou, China
| | - Zhuguang Yi
- Department of Otolaryngology, Guizhou Provincial People's Hospital Guiyang, Guizhou, China
| |
Collapse
|
5
|
Jeanmard N, Bissanum R, Sriplung H, Charoenlappanit S, Roytrakul S, Navakanitworakul R. Proteomic profiling of urinary extracellular vesicles differentiates breast cancer patients from healthy women. PLoS One 2023; 18:e0291574. [PMID: 37922300 PMCID: PMC10624262 DOI: 10.1371/journal.pone.0291574] [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: 03/01/2023] [Accepted: 08/31/2023] [Indexed: 11/05/2023] Open
Abstract
Urinary extracellular vesicles (uEVs) reflect the biological conditions of the producing cells. The protein profiling of uEVs allow us to better understand cancer progression in several cancers such as bladder cancer, prostate cancer and kidney cancer but has not been reported in breast cancer. We have, herein, aimed at quantifying the concentration and at generating the proteomic profile of uEVs in patients with breast cancer (BC) as compared to that of healthy controls (CT). Urine samples were collected from 29 CT and 47 patients with BC. uEVs were isolated by using differential ultracentrifugation, and were then characterized by Western blotting and transmission electron microscopy. Moreover, a nanoparticle tracking analysis was used in order to measure the concentration and the size distribution of urine particles and uEVs. The proteomic profiling of the uEVs was facilitated through LC-MS/MS. The uEV concentration was not significantly different between the assessed groups. The undertaken proteomic analysis revealed 15,473 and 11,278 proteins in the BC patients' group and the CT group, respectively. Furthermore, a heat map analysis revealed a differential protein expression, while a principal component analysis highlighted two clusters. The volcano plot indicated 259 differentially expressed proteins (DEPs; 155 up- and 104 down-regulated proteins) in patients with BC compared with CT. The up-regulated proteins from BC-derived uEVs were enriched in pathways related to cancer progression (i.e., cell proliferation, cell survival, cell cycle, cell migration, carbohydrate metabolism, and angiogenesis). Moreover, we verified the expression of the upregulated DEPs using UALCAN for web-based validation. Remarkably, the results indicated that 6 of 155 up-regulated proteins (POSTN, ATAD2, BCAS4, GSK3β, HK1, and Ki-67) were overexpressed in BC compared with normal samples. Since these six proteins often act as markers of cell proliferation and progression, they may be potential biomarkers for BC screening and diagnosis. However, this requires validation in larger cohorts.
Collapse
Affiliation(s)
- Nilobon Jeanmard
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Rassanee Bissanum
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Hutcha Sriplung
- Department of Epidemiology, Faculty of Medicine, Prince of Songkla University, Hat Yai, Thailand
| | - Sawanya Charoenlappanit
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | - Sittiruk Roytrakul
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | - Raphatphorn Navakanitworakul
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| |
Collapse
|
6
|
Neophytou CM, Katsonouri A, Christodoulou MI, Papageorgis P. In Vivo Investigation of the Effect of Dietary Acrylamide and Evaluation of Its Clinical Relevance in Colon Cancer. TOXICS 2023; 11:856. [PMID: 37888706 PMCID: PMC10610724 DOI: 10.3390/toxics11100856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/06/2023] [Accepted: 10/09/2023] [Indexed: 10/28/2023]
Abstract
Dietary exposure to acrylamide (AA) has been linked with carcinogenicity in the gastrointestinal (GI) tract. However, epidemiologic data on AA intake in relation to cancer risk are limited and contradictory, while the potential cancer-inducing molecular pathways following AA exposure remain elusive. In this study, we collected mechanistic information regarding the induction of carcinogenesis by dietary AA in the colon, using an established animal model. Male Balb/c mice received AA orally (0.1 mg/kg/day) daily for 4 weeks. RNA was extracted from colon tissue samples, followed by RNA sequencing. Comparative transcriptomic analysis between AA and mock-treated groups revealed a set of differentially expressed genes (DEGs) that were further processed using different databases through the STRING-DB portal, to reveal deregulated protein-protein interaction networks. We found that genes implicated in RNA metabolism, processing and formation of the ribosomal subunits and protein translation and metabolism are upregulated in AA-exposed colon tissue; these genes were also overexpressed in human colon adenocarcinoma samples and were negatively correlated with patient overall survival (OS), based on publicly available datasets. Further investigation of the potential role of these genes during the early stages of colon carcinogenesis may shed light into the underlying mechanisms induced by dietary AA exposure.
Collapse
Affiliation(s)
- Christiana M Neophytou
- Tumor Microenvironment, Metastasis and Experimental Therapeutics Group, Basic and Translational Cancer Research Center, Department of Life Sciences, European University Cyprus, 2404 Nicosia, Cyprus
- State General Laboratory, Ministry of Health, 2081 Nicosia, Cyprus
| | | | - Maria-Ioanna Christodoulou
- State General Laboratory, Ministry of Health, 2081 Nicosia, Cyprus
- Tumor Immunology and Biomarkers Group, Basic and Translational Cancer Research Center, Department of Life Sciences, European University Cyprus, 2404 Nicosia, Cyprus
| | - Panagiotis Papageorgis
- Tumor Microenvironment, Metastasis and Experimental Therapeutics Group, Basic and Translational Cancer Research Center, Department of Life Sciences, European University Cyprus, 2404 Nicosia, Cyprus
- State General Laboratory, Ministry of Health, 2081 Nicosia, Cyprus
| |
Collapse
|
7
|
Fang D, Hu H, Zhao K, Xu A, Yu C, Zhu Y, Yu N, Yao B, Tang S, Wu X, Mei Y. MLF2 Negatively Regulates P53 and Promotes Colorectal Carcinogenesis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303336. [PMID: 37438558 PMCID: PMC10502657 DOI: 10.1002/advs.202303336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Indexed: 07/14/2023]
Abstract
Inactivation of the p53 pathway is linked to a variety of human cancers. As a critical component of the p53 pathway, ubiquitin-specific protease 7 (USP7) acts as a deubiquitinase for both p53 and its ubiquitin E3 ligase mouse double minute 2 homolog. Here, myeloid leukemia factor 2 (MLF2) is reported as a new negative regulator of p53. MLF2 interacts with both p53 and USP7. Via these interactions, MLF2 inhibits the binding of USP7 to p53 and antagonizes USP7-mediated deubiquitination of p53, thereby leading to p53 destabilization. Functionally, MLF2 plays an oncogenic role in colorectal cancer, at least partially, via the negative regulation of p53. Clinically, MLF2 is elevated in colorectal cancer and its high expression is associated with poor prognosis in patients with colorectal cancer. In wild-type-p53-containing colorectal cancer, MLF2 and p53 expressions are inversely correlated. These findings establish MLF2 as an important suppressor of p53 function. The study also reveals a critical role for the MLF2-p53 axis in promoting colorectal carcinogenesis.
Collapse
Affiliation(s)
- Debao Fang
- Department of Thoracic Surgery, The First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230001China
- The CAS Key Laboratory of Innate Immunity and Chronic DiseaseDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027China
| | - Hao Hu
- The CAS Key Laboratory of Innate Immunity and Chronic DiseaseDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027China
| | - Kailiang Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230001China
- The CAS Key Laboratory of Innate Immunity and Chronic DiseaseDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027China
| | - Aman Xu
- Department of General SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
| | - Changjun Yu
- Department of General SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
| | - Yong Zhu
- Department of General SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
| | - Ning Yu
- The CAS Key Laboratory of Innate Immunity and Chronic DiseaseDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027China
| | - Bo Yao
- The CAS Key Laboratory of Innate Immunity and Chronic DiseaseDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027China
| | - Suyun Tang
- The CAS Key Laboratory of Innate Immunity and Chronic DiseaseDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027China
| | - Xianning Wu
- Department of Thoracic Surgery, The First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230001China
| | - Yide Mei
- Department of Thoracic Surgery, The First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230001China
- The CAS Key Laboratory of Innate Immunity and Chronic DiseaseDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027China
- Center for Advanced Interdisciplinary Science and Biomedicine of IHMDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027China
| |
Collapse
|
8
|
Alsamman K, Alamri AM, Vatte C, Owaidah AY, Alhassan F, Mubarki R, El-Masry OS. Potential Candidate Genes for Therapeutic Targeting in Chronic Myeloid Leukemia: A Pilot Study. Asian Pac J Cancer Prev 2023; 24:3077-3085. [PMID: 37774059 PMCID: PMC10762750 DOI: 10.31557/apjcp.2023.24.9.3077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 09/17/2023] [Indexed: 10/01/2023] Open
Abstract
BACKGROUND Chronic myeloid leukemia (CML) is a prevalent hematological malignancy known for the presence of the Philadelphia chromosome and activation of the BCR-Abl kinase activity. Although tyrosine kinase inhibitors are widely used as the standard treatment, resistance remains a concern among certain patients. This study aimed to investigate the gene expression profile of a group of CML patients in comparison to a control group in order to identify novel candidate genes associated with the disease. METHODS Whole transcriptome sequencing was performed, and gene expression levels were validated using quantitative real-time PCR. Additionally, single nucleotide and insertion/deletion variants were analyzed in the selected candidate genes among 10 CML patients and 4 healthy control subjects. RESULTS Analysis revealed a set of differentially expressed genes, whose up- or downregulation was further confirmed by qRT-PCR. Among the upregulated genes in the patient group were ribosomal protein like (RPL) members, specifically RPL9, RPL34, RPL36A, and RPL39, while downregulation was observed in CCDC170, LDB1, and SBF1 compared to the healthy subjects. Furthermore, gene variant studies identified novel genetic changes in these candidate genes, suggesting potential clinical significance in CML. CONCLUSIONS This study highlights RPL9, RPL34, RPL36A, RPL39, CCDC170, LDB1, and SBF1 as potential targets in CML. Additionally, it underscores the importance of investigating these genes and their variants in larger cohort studies to assess their clinical significance in CML patients.
Collapse
MESH Headings
- Humans
- Pilot Projects
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- LIM-Homeodomain Proteins
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Chronic Disease
- Protein Kinase Inhibitors/pharmacology
- Drug Resistance, Neoplasm
Collapse
Affiliation(s)
- Khaldoon Alsamman
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Imam Abdulrahaman Bin Faisal University, Dammam, Saudi Arabia.
| | - Ali M. Alamri
- Department of Internal Medicine, King Fahd Hospital of the University, Imam Abdulrahaman Bin Faisal University, Alkhobar, Saudi Arabia.
| | - Chittibabu Vatte
- Department of Clinical Biochemistry, College of Medicine, Imam Abdulrahman bin Faisal University, Dammam, Saudi Arabia.
| | - Amani Y. Owaidah
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Imam Abdulrahaman Bin Faisal University, Dammam, Saudi Arabia.
| | - Fatimah Alhassan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Imam Abdulrahaman Bin Faisal University, Dammam, Saudi Arabia.
| | - Roba Mubarki
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Imam Abdulrahaman Bin Faisal University, Dammam, Saudi Arabia.
| | - Omar S. El-Masry
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Imam Abdulrahaman Bin Faisal University, Dammam, Saudi Arabia.
| |
Collapse
|
9
|
Sun J, Peterson EA, Chen X, Wang J. hapln1a + cells guide coronary growth during heart morphogenesis and regeneration. Nat Commun 2023; 14:3505. [PMID: 37311876 PMCID: PMC10264374 DOI: 10.1038/s41467-023-39323-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 06/07/2023] [Indexed: 06/15/2023] Open
Abstract
Although several tissues and chemokines orchestrate coronary formation, the guidance cues for coronary growth remain unclear. Here, we profile the juvenile zebrafish epicardium during coronary vascularization and identify hapln1a+ cells enriched with vascular-regulating genes. hapln1a+ cells not only envelop vessels but also form linear structures ahead of coronary sprouts. Live-imaging demonstrates that coronary growth occurs along these pre-formed structures, with depletion of hapln1a+ cells blocking this growth. hapln1a+ cells also pre-lead coronary sprouts during regeneration and hapln1a+ cell loss inhibits revascularization. Further, we identify serpine1 expression in hapln1a+ cells adjacent to coronary sprouts, and serpine1 inhibition blocks vascularization and revascularization. Moreover, we observe the hapln1a substrate, hyaluronan, forming linear structures along and preceding coronary vessels. Depletion of hapln1a+ cells or serpine1 activity inhibition disrupts hyaluronan structure. Our studies reveal that hapln1a+ cells and serpine1 are required for coronary production by establishing a microenvironment to facilitate guided coronary growth.
Collapse
Affiliation(s)
- Jisheng Sun
- Cardiology Division, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Elizabeth A Peterson
- Cardiology Division, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Xin Chen
- Cardiology Division, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Jinhu Wang
- Cardiology Division, School of Medicine, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
10
|
Reddy TP, Glynn SA, Billiar TR, Wink DA, Chang JC. Targeting Nitric Oxide: Say NO to Metastasis. Clin Cancer Res 2023; 29:1855-1868. [PMID: 36520504 PMCID: PMC10183809 DOI: 10.1158/1078-0432.ccr-22-2791] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/24/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022]
Abstract
Utilizing targeted therapies capable of reducing cancer metastasis, targeting chemoresistant and self-renewing cancer stem cells, and augmenting the efficacy of systemic chemo/radiotherapies is vital to minimize cancer-associated mortality. Targeting nitric oxide synthase (NOS), a protein within the tumor microenvironment, has gained interest as a promising therapeutic strategy to reduce metastatic capacity and augment the efficacy of chemo/radiotherapies in various solid malignancies. Our review highlights the influence of nitric oxide (NO) in tumor progression and cancer metastasis, as well as promising preclinical studies that evaluated NOS inhibitors as anticancer therapies. Lastly, we highlight the prospects and outstanding challenges of using NOS inhibitors in the clinical setting.
Collapse
Affiliation(s)
- Tejaswini P. Reddy
- Texas A&M University Health Science Center, Bryan, Texas
- Houston Methodist Research Institute, Houston, Texas
- Houston Methodist Neal Cancer Center, Houston, Texas
| | - Sharon A. Glynn
- Prostate Cancer Institute, National University of Ireland Galway, Galway, Ireland
| | - Timothy R. Billiar
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - David A. Wink
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institute of Health, Frederick, Maryland
| | - Jenny C. Chang
- Houston Methodist Research Institute, Houston, Texas
- Houston Methodist Neal Cancer Center, Houston, Texas
| |
Collapse
|
11
|
Xu H, Zhao F, Wu D, Zhang Y, Bao X, Shi F, Cai Y, Dou J. Eliciting effective tumor immunity against ovarian cancer by cancer stem cell vaccination. Biomed Pharmacother 2023; 161:114547. [PMID: 36933377 DOI: 10.1016/j.biopha.2023.114547] [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: 02/18/2023] [Revised: 03/12/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023] Open
Abstract
Advanced ovarian cancer (OC) patients have limited benefit from current relevant cytotoxic and targeted therapies following debulking surgery. Therefore, new therapeutic strategies are in urgent need. Immunotherapy has shown great potential in tumor treatment, especially in tumor vaccine development. The study objective was to evaluate the immune effects of cancer stem cells (CSCs) vaccines on OC. The CD44+CD117+CSCs were isolated from human OC HO8910 and SKOV3 cells using the magnetic cell sorting system; the cancer stem-like cells were selected from murine OC ID8 cell by no-serum formed sphere culture. The CSC vaccines were prepared by freezing and thawing these CSCs, which were then injected into mice followed by challenging the different OC cells. The in vivo antitumor efficacy of CSC immunization revealed the vaccines were capable of significantly provoking immune responses to autologous tumor antigens in vaccinated mice as the mice were found to have markedly inhibited tumor growth, prolonged survival, and decreased CSC counts in OC tissues when compared to mice without the CSC vaccination. The in vitro cytotoxicities of immunocytes toward SKOV3, HO8910 and ID8 cells indicated a significant killing efficacy compared with the controls. However, the antitumor efficacy was remarkably reduced whilst the mucin-1 expression in CSC vaccines was down-regulated by small interfering RNA. Overall, findings from this study provided the evidence that has deepened our understanding of CSC vaccine immunogenicity and anti-OC efficacy, particularly for the role of dominant antigen mucin-1. It is possible to turn the CSC vaccine into an immunotherapeutic approach against ovarian cancer.
Collapse
Affiliation(s)
- Hui Xu
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing 210009, China; Department of Gynecology & Obstetrics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Fengshu Zhao
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Di Wu
- Department of Gynecology & Obstetrics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Yunxia Zhang
- Department of Gynecology & Obstetrics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xueyang Bao
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Fangfang Shi
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Yunlang Cai
- Department of Gynecology & Obstetrics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Jun Dou
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing 210009, China.
| |
Collapse
|
12
|
Cheng RYS, Burkett S, Ambs S, Moody T, Wink DA, Ridnour LA. Chronic Exposure to Nitric Oxide Induces P53 Mutations and Malignant-like Features in Human Breast Epithelial Cells. Biomolecules 2023; 13:311. [PMID: 36830680 PMCID: PMC9953427 DOI: 10.3390/biom13020311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 01/31/2023] [Accepted: 02/04/2023] [Indexed: 02/11/2023] Open
Abstract
The small endogenous signaling molecule nitric oxide (NO) has been linked with chronic inflammation and cancer. The effects of NO are both concentration and temporally dependent; under some conditions, NO protects against damage caused by reactive oxygen species and activates P53 signaling. During chronic inflammation, NO causes DNA damage and inhibits repair proteins. To extend our understanding of the roles of NO during carcinogenesis, we investigated the possible effects of chronic NO exposure on MCF10A breast epithelial cells, as defined by changes in cellular morphology, chromosome/genomic stability, RNA, and protein expression, and altered cell phenotypes. Human MCF10A cells were maintained in varying doses of the NO donor DETANO for three weeks. Distinct patterns of genomic modifications in TP53 and KRAS target genes were detected in NO-treated cells when compared to background mutations. In addition, quantitative real-time PCR demonstrated an increase in the expression of cancer stem cell (CSC) marker CD44 after prolonged exposure to 300 μM DETANO. While similar changes in cell morphology were found in cells exposed to 300-500 μM DETANO, cells cultured in 100 μM DETANO exhibited enhanced motility. In addition, 100 μM NO-treated cells proliferated in serum-free media and selected clonal populations and pooled cells formed colonies in soft agar that were clustered and disorganized. These findings show that chronic exposure to NO generates altered breast epithelial cell phenotypes with malignant characteristics.
Collapse
Affiliation(s)
- Robert Y. S. Cheng
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Sandra Burkett
- Molecular Cytogenetics Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Stefan Ambs
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Terry Moody
- Center for Cancer Training Office of Training and Education, National Cancer Institute, Bethesda, MD 20892, USA
| | - David A. Wink
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Lisa A. Ridnour
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| |
Collapse
|
13
|
Lüchtenborg AM, Metzger P, Cosenza Contreras M, Oria V, Biniossek ML, Lindner F, Fröhlich K, Malyi A, Erbes T, Gensch N, Maurer J, Thomsen A, Boerries M, Schilling O, Werner M, Bronsert P. Krüppel-like factor 7 influences translation and pathways involved in ribosomal biogenesis in breast cancer. BREAST CANCER RESEARCH : BCR 2022; 24:65. [PMID: 36192788 PMCID: PMC9531505 DOI: 10.1186/s13058-022-01562-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 09/19/2022] [Indexed: 11/27/2022]
Abstract
Background Ribosomal biogenesis and ribosomal proteins have attracted attention in the context of tumor biology in recent years. Instead of being mere translational machineries, ribosomes might play an active role in tumor initiation and progression. Despite its importance, regulation of ribosomal biogenesis is still not completely understood.
Methods Using Gene Set Enrichment Analysis of RNA sequencing and proteomical mass spectrometry data in breast cancer cells expressing Krüppel-like factor 7 (KLF7), we identified processes altered by this transcription factor. In silico analyses of a cohort of breast cancer patients in The Cancer Genome Atlas confirmed our finding. We further verified the role of KLF7 the identified ribosomal processes in in vitro assays of mammary carcinoma cell lines and analyses of breast cancer patients’ tissue slices.
Results We identified the transcription factor Krüppel-like factor 7 (KLF7) as a regulator of ribosomal biogenesis and translation in breast cancer cells and tissue. Highly significant overlapping processes related to ribosomal biogenesis were identified in proteomics and transcriptomics data and confirmed in patients’ breast cancer RNA Seq data. Further, nucleoli, the sites of ribosomal biogenesis, were morphologically altered and quantitatively increased in KLF7-expressing cells. Pre-rRNA processing was identified as one potential process affected by KLF7. In addition, an increase in global translation independent from proliferation and transcription was observed upon exogenous KLF7 expression in vitro. Importantly, in a cohort of breast cancer patients, KLF7-expression levels correlated with aggressiveness of the intrinsic breast cancer subtype and tumor grading. Moreover, KLF7 correlated with nucleolar characteristics in human breast tumor tissue, indicating a role for KLF7 in ribosomal biogenesis. Conclusion In mammary carcinoma, KLF7 is involved in ribosomal biogenesis. Alterations of ribosomal biogenesis has far reaching quantitative and qualitative implications for the proteome of the cancer cells. This might influence the aggressiveness of cancer cells. Supplementary Information The online version contains supplementary material available at 10.1186/s13058-022-01562-8.
Collapse
Affiliation(s)
- Anne-Marie Lüchtenborg
- Institute for Surgical Pathology, Medical Center - University of Freiburg, Breisacher Straße 115A, 79106, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK) Partner Site Freiburg and Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Patrick Metzger
- Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Institute of Medical Bioinformatics and Systems Medicine, Medical Center - University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Miguel Cosenza Contreras
- Institute for Surgical Pathology, Medical Center - University of Freiburg, Breisacher Straße 115A, 79106, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Victor Oria
- Faculty of Biology, University of Freiburg, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine, Faculty of Biology, University of Freiburg, Freiburg, Germany.,Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Martin L Biniossek
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Franziska Lindner
- Institute for Surgical Pathology, Medical Center - University of Freiburg, Breisacher Straße 115A, 79106, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Klemens Fröhlich
- Institute for Surgical Pathology, Medical Center - University of Freiburg, Breisacher Straße 115A, 79106, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Ambrus Malyi
- 2Nd Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Thalia Erbes
- Department of Obstetrics and Gynecology, Medical Center - University of Freiburg, Freiburg, Germany
| | - Nicole Gensch
- Core Facility Signaling Factory, BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany
| | - Jochen Maurer
- Department of Obstetrics and Gynecology, University Hospital Aachen (UKA), Aachen, Germany
| | - Andreas Thomsen
- Department of Radiation Oncology, Medical Center - University of Freiburg, Freiburg, Germany
| | - Melanie Boerries
- Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK) Partner Site Freiburg and Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute of Medical Bioinformatics and Systems Medicine, Medical Center - University of Freiburg, Freiburg, Germany
| | - Oliver Schilling
- Institute for Surgical Pathology, Medical Center - University of Freiburg, Breisacher Straße 115A, 79106, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK) Partner Site Freiburg and Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martin Werner
- Institute for Surgical Pathology, Medical Center - University of Freiburg, Breisacher Straße 115A, 79106, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK) Partner Site Freiburg and Cancer Research Center (DKFZ), Heidelberg, Germany.,Tumorbank Comprehensive Cancer Center Freiburg, Medical Center - University of Freiburg, Freiburg, Germany.,Core Facility for Histopathology and Digital Pathology, Medical Center - University of Freiburg, Freiburg, Germany
| | - Peter Bronsert
- Institute for Surgical Pathology, Medical Center - University of Freiburg, Breisacher Straße 115A, 79106, Freiburg, Germany. .,Faculty of Medicine, University of Freiburg, Freiburg, Germany. .,German Cancer Consortium (DKTK) Partner Site Freiburg and Cancer Research Center (DKFZ), Heidelberg, Germany. .,Core Facility for Histopathology and Digital Pathology, Medical Center - University of Freiburg, Freiburg, Germany.
| |
Collapse
|
14
|
Luo W, Lin S, Huang Y, Zhu K, Zhang F, Lin J, Qin Y, Zhou Z, Wu W, Liu C. Bioinformatic Analysis and In Vitro and In Vivo Experiments Reveal That Fibrillarin Participates in the Promotion of Lung Metastasis in Hepatocellular Carcinoma. Bioengineering (Basel) 2022; 9:bioengineering9080396. [PMID: 36004921 PMCID: PMC9405174 DOI: 10.3390/bioengineering9080396] [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: 06/23/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 12/24/2022] Open
Abstract
Lung metastasis, the most frequent metastatic pattern in hepatocellular carcinoma, is an important contributor to poor prognosis. However, the mechanisms responsible for lung metastasis in hepatocellular carcinoma remain unknown. Aiming to explore these mechanisms, weighted gene coexpression network analysis (WGCNA) was firstly used to find hub genes related to lung metastasis. Then, we obtained 67 genes related to lung metastasis in hepatocellular carcinoma which were mainly related to ribosomal pathways and functions, and a protein interaction network analysis identified that fibrillarin (FBL) might be an important hub gene. Furthermore, we found that FBL is highly expressed in hepatocellular carcinoma and that its high expression increases the rate of lung metastasis and indicates a poor prognosis. Knockdown of FBL could significantly reduce proliferation and stemness as well as inhibiting the migration and invasion of hepatocellular carcinoma cells. Moreover, we found that FBL might be involved in the regulation of MYC and E2F pathways in hepatocellular carcinoma. Finally, we demonstrated that the knockdown of FBL could suppress hepatocellular carcinoma cell growth in vivo. In conclusion, ribosome-biogenesis-related proteins, especially Fibrillarin, play important roles in lung metastasis from hepatocellular carcinoma.
Collapse
Affiliation(s)
- Weixin Luo
- Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Shusheng Lin
- Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Yipei Huang
- Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Ke Zhu
- Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Fapeng Zhang
- Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Junlong Lin
- Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Yufei Qin
- Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Ziyu Zhou
- Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Wenrui Wu
- Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Correspondence:
| | - Chao Liu
- Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| |
Collapse
|
15
|
Azevedo ALKD, Gomig THB, Giner IS, Batista M, Marchini FK, Lima RS, de Andrade Urban C, Sebastião APM, Cavalli IJ, Ribeiro EMDSF. Comprehensive analysis of the large and small ribosomal proteins in breast cancer: Insights on proteomic and transcriptomic expression patterns, regulation, mutational landscape, and prognostic significance. Comput Biol Chem 2022; 100:107746. [DOI: 10.1016/j.compbiolchem.2022.107746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 07/11/2022] [Accepted: 07/25/2022] [Indexed: 11/29/2022]
|
16
|
Salihi A, Al-Naqshabandi MA, Khudhur ZO, Housein Z, Hama HA, Abdullah RM, Hussen BM, Alkasalias T. Gasotransmitters in the tumor microenvironment: Impacts on cancer chemotherapy (Review). Mol Med Rep 2022; 26:233. [PMID: 35616143 PMCID: PMC9178674 DOI: 10.3892/mmr.2022.12749] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 04/07/2022] [Indexed: 11/23/2022] Open
Abstract
Nitric oxide, carbon monoxide and hydrogen sulfide are three endogenous gasotransmitters that serve a role in regulating normal and pathological cellular activities. They can stimulate or inhibit cancer cell proliferation and invasion, as well as interfere with cancer cell responses to drug treatments. Understanding the molecular pathways governing the interactions between these gases and the tumor microenvironment can be utilized for the identification of a novel technique to disrupt cancer cell interactions and may contribute to the conception of effective and safe cancer therapy strategies. The present review discusses the effects of these gases in modulating the action of chemotherapies, as well as prospective pharmacological and therapeutic interfering approaches. A deeper knowledge of the mechanisms that underpin the cellular and pharmacological effects, as well as interactions, of each of the three gases could pave the way for therapeutic treatments and translational research.
Collapse
Affiliation(s)
- Abbas Salihi
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region 44001, Iraq
- Center of Research and Strategic Studies, Lebanese French University, Erbil, Kurdistan Region 44002, Iraq
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum, Karolinska Institutet, SE-17165 Stockholm, Sweden
| | - Mohammed A. Al-Naqshabandi
- Department of Clinical Biochemistry, College of Health Sciences, Hawler Medical University, Erbil, Kurdistan Region 44001, Iraq
| | - Zhikal Omar Khudhur
- Department of Medical Analysis, Faculty of Applied Science, Tishk International University, Erbil, Kurdistan Region 44001, Iraq
| | - Zjwan Housein
- Department of Medical Laboratory Technology, Technical Health and Medical College, Erbil Polytechnique University, Erbil, Kurdistan Region 44002, Iraq
| | - Harmand A. Hama
- Department of Biology, Faculty of Education, Tishk International University, Erbil, Kurdistan Region 44002, Iraq
| | - Ramyar M. Abdullah
- College of Medicine, Hawler Medical University, Erbil, Kurdistan Region 44002, Iraq
| | - Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Erbil, Kurdistan Region 44002, Iraq
| | - Twana Alkasalias
- General Directorate of Scientific Research Center, Salahaddin University-Erbil, Erbil, Kurdistan Region 44002, Iraq
- Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, SE-17176 Stockholm, Sweden
| |
Collapse
|
17
|
Nguyen TTA, Postlewait LM, Zhang C, Meisel JL, O'Regan R, Badve S, Kalinsky K, Li X. Utility of Oncotype DX score in clinical management for T1 estrogen receptor positive, HER2 negative, and lymph node negative breast cancer. Breast Cancer Res Treat 2022; 192:509-516. [PMID: 35084624 DOI: 10.1007/s10549-022-06530-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/17/2022] [Indexed: 12/16/2022]
Abstract
BACKGROUND The management of estrogen receptor positive (ER+)/HER2- and lymph node (LN) negative breast cancers can be influenced by Oncotype DX recurrence score (RS) in the USA. However, the benefit of RS in T1 tumors (≤ 1 cm) is not clear. METHODS We retrieved 199 T1 ER+/HER2-/LN- breast cancer diagnosed between 1993 and 2016 that had undergone RS testing. The median follow-up time was 51 months. We examined the disease-free survival (DFS) and distant metastasis and their association with RS and other clinicopathologic features. RESULTS Of the 199 cases, 40 were T1a (≤ 0.5 cm) and 159 were T1b (> 0.5 cm to 1 cm) tumors. In the 40 T1a tumors, 11 would benefit from chemotherapy by the TAILORx study results. Of these T1a tumors, 36 were Nottingham grade 1/2, 3 were grade 3, and 1 was microinvasive carcinoma; 2 (5%) had local recurrence and 1 (2.5%) had distant metastasis to the bone. The only patient with T1a tumor (Nottingham grade 3, RS = 42) and distant metastasis to bone had received adjuvant chemotherapy. In the 159 T1b tumors, 25 would benefit chemotherapy by the TAILORx results. Of the T1b tumors, 149 were Nottingham grade 1/2 and 10 were grade 3. Nine (5.7%) had local recurrence and 2 (1.3%) had distant metastasis to bone and mediastinum, respectively. The two T1b tumors with distant metastasis had a RS 20 and Nottingham grade 2, and RS 27 and Nottingham grade 3, respectively. Both patients received adjuvant chemotherapy. In multivariate analysis of the entire cohort (T1a and T1b tumors), Nottingham tumor grade and receiving chemotherapy were significantly associated with DFS. In univariate analysis of the entire cohort, Nottingham tumor grade, receiving adjuvant chemotherapy, and RS were significantly associated with distant metastasis. CONCLUSION This study demonstrates that the metastatic rate of T1a and T1b ER+/HER2-/LN- breast cancer is very low. Patients with low grade (1 or 2), T1a ER+/HER2-/LN- breast cancer may not need RS for treatment decision-making; however, in patients with high-grade T1a or T1b ER+/HER2-/LN- breast cancer, RS analysis should be strongly considered.
Collapse
Affiliation(s)
- Thi Truc Anh Nguyen
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, USA
| | - Lauren M Postlewait
- Division of Surgical Oncology, Department of Surgery, Emory University, Atlanta, GA, USA
| | - Chao Zhang
- Pediatric Biostatistics Core, Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Jane L Meisel
- Department of Hematology and Oncology, Emory University, Atlanta, GA, USA
| | - Ruth O'Regan
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Sunil Badve
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, USA
| | - Kevin Kalinsky
- Department of Hematology and Oncology, Emory University, Atlanta, GA, USA
| | - Xiaoxian Li
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, USA.
| |
Collapse
|
18
|
Deregulation of ribosomal proteins in human cancers. Biosci Rep 2021; 41:230380. [PMID: 34873618 PMCID: PMC8685657 DOI: 10.1042/bsr20211577] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 10/28/2021] [Accepted: 11/22/2021] [Indexed: 12/26/2022] Open
Abstract
The ribosome, the site for protein synthesis, is composed of ribosomal RNAs (rRNAs) and ribosomal proteins (RPs). The latter have been shown to have many ribosomal and extraribosomal functions. RPs are implicated in a variety of pathological processes, especially tumorigenesis and cell transformation. In this review, we will focus on the recent advances that shed light on the effects of RPs deregulation in different types of cancer and their roles in regulating the tumor cell fate.
Collapse
|
19
|
Zou Q, Yang L, Shi R, Qi Y, Zhang X, Qi H. Proteostasis regulated by testis-specific ribosomal protein RPL39L maintains mouse spermatogenesis. iScience 2021; 24:103396. [PMID: 34825148 PMCID: PMC8605100 DOI: 10.1016/j.isci.2021.103396] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/20/2021] [Accepted: 10/27/2021] [Indexed: 01/03/2023] Open
Abstract
Maintaining proteostasis is important for animal development. How proteostasis influences spermatogenesis that generates male gametes, spermatozoa, is not clear. We show that testis-specific paralog of ribosomal large subunit protein RPL39, RPL39L, is required for mouse spermatogenesis. Deletion of Rpl39l in mouse caused reduced proliferation of spermatogonial stem cells, malformed sperm mitochondria and flagella, leading to sub-fertility in males. Biochemical analyses revealed that lack of RPL39L deteriorated protein synthesis and protein quality control in spermatogenic cells, partly due to reduced biogenesis of ribosomal subunits and ribosome homeostasis. RPL39/RPL39L is likely assembled into ribosomes via H/ACA domain containing NOP10 complex early in ribosome biogenesis pathway. Furthermore, Rpl39l null mice exhibited compromised regenerative spermatogenesis after chemical insult and early degenerative spermatogenesis in aging mice. These data demonstrate that maintaining proteostasis is important for spermatogenesis, of which ribosome homeostasis maintained by ribosomal proteins coordinates translation machinery to the regulation of cellular growth. Rpl39l deletion causes reduced spermatogenesis and subfertility in male mice SSC proliferation, mitochondria and sperm flagella compromised in Rpl39l–/– mice Rpl39l deletion affects ribosomal LSU formation and protein quality control Aberrant proteostasis affects spermatogenesis and regeneration
Collapse
Affiliation(s)
- Qianxing Zou
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510630, China.,Center for Cell Lineage and Development, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510630, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510630, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lele Yang
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510630, China.,Center for Cell Lineage and Development, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510630, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510630, China
| | - Ruona Shi
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510630, China.,Center for Cell Lineage and Development, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510630, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510630, China.,Department of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230000, China
| | - Yuling Qi
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510630, China.,Center for Cell Lineage and Development, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510630, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510630, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,Center for Cell Lineage and Atlas (CCLA), Bioland Laboratory, Guangzhou 510630, China
| | - Xiaofei Zhang
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510630, China.,Center for Cell Lineage and Development, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510630, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510630, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,Center for Cell Lineage and Atlas (CCLA), Bioland Laboratory, Guangzhou 510630, China
| | - Huayu Qi
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510630, China.,Center for Cell Lineage and Development, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510630, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510630, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
20
|
Chung AW, Anand K, Anselme AC, Chan AA, Gupta N, Venta LA, Schwartz MR, Qian W, Xu Y, Zhang L, Kuhn J, Patel T, Rodriguez AA, Belcheva A, Darcourt J, Ensor J, Bernicker E, Pan PY, Chen SH, Lee DJ, Niravath PA, Chang JC. A phase 1/2 clinical trial of the nitric oxide synthase inhibitor L-NMMA and taxane for treating chemoresistant triple-negative breast cancer. Sci Transl Med 2021; 13:eabj5070. [PMID: 34910551 DOI: 10.1126/scitranslmed.abj5070] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Andrew W Chung
- Texas A&M University Health Science Center, Bryan, TX 77807, USA.,Houston Methodist Research Institute, Houston, TX 77030, USA.,Houston Methodist Cancer Center, Houston, TX 77030, USA
| | - Kartik Anand
- Houston Methodist Cancer Center, Houston, TX 77030, USA
| | - Ann C Anselme
- Texas A&M University Health Science Center, Bryan, TX 77807, USA.,Houston Methodist Research Institute, Houston, TX 77030, USA.,Houston Methodist Cancer Center, Houston, TX 77030, USA
| | | | - Nakul Gupta
- Department of Radiology, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Luz A Venta
- Department of Radiology, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Mary R Schwartz
- Houston Methodist Department of Pathology and Genomic Medicine, Houston, TX 77030, USA
| | - Wei Qian
- Houston Methodist Research Institute, Houston, TX 77030, USA.,Houston Methodist Cancer Center, Houston, TX 77030, USA
| | - Yitian Xu
- Houston Methodist Research Institute, Houston, TX 77030, USA.,Houston Methodist Cancer Center, Houston, TX 77030, USA
| | - Licheng Zhang
- Houston Methodist Research Institute, Houston, TX 77030, USA.,Houston Methodist Cancer Center, Houston, TX 77030, USA
| | - John Kuhn
- University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Tejal Patel
- Houston Methodist Cancer Center, Houston, TX 77030, USA.,Department of General Oncology MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Anna Belcheva
- Houston Methodist Cancer Center, Houston, TX 77030, USA
| | | | - Joe Ensor
- Houston Methodist Cancer Center, Houston, TX 77030, USA
| | | | - Ping-Ying Pan
- Houston Methodist Research Institute, Houston, TX 77030, USA.,Houston Methodist Cancer Center, Houston, TX 77030, USA
| | - Shu Hsia Chen
- Houston Methodist Research Institute, Houston, TX 77030, USA.,Houston Methodist Cancer Center, Houston, TX 77030, USA
| | - Delphine J Lee
- Lundquist Institute, Torrance, CA 90502, USA.,David Geffen School of Medicine at Los Angeles, CA 90095, USA
| | | | - Jenny C Chang
- Houston Methodist Research Institute, Houston, TX 77030, USA.,Houston Methodist Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
21
|
Wang Z, Li Z, Zhou K, Wang C, Jiang L, Zhang L, Yang Y, Luo W, Qiao W, Wang G, Ni Y, Dai S, Guo T, Ji G, Xu M, Liu Y, Su Z, Che G, Li W. Deciphering cell lineage specification of human lung adenocarcinoma with single-cell RNA sequencing. Nat Commun 2021; 12:6500. [PMID: 34764257 PMCID: PMC8586023 DOI: 10.1038/s41467-021-26770-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 10/22/2021] [Indexed: 02/08/2023] Open
Abstract
Lung adenocarcinomas (LUAD) arise from precancerous lesions such as atypical adenomatous hyperplasia, which progress into adenocarcinoma in situ and minimally invasive adenocarcinoma, then finally into invasive adenocarcinoma. The cellular heterogeneity and molecular events underlying this stepwise progression remain unclear. In this study, we perform single-cell RNA sequencing of 268,471 cells collected from 25 patients in four histologic stages of LUAD and compare them to normal cell types. We detect a group of cells closely resembling alveolar type 2 cells (AT2) that emerged during atypical adenomatous hyperplasia and whose transcriptional profile began to diverge from that of AT2 cells as LUAD progressed, taking on feature characteristic of stem-like cells. We identify genes related to energy metabolism and ribosome synthesis that are upregulated in early stages of LUAD and may promote progression. MDK and TIMP1 could be potential biomarkers for understanding LUAD pathogenesis. Our work shed light on the underlying transcriptional signatures of distinct histologic stages of LUAD progression and our findings may facilitate early diagnosis.
Collapse
Affiliation(s)
- Zhoufeng Wang
- grid.412901.f0000 0004 1770 1022Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan China ,grid.412901.f0000 0004 1770 1022Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan China ,grid.412901.f0000 0004 1770 1022Research Units of West China, Chinese Academy of Medical Sciences, West China Hospital, Chengdu, Sichuan China
| | - Zhe Li
- Singlera Genomics Ltd, Shanghai, China
| | - Kun Zhou
- grid.13402.340000 0004 1759 700XDepartment of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China ,grid.412901.f0000 0004 1770 1022Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan China
| | - Chengdi Wang
- grid.412901.f0000 0004 1770 1022Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan China
| | - Lili Jiang
- grid.412901.f0000 0004 1770 1022Department of Pathology, West China Hospital of Sichuan University, Chengdu, China
| | - Li Zhang
- grid.412901.f0000 0004 1770 1022Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan China
| | - Ying Yang
- grid.412901.f0000 0004 1770 1022Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan China
| | - Wenxin Luo
- grid.412901.f0000 0004 1770 1022Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan China
| | - Wenliang Qiao
- grid.412901.f0000 0004 1770 1022Lung Cancer Center, West China Hospital Sichuan University, Chengdu, Sichuan China
| | - Gang Wang
- grid.412901.f0000 0004 1770 1022Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan China
| | - Yinyun Ni
- grid.412901.f0000 0004 1770 1022Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan China
| | - Shuiping Dai
- grid.412901.f0000 0004 1770 1022Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan China
| | - Tingting Guo
- grid.412901.f0000 0004 1770 1022Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan China
| | - Guiyi Ji
- grid.412901.f0000 0004 1770 1022Health Management Center, West China Hospital, Sichuan University, Chengdu, Sichuan China
| | - Minjie Xu
- Singlera Genomics Ltd, Shanghai, China
| | | | - Zhixi Su
- Singlera Genomics Ltd, Shanghai, China
| | - Guowei Che
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Weimin Li
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China. .,Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China. .,Research Units of West China, Chinese Academy of Medical Sciences, West China Hospital, Chengdu, Sichuan, China.
| |
Collapse
|
22
|
Chung WM, Molony RD, Lee YF. Non-stem bladder cancer cell-derived extracellular vesicles promote cancer stem cell survival in response to chemotherapy. Stem Cell Res Ther 2021; 12:533. [PMID: 34627375 PMCID: PMC8502272 DOI: 10.1186/s13287-021-02600-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 09/23/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Chemosenstive non-stem cancer cells (NSCCs) constitute the bulk of tumors and are considered as part of the cancer stem cell (CSC) niche in the tumor microenvironment (TME). Tumor-derived extracellular vesicles (EVs) mediate the communication between tumors and the TME. In this study, we sought to investigate the impacts of EVs released by NSCCs on the maintenance of CSC properties and chemoresistance. METHODS We employed murine MB49 bladder cancer (BC) sub-lines representing CSCs and NSCCs as a model system. Chemotherapy drugs were used to treat NSCCs in order to collect conditioned EVs. The impacts of NSCC-derived EVs on CSC progression were evaluated through sphere formation, cytotoxicity, migration, and invasion assays, and by analyzing surface marker expression on these BC cells. Differential proteomic analyses were conducted to identify cargo protein candidates involved in the EV-mediated communication between NSCCs and CSCs. RESULTS NSCC-derived EVs contained cargo proteins enriched in proteostasis-related functions, and significantly altered the development of CSCs such that they were more intrinsically chemoresistant, aggressive, and better able to undergo self-renewal. CONCLUSIONS We thus identified a novel communication mechanism whereby NSCC-EVs can alter the relative fitness of CSCs to promote disease progression and the acquisition of chemoresistance.
Collapse
Affiliation(s)
- Wei-Min Chung
- Department of Urology, University of Rochester Medical Center, 601 Elmwood Ave, Box 656, Rochester, NY, 14642, USA
| | - Ryan D Molony
- Department of Urology, University of Rochester Medical Center, 601 Elmwood Ave, Box 656, Rochester, NY, 14642, USA
| | - Yi-Fen Lee
- Department of Urology, University of Rochester Medical Center, 601 Elmwood Ave, Box 656, Rochester, NY, 14642, USA.
- Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA.
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
23
|
Zou Q, Qi H. Deletion of ribosomal paralogs Rpl39 and Rpl39l compromises cell proliferation via protein synthesis and mitochondrial activity. Int J Biochem Cell Biol 2021; 139:106070. [PMID: 34428590 DOI: 10.1016/j.biocel.2021.106070] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 07/25/2021] [Accepted: 08/19/2021] [Indexed: 11/15/2022]
Abstract
Accumulating evidences suggest that the composition and functional roles of ribosomes are heterogeneous in cells, partly due to the temporal-spatial expression of paralogous ribosomal proteins (RPs), of which functional relationships remain largely unexplored. In mouse, the X chromosome-linked RPL39 and its male germline specific paralog RPL39L are thought to express mutually exclusively due to the meiotic sex chromosome inactivation, hinders the understanding of their functional relationships. In the present study, we investigated the expression and functional relations of Rpl39 and Rpl39l in a proliferative mouse cell line, in which both genes are expressed simultaneously, with the expression level of Rpl39 higher than that of Rpl39l. Disruption of Rpl39 via CRISPR/Cas9 method caused decreased cell proliferation, nascent protein synthesis and altered mitochondrial functions, whereas double mutations of Rpl39 and Rpl39l augmented these phenotypes, suggesting that both RPs contribute to the cellular physiology. Consistently, overexpression of Rpl39, Rpl39l or an alanine mutant of RPL39, rescued cell proliferation similarly in Rpl39-/-::Rpl39l-/- dual gene null cells. Deletion of Rpl39l induced compensatory expression of Rpl39, rendering the deleterious effects of Rpl39l mutation. Supporting this, Rpl39l mutation was more detrimental to cells under a low serum condition, under which the compensatory expression of Rpl39 was inhibited. Moreover, the low serum condition induced expression of both genes, suggesting that they possess stress responsive roles. Taken together, these data indicate that both RPL39 and RPL39L influence cell proliferation via protein synthesis and mitochondrial functions, suggesting a link between protein translation and cellular metabolism through these ribosomal protein paralogs.
Collapse
Affiliation(s)
- Qianxing Zou
- CAS Key Laboratory of Regenerative Biology; Center for Cell Lineage and Development; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huayu Qi
- CAS Key Laboratory of Regenerative Biology; Center for Cell Lineage and Development; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
24
|
Transcriptome profiling reveals new insights into the roles of neuronal nitric oxide synthase on macrophage polarization towards classically activated phenotype. PLoS One 2021; 16:e0257908. [PMID: 34587205 PMCID: PMC8480887 DOI: 10.1371/journal.pone.0257908] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 09/13/2021] [Indexed: 01/09/2023] Open
Abstract
In response to various stimuli, naïve macrophages usually polarize to M1 (classically activated) or M2 (alternatively activated) cells with distinct biological functions. Neuronal nitric oxide synthase (NOS1) is involved in M1 macrophage polarization at an early stage. Here, we show for the first time that NOS1 is dispensable for M2 macrophage polarization for the first time. Further, differentially expressed genes (DEGs) regulated by NOS1 signaling in M1-polarized macrophages stimulated with lipopolysaccharide (LPS) were characterized by transcriptome analysis of wild-type (WT) and NOS1 knockout mouse macrophages. Thousands of affected genes were detected 2 h post LPS challenge, and this wide-ranging effect became greater with a longer stimulation time (8 h post LPS). NOS1 deficiency caused dysregulated expression of hundreds of LPS-responsive genes. Most DEGs were enriched in biological processes related to transcription and regulation of the immune and inflammatory response. At 2 h post-LPS, the toll-like receptor (TLR) signaling pathway, cytokine-cytokine receptor interaction, and NOD-like receptor signaling pathway were the major pathways affected, whereas the main pathways affected at 8 h post-LPS were Th1 and Th2 cell differentiation, FoxO, and AMPK signaling pathway. Identified DEGs were validated by real-time quantitative PCR and interacted in a complicated signaling pathway network. Collectively, our data show that NOS1 is dispensable for M2 macrophage polarization and reveal novel insights in the role of NOS1 signaling at different stages of M1 macrophage polarization through distinct TLR4 plasma membrane-localized and endosome-internalized signaling pathways.
Collapse
|
25
|
Ogunleye AJ, Romanova E, Medvedeva YA. Genome-wide regulation of CpG methylation by ecCEBPα in acute myeloid leukemia. F1000Res 2021; 10:204. [PMID: 34557292 PMCID: PMC8444155 DOI: 10.12688/f1000research.28146.2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/23/2021] [Indexed: 01/08/2023] Open
Abstract
Background: Acute myeloid leukemia (AML) is a hematopoietic malignancy characterized by genetic and epigenetic aberrations that alter the differentiation capacity of myeloid progenitor cells. The transcription factor
CEBPα is frequently mutated in AML patients leading to an increase in DNA methylation in many genomic locations. Previously, it has been shown that
ecCEBPα (extra coding CEBP
α) - a lncRNA transcribed in the same direction as
CEBPα gene - regulates DNA methylation of
CEBPα promoter in
cis. Here, we hypothesize that
ecCEBPα could participate in the regulation of DNA methylation in
trans. Method: First, we retrieved the methylation profile of AML patients with mutated
CEBPα locus from The Cancer Genome Atlas (TCGA). We then predicted the
ecCEBPα secondary structure in order to check the potential of
ecCEBPα to form triplexes around CpG loci and checked if triplex formation influenced CpG methylation, genome-wide. Results: Using DNA methylation profiles of AML patients with a mutated
CEBPα locus, we show that
ecCEBPα could interact with DNA by forming DNA:RNA triple helices and protect regions near its binding sites from global DNA methylation. Further analysis revealed that triplex-forming oligonucleotides in
ecCEBPα are structurally unpaired supporting the DNA-binding potential of these regions.
ecCEBPα triplexes supported with the RNA-chromatin co-localization data are located in the promoters of leukemia-linked transcriptional factors such as MLF2. Discussion: Overall, these results suggest a novel regulatory mechanism for
ecCEBPα as a genome-wide epigenetic modulator through triple-helix formation which may provide a foundation for sequence-specific engineering of RNA for regulating methylation of specific genes.
Collapse
Affiliation(s)
- Adewale J Ogunleye
- Department of Biological and Medical Physics, Moscow Institute of Physics and Technology, Moscow, Russian Federation
| | - Ekaterina Romanova
- Research Center of Biotechnology, Institute of Bioengineering, Russian Academy of Sciences, Moscow, Russian Federation
| | - Yulia A Medvedeva
- Department of Biological and Medical Physics, Moscow Institute of Physics and Technology, Moscow, Russian Federation.,Research Center of Biotechnology, Institute of Bioengineering, Russian Academy of Sciences, Moscow, Russian Federation
| |
Collapse
|
26
|
Zhao W, Li X, Nian W, Wang J, Wang X, Sun L, Zhu Y, Tong Z. Ribosome Proteins Represented by RPL27A Mark the Development and Metastasis of Triple-Negative Breast Cancer in Mouse and Human. Front Cell Dev Biol 2021; 9:716730. [PMID: 34497807 PMCID: PMC8419227 DOI: 10.3389/fcell.2021.716730] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 07/28/2021] [Indexed: 12/14/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is known to have a poor prognosis and limited treatment options. The lack of targeted therapies and poor prognosis of patients with TNBC have made it urgent to discover novel critical diagnosis and therapeutic targets in the TNBC field. Here, in the current study, we integrated the single-cell RNA-sequencing (scRNA-seq) data from four normal mouse mammary tissues and four mouse breast tumors. Comparative analysis was conducted to identify the gene profiles of normal epithelial cells and cancer cells at different models. Surprisingly, two ribosomal protein genes, Rpl27a and Rpl15, were significantly upregulated in the cancer cells in all the TNBC models. Next, we accessed the scRNA-seq data from human primary and metastatic TNBC tissues, and comparative analysis revealed gene profiles of human primary and metastatic TNBC cancer cells. Ribosomal protein genes, represented by RPL27A and RPL15, showed significantly upregulated expression in metastatic TNBC cancer cells. Pathway analysis on the upregulated genes of the metastatic TNBC cancer cells identified the key regulators and signaling pathways that were driving the metastasis of the TNBC cancer cells. Specifically, EIF2 signaling was significantly activated, and major member genes of this signaling pathway were upregulated. In vitro study revealed that targeting RPL27A or EIF2 signaling in a TNBC cell line, MDA-MB-231, significantly reduced cell migration and invasion. Altogether, these data suggested that the RPL27A gene is conducting critical functions in TNBC cancer development and metastasis and is a potential therapeutic target for TNBC.
Collapse
Affiliation(s)
- Weipeng Zhao
- Key Laboratory of Cancer Prevention and Therapy, Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Xichuan Li
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, Tianjin, China
| | - Weiqi Nian
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Jun Wang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Xiaorui Wang
- Key Laboratory of Cancer Prevention and Therapy, Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Linlin Sun
- Key Laboratory of Cancer Prevention and Therapy, Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Ye Zhu
- Key Laboratory of Cancer Prevention and Therapy, Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Zhongsheng Tong
- Key Laboratory of Cancer Prevention and Therapy, Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
27
|
Jie Q, Sun F, Li Q, Zhu J, Wei Y, Yang H, Long P, Wang Z, Yang X, Li D, Huang L, Ma Y. Downregulated ribosomal protein L39 inhibits trophoblast cell migration and invasion by targeting E-cadherin in the placenta of patients with preeclampsia. FASEB J 2021; 35:e21322. [PMID: 33710681 DOI: 10.1096/fj.202002061r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/06/2020] [Accepted: 12/14/2020] [Indexed: 12/27/2022]
Abstract
Early-onset preeclampsia (PE) is a pregnancy complication that can lead to severe adverse maternal and fetal outcomes. However, the mechanisms underlying the development of early-onset PE are not fully understood. Ribosomal protein L39 (RPL39) is a member of the S39E family of ribosomal proteins that plays an important role in stem cell self-renewal, cancer metastasis, and chemoresistance. In this study, we aimed to explore the potential function of RPL39 in placental trophoblast cells. We analyzed the expression of RPL39 in early-onset PE and normal placental tissues using real-time PCR, western blot analysis, and immunohistochemistry. The results showed that RPL39 was markedly downregulated in early-onset PE placental tissues. RPL39 knockdown inhibited trophoblast cell proliferation, migration, and invasion, as well as placental explant outgrowth. Flow cytometry analysis suggested that knockdown of RPL39 resulted in cell cycle arrest at the G0/G1 phase, but had no significant effect on cell apoptosis. We also found that RPL39 knockdown could alter cell morphology. We then measured the expression of the epithelial cell marker E-cadherin following knockdown of RPL39 in Bewo and HTR8/SVneo cells. RPL39 knockdown increased the expression of E-cadherin. Furthermore, E-cadherin expression was upregulated in placental explant outgrowth tissues transfected with RPL39 small interfering RNA. In conclusion, RPL39 plays an essential role in proliferation, invasion, and migration of trophoblast cells by targeting E-cadherin. Our findings provide novel insight into the mechanisms underlying the occurrence of early-onset PE.
Collapse
Affiliation(s)
- Qiuling Jie
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Hainan Provincial Key Laboratory for human reproductive medicine and Genetic Research, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Reproductive Medicine, the First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Fei Sun
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Hainan Provincial Key Laboratory for human reproductive medicine and Genetic Research, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Reproductive Medicine, the First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Qi Li
- Department of Hainan Provincial Key Laboratory for human reproductive medicine and Genetic Research, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Reproductive Medicine, the First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Juan Zhu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Hainan Provincial Key Laboratory for human reproductive medicine and Genetic Research, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Reproductive Medicine, the First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Yunjian Wei
- Department of Hainan Provincial Key Laboratory for human reproductive medicine and Genetic Research, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Reproductive Medicine, the First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Huamei Yang
- Department of Hainan Provincial Key Laboratory for human reproductive medicine and Genetic Research, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Reproductive Medicine, the First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Ping Long
- Department of Hainan Provincial Key Laboratory for human reproductive medicine and Genetic Research, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Reproductive Medicine, the First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Zhen Wang
- Department of Hainan Provincial Key Laboratory for human reproductive medicine and Genetic Research, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Reproductive Medicine, the First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Xiaohui Yang
- Department of Hainan Provincial Key Laboratory for human reproductive medicine and Genetic Research, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Reproductive Medicine, the First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Dan Li
- Department of Hainan Provincial Key Laboratory for human reproductive medicine and Genetic Research, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Reproductive Medicine, the First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Liping Huang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yanlin Ma
- Department of Hainan Provincial Key Laboratory for human reproductive medicine and Genetic Research, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Reproductive Medicine, the First Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
28
|
Ribosome Biogenesis and Cancer: Overview on Ribosomal Proteins. Int J Mol Sci 2021; 22:ijms22115496. [PMID: 34071057 PMCID: PMC8197113 DOI: 10.3390/ijms22115496] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/16/2021] [Accepted: 05/19/2021] [Indexed: 12/11/2022] Open
Abstract
Cytosolic ribosomes (cytoribosomes) are macromolecular ribonucleoprotein complexes that are assembled from ribosomal RNA and ribosomal proteins, which are essential for protein biosynthesis. Mitochondrial ribosomes (mitoribosomes) perform translation of the proteins essential for the oxidative phosphorylation system. The biogenesis of cytoribosomes and mitoribosomes includes ribosomal RNA processing, modification and binding to ribosomal proteins and is assisted by numerous biogenesis factors. This is a major energy-consuming process in the cell and, therefore, is highly coordinated and sensitive to several cellular stressors. In mitochondria, the regulation of mitoribosome biogenesis is essential for cellular respiration, a process linked to cell growth and proliferation. This review briefly overviews the key stages of cytosolic and mitochondrial ribosome biogenesis; summarizes the main steps of ribosome biogenesis alterations occurring during tumorigenesis, highlighting the changes in the expression level of cytosolic ribosomal proteins (CRPs) and mitochondrial ribosomal proteins (MRPs) in different types of tumors; focuses on the currently available information regarding the extra-ribosomal functions of CRPs and MRPs correlated to cancer; and discusses the role of CRPs and MRPs as biomarkers and/or molecular targets in cancer treatment.
Collapse
|
29
|
Context-specific action of macrolide antibiotics on the eukaryotic ribosome. Nat Commun 2021; 12:2803. [PMID: 33990576 PMCID: PMC8121947 DOI: 10.1038/s41467-021-23068-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 04/14/2021] [Indexed: 01/09/2023] Open
Abstract
Macrolide antibiotics bind in the nascent peptide exit tunnel of the bacterial ribosome and prevent polymerization of specific amino acid sequences, selectively inhibiting translation of a subset of proteins. Because preventing translation of individual proteins could be beneficial for the treatment of human diseases, we asked whether macrolides, if bound to the eukaryotic ribosome, would retain their context- and protein-specific action. By introducing a single mutation in rRNA, we rendered yeast Saccharomyces cerevisiae cells sensitive to macrolides. Cryo-EM structural analysis showed that the macrolide telithromycin binds in the tunnel of the engineered eukaryotic ribosome. Genome-wide analysis of cellular translation and biochemical studies demonstrated that the drug inhibits eukaryotic translation by preferentially stalling ribosomes at distinct sequence motifs. Context-specific action markedly depends on the macrolide structure. Eliminating macrolide-arrest motifs from a protein renders its translation macrolide-tolerant. Our data illuminate the prospects of adapting macrolides for protein-selective translation inhibition in eukaryotic cells.
Collapse
|
30
|
Zhou S, Liu Y, Jin L, Guo P, Liu Q, Shan J, Luo X, He H, Ma W, Zhang T. Estrogen enhances the cytotoxicity of PARP inhibitors on breast cancer cells through stimulating nitric oxide production. J Steroid Biochem Mol Biol 2021; 209:105853. [PMID: 33617965 DOI: 10.1016/j.jsbmb.2021.105853] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 02/10/2021] [Accepted: 02/16/2021] [Indexed: 02/08/2023]
Abstract
Inhibition of Poly(ADP-ribose) polymerase (PARP) is effective for breast cancer susceptibility genes 1 (BRCA1)-deficient breast cancers. Although hormones play critical roles on the occurrence as well as being used in conventional therapies of breast cancer, their impacts on PARP-targeted therapy have been poorly addressed. Here, we showed that addition of estrogen could enhance the cytotoxicity of PARP inhibitors on estrogen receptor (ER)-positive breast cancer cells, causing significant suppression of cell growth. Further analysis revealed that the impact was due to estrogen's stimulating the production of nitric oxide (NO), which could be abrogated when blocking NO formation. Moreover, the effect of estrogen can be resembled by two exogenous nitric oxide donors (SNAP and GSNO). Using ER-negative cell line MDA-MB231, estrogen could not enhance the cell killing of PARP inhibitors any more, but addition of NO donors re-established the enhancing effects. The increased NO level led to accumulation of DNA double strand breaks (DSBs) based on the formation of H2AX foci. Consistent with earlier studies, we demonstrated that NO suppressed the expression of BRCA1, a key player involved in DSB recombination repair. Taken together, these data reveal an important role of estrogen on the treatment of PARP inhibitors, which may affect its clinical treatment and should be considered in precision therapies for ER-positive and negative cancers.
Collapse
Affiliation(s)
- Sa Zhou
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, PR China
| | - Yupeng Liu
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, PR China
| | - Lijun Jin
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, PR China
| | - Peiyan Guo
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, PR China
| | - Qingxi Liu
- Qilu Institute of Technology, Shandong, 250200, PR China
| | - Jingbo Shan
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, PR China
| | - Xuegang Luo
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, PR China
| | - Hongpeng He
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, PR China
| | - Wenjian Ma
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, PR China; Qilu Institute of Technology, Shandong, 250200, PR China.
| | - TongCun Zhang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, PR China; Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, 430081, PR China.
| |
Collapse
|
31
|
Jafari A, Babajani A, Abdollahpour-Alitappeh M, Ahmadi N, Rezaei-Tavirani M. Exosomes and cancer: from molecular mechanisms to clinical applications. Med Oncol 2021; 38:45. [PMID: 33743101 DOI: 10.1007/s12032-021-01491-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/04/2021] [Indexed: 12/15/2022]
Abstract
Exosomes are extracellular nanovesicles secreted from almost all types of normal and cancer cells. Collective evidence suggests that exosomes participate in cell-cell communication via transmitting their cargo, including nucleic acids, proteins, and metabolites to recipient cells. Tumor-derived exosomes (TEXs) play prominent roles in the regulation of molecular pathways in malignancies. Internalization of exosomes by tumor cells affects cellular pathways and several cancer hallmarks, including reprogramming of stromal cells, modulating immune responses, reconstructing extracellular matrix architecture, or even endowing tumor cells with drug features resistance. The unique biogenesis pathways of exosomes, their composition, low immunogenicity, and nontoxicity, together with their ability to target tumor cells, bring them up as an attractive vesicles for cancer therapy. Thus, understanding the molecular mechanisms of exosomes' participation in tumorigenesis will be critical for the next generation of cancer therapeutics. This review aims to summarize the exosomes' roles in different mechanisms underlying cancer progression for the rational design of tailored strategies against this illness. The present study also highlights the new findings on using these smart vesicles as therapeutic targets and potential biomarkers. Recent advances in exosome biology will open up new, more effective, less invasive, and more individualized clinical applications for treating cancer patients.
Collapse
Affiliation(s)
- Ameneh Jafari
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirhesam Babajani
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Nayebali Ahmadi
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Proteomics Research Center, Department of Medical Lab Technology, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Rezaei-Tavirani
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
32
|
Dou J, Li L, Guo M, Mei F, Zheng D, Xu H, Xue R, Bao X, Zhao F, Zhang Y. Iron Oxide Nanoparticles Combined with Cytosine Arabinoside Show Anti-Leukemia Stem Cell Effects on Acute Myeloid Leukemia by Regulating Reactive Oxygen Species. Int J Nanomedicine 2021; 16:1231-1244. [PMID: 33633448 PMCID: PMC7900778 DOI: 10.2147/ijn.s278885] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 12/15/2020] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND AND AIM Acute myeloid leukemia (AML), initiated and maintained by leukemia stem cells (LSCs), is often relapsed or refractory to therapy. The present study aimed at assessing the effects of nanozyme-like Fe3O4 nanoparticles (IONPs) combined with cytosine arabinoside (Ara-C) on LSCs in vitro and in vivo. METHODS The CD34+CD38-LSCs, isolated from human AML cell line KG1a by a magnetic activated cell sorting method, were treated with Ara-C, IONPs, and Ara-C+ IONPs respectively in vitro. The cellular proliferation, apoptosis, reactive oxygen species (ROS), and the related molecular expression levels in LSCs were analyzed using flow cytometry, RT-qPCR, and Western blot. The nonobese diabetic/severe combined immune deficiency mice were transplanted with LSCs or non-LSCs via tail vein, and then the mice were treated with Ara-C, IONPs and IONPs plus Ara-C, respectively. The therapeutic effects on the AML bearing mice were further evaluated. RESULTS LSCs indicated stronger cellular proliferation, more clone formation, and more robust resistance to Ara-C than non-LSCs. Compared with LSCs treated with Ara-C alone, LSCs treated with IONPs plus Ara-C showed a significant increase in apoptosis and ROS levels that might be regulated by nanozyme-like IONPs via improving the expression of pro-oxidation molecule gp91-phox but decreasing the expression of antioxidation molecule superoxide dismutase 1. The in vivo results suggested that, compared with the AML bearing mice treated with Ara-C alone, the mice treated with IONPs plus Ara-C markedly reduced the abnormal leukocyte numbers in peripheral blood and bone marrow and significantly extended the survival of AML bearing mice. CONCLUSION IONPs combined with Ara-C showed the effectiveness on reducing AML burden in the mice engrafted with LSCs and extending mouse survival by increasing LSC's ROS level to induce LSC apoptosis. Our findings suggest that targeting LSCs could control the AML relapse by using IONPs plus Ara-C.
Collapse
Affiliation(s)
- Jun Dou
- Department of Pathogenic Biology and Immunology, Medical College, Southeast University, Nanjing, 210009, People’s Republic of China
| | - Luoyang Li
- Department of Pathogenic Biology and Immunology, Medical College, Southeast University, Nanjing, 210009, People’s Republic of China
| | - Mei Guo
- Department of Pathogenic Biology and Immunology, Medical College, Southeast University, Nanjing, 210009, People’s Republic of China
| | - Feng Mei
- Department of Pathogenic Biology and Immunology, Medical College, Southeast University, Nanjing, 210009, People’s Republic of China
| | - Danfeng Zheng
- Department of Pathogenic Biology and Immunology, Medical College, Southeast University, Nanjing, 210009, People’s Republic of China
| | - Hui Xu
- Department of Pathogenic Biology and Immunology, Medical College, Southeast University, Nanjing, 210009, People’s Republic of China
| | - Rui Xue
- Department of Pathogenic Biology and Immunology, Medical College, Southeast University, Nanjing, 210009, People’s Republic of China
| | - Xueyang Bao
- Department of Pathogenic Biology and Immunology, Medical College, Southeast University, Nanjing, 210009, People’s Republic of China
| | - Fengshu Zhao
- Department of Pathogenic Biology and Immunology, Medical College, Southeast University, Nanjing, 210009, People’s Republic of China
| | - Yu Zhang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210009, People’s Republic of China
| |
Collapse
|
33
|
Jiao D, Zhang J, Chen P, Guo X, Qiao J, Zhu J, Wang L, Lu Z, Liu Z. HN1L promotes migration and invasion of breast cancer by up-regulating the expression of HMGB1. J Cell Mol Med 2021; 25:397-410. [PMID: 33191617 PMCID: PMC7810958 DOI: 10.1111/jcmm.16090] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 09/01/2020] [Accepted: 10/11/2020] [Indexed: 02/06/2023] Open
Abstract
Recent reports showed that haematological and neurological expressed 1-like (HN1L) gene participated in tumorigenesis and tumour invasion. However, the expression and role of HN1L in breast cancer remain to be investigated. Here, bioinformatics, western blot and immunohistochemistry were used to detect the expression of HN1L in breast cancer. Wound healing, transwell assay, immunofluorescence assay and mass spectrum were used to explore the role and mechanism of HN1L on the migration and invasion of breast cancer, which was confirmed in vivo using a nude mice model. Results showed that HN1L was significantly over-expressed in breast cancer tissues, which was positively correlated with M metastasis of breast cancer patients. Silencing HN1L significantly inhibited the invasion and metastasis of breast cancer cells in vitro and lung metastasis in nude mice metastasis model of breast cancer. Mechanistically, HN1L interacted with HSPA9 and affected the expression of HMGB1, playing a key role in promoting the invasion and metastasis of breast cancer cell. These results suggested that HN1L was an appealing drug target for breast cancer.
Collapse
Affiliation(s)
- Dechuang Jiao
- Department of Breast DiseaseHenan Breast Cancer CenterAffiliated Cancer Hospital of Zhengzhou University & Henan Cancer HospitalZhengzhouChina
| | - Jingyang Zhang
- Department of Breast DiseaseHenan Breast Cancer CenterAffiliated Cancer Hospital of Zhengzhou University & Henan Cancer HospitalZhengzhouChina
| | - Ping Chen
- College of Basic Medical SciencesCollaborative Innovation Center of Henan Province for Cancer ChemopreventionZhengzhou UniversityZhengzhouChina
| | - Xuhui Guo
- Department of Breast DiseaseHenan Breast Cancer CenterAffiliated Cancer Hospital of Zhengzhou University & Henan Cancer HospitalZhengzhouChina
| | - Jianghua Qiao
- Department of Breast DiseaseHenan Breast Cancer CenterAffiliated Cancer Hospital of Zhengzhou University & Henan Cancer HospitalZhengzhouChina
| | - Jiujun Zhu
- Department of Breast DiseaseHenan Breast Cancer CenterAffiliated Cancer Hospital of Zhengzhou University & Henan Cancer HospitalZhengzhouChina
| | - Lina Wang
- Department of Breast DiseaseHenan Breast Cancer CenterAffiliated Cancer Hospital of Zhengzhou University & Henan Cancer HospitalZhengzhouChina
| | - Zhenduo Lu
- Department of Breast DiseaseHenan Breast Cancer CenterAffiliated Cancer Hospital of Zhengzhou University & Henan Cancer HospitalZhengzhouChina
| | - Zhenzhen Liu
- Department of Breast DiseaseHenan Breast Cancer CenterAffiliated Cancer Hospital of Zhengzhou University & Henan Cancer HospitalZhengzhouChina
| |
Collapse
|
34
|
Baek M, Chang JT, Echeverria GV. Methodological Advancements for Investigating Intra-tumoral Heterogeneity in Breast Cancer at the Bench and Bedside. J Mammary Gland Biol Neoplasia 2020; 25:289-304. [PMID: 33300087 PMCID: PMC7960623 DOI: 10.1007/s10911-020-09470-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 11/12/2020] [Indexed: 12/20/2022] Open
Abstract
There is a major need to overcome therapeutic resistance and metastasis that eventually arises in many breast cancer patients. Therapy resistant and metastatic tumors are increasingly recognized to possess intra-tumoral heterogeneity (ITH), a diversity of cells within an individual tumor. First hypothesized in the 1970s, the possibility that this complex ITH may endow tumors with adaptability and evolvability to metastasize and evade therapies is now supported by multiple lines of evidence. Our understanding of ITH has been driven by recent methodological advances including next-generation sequencing, computational modeling, lineage tracing, single-cell technologies, and multiplexed in situ approaches. These have been applied across a range of specimens, including patient tumor biopsies, liquid biopsies, cultured cell lines, and mouse models. In this review, we discuss these approaches and how they have deepened our understanding of the mechanistic origins of ITH amongst tumor cells, including stem cell-like differentiation hierarchies and Darwinian evolution, and the functional role for ITH in breast cancer progression. While ITH presents a challenge for combating tumor evolution, in-depth analyses of ITH in clinical biopsies and laboratory models hold promise to elucidate therapeutic strategies that should ultimately improve outcomes for breast cancer patients.
Collapse
Affiliation(s)
- Mokryun Baek
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jeffrey T Chang
- Department of Pharmacology and Integrative Biology, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Gloria V Echeverria
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
35
|
Jindal S, Pennock ND, Klug A, Narasimhan J, Calhoun A, Roberts MR, Tamimi RM, Eliassen AH, Weinmann S, Borges VF, Schedin P. S-nitrosylated and non-nitrosylated COX2 have differential expression and distinct subcellular localization in normal and breast cancer tissue. NPJ Breast Cancer 2020; 6:62. [PMID: 33298921 PMCID: PMC7686348 DOI: 10.1038/s41523-020-00204-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 10/21/2020] [Indexed: 02/07/2023] Open
Abstract
Immunohistochemical (IHC) staining in breast cancer shows both gain and loss of COX2 expression with disease risk and progression. We investigated four common COX2 antibody clones and found high specificity for purified human COX2 for three clones; however, recognition of COX2 in cell lysates was clone dependent. Biochemical characterization revealed two distinct forms of COX2, with SP21 recognizing an S-nitrosylated form, and CX229 and CX294 recognizing non-nitrosylated COX2 antigen. We found S-nitrosylated and non-nitrosylated COX2 occupy different subcellular locations in normal and breast cancer tissue, implicating distinct synthetic/trafficking pathways and function. Dual stains of ~2000 breast cancer cases show early-onset breast cancer had increased expression of both forms of COX2 compared to postmenopausal cases. Our results highlight the strengths of using multiple, highly characterized antibody clones for COX2 IHC studies and raise the prospect that S-nitrosylation of COX2 may play a role in breast cancer biology.
Collapse
Affiliation(s)
- Sonali Jindal
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, 2720 SW Moody Ave., Mailing Code: KR-CDCB, Portland, OR, 97201, USA
- Knight Cancer Institute, Oregon Health & Science University, 2720 SW Moody Ave., Mailing Code: KR-ADM, Portland, OR, 97201, USA
| | - Nathan D Pennock
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, 2720 SW Moody Ave., Mailing Code: KR-CDCB, Portland, OR, 97201, USA
| | - Alex Klug
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, 2720 SW Moody Ave., Mailing Code: KR-CDCB, Portland, OR, 97201, USA
| | - Jayasri Narasimhan
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, 2720 SW Moody Ave., Mailing Code: KR-CDCB, Portland, OR, 97201, USA
| | - Andrea Calhoun
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, 2720 SW Moody Ave., Mailing Code: KR-CDCB, Portland, OR, 97201, USA
| | - Michelle R Roberts
- Department of Dermatology, Massachusetts General Hospital, 50 Staniford Street, Suite 200, Boston, MA, 02114, USA
| | - Rulla M Tamimi
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, 02114, USA
| | - A Heather Eliassen
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, 02114, USA
| | - Sheila Weinmann
- Center for Health Research, Kaiser Permanente Northwest, 3800 N. Interstate Ave., Portland, OR, 97227, USA
| | - Virginia F Borges
- School of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, MS8117, RC-1S, 8401K, 12801 E 17th Ave., Aurora, CO, 80045, USA
- Young Women's Breast Cancer Translational Program, School of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, MS8117, RC-1S, 8401K, 12801 E 17th Ave., Aurora, CO, 80045, USA
| | - Pepper Schedin
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, 2720 SW Moody Ave., Mailing Code: KR-CDCB, Portland, OR, 97201, USA.
- Knight Cancer Institute, Oregon Health & Science University, 2720 SW Moody Ave., Mailing Code: KR-ADM, Portland, OR, 97201, USA.
- School of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, MS8117, RC-1S, 8401K, 12801 E 17th Ave., Aurora, CO, 80045, USA.
- Young Women's Breast Cancer Translational Program, School of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, MS8117, RC-1S, 8401K, 12801 E 17th Ave., Aurora, CO, 80045, USA.
| |
Collapse
|
36
|
Ávalos-Moreno M, López-Tejada A, Blaya-Cánovas JL, Cara-Lupiañez FE, González-González A, Lorente JA, Sánchez-Rovira P, Granados-Principal S. Drug Repurposing for Triple-Negative Breast Cancer. J Pers Med 2020; 10:E200. [PMID: 33138097 PMCID: PMC7711505 DOI: 10.3390/jpm10040200] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/20/2020] [Accepted: 10/28/2020] [Indexed: 12/12/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive type of breast cancer which presents a high rate of relapse, metastasis, and mortality. Nowadays, the absence of approved specific targeted therapies to eradicate TNBC remains one of the main challenges in clinical practice. Drug discovery is a long and costly process that can be dramatically improved by drug repurposing, which identifies new uses for existing drugs, both approved and investigational. Drug repositioning benefits from improvements in computational methods related to chemoinformatics, genomics, and systems biology. To the best of our knowledge, we propose a novel and inclusive classification of those approaches whereby drug repurposing can be achieved in silico: structure-based, transcriptional signatures-based, biological networks-based, and data-mining-based drug repositioning. This review specially emphasizes the most relevant research, both at preclinical and clinical settings, aimed at repurposing pre-existing drugs to treat TNBC on the basis of molecular mechanisms and signaling pathways such as androgen receptor, adrenergic receptor, STAT3, nitric oxide synthase, or AXL. Finally, because of the ability and relevance of cancer stem cells (CSCs) to drive tumor aggressiveness and poor clinical outcome, we also focus on those molecules repurposed to specifically target this cell population to tackle recurrence and metastases associated with the progression of TNBC.
Collapse
Affiliation(s)
- Marta Ávalos-Moreno
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración, 18016 Granada, Spain; (M.Á.-M.); (A.L.-T.); (J.L.B.-C.); (F.E.C.-L.); (A.G.-G.); (J.A.L.)
| | - Araceli López-Tejada
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración, 18016 Granada, Spain; (M.Á.-M.); (A.L.-T.); (J.L.B.-C.); (F.E.C.-L.); (A.G.-G.); (J.A.L.)
- UGC de Oncología Médica, Complejo Hospitalario de Jaén, 23007 Jaén, Spain;
| | - Jose L. Blaya-Cánovas
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración, 18016 Granada, Spain; (M.Á.-M.); (A.L.-T.); (J.L.B.-C.); (F.E.C.-L.); (A.G.-G.); (J.A.L.)
- UGC de Oncología Médica, Complejo Hospitalario de Jaén, 23007 Jaén, Spain;
| | - Francisca E. Cara-Lupiañez
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración, 18016 Granada, Spain; (M.Á.-M.); (A.L.-T.); (J.L.B.-C.); (F.E.C.-L.); (A.G.-G.); (J.A.L.)
- UGC de Oncología Médica, Complejo Hospitalario de Jaén, 23007 Jaén, Spain;
| | - Adrián González-González
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración, 18016 Granada, Spain; (M.Á.-M.); (A.L.-T.); (J.L.B.-C.); (F.E.C.-L.); (A.G.-G.); (J.A.L.)
- UGC de Oncología Médica, Complejo Hospitalario de Jaén, 23007 Jaén, Spain;
| | - Jose A. Lorente
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración, 18016 Granada, Spain; (M.Á.-M.); (A.L.-T.); (J.L.B.-C.); (F.E.C.-L.); (A.G.-G.); (J.A.L.)
- Department of Legal Medicine, School of Medicine—PTS—University of Granada, 18016 Granada, Spain
| | | | - Sergio Granados-Principal
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración, 18016 Granada, Spain; (M.Á.-M.); (A.L.-T.); (J.L.B.-C.); (F.E.C.-L.); (A.G.-G.); (J.A.L.)
- UGC de Oncología Médica, Complejo Hospitalario de Jaén, 23007 Jaén, Spain;
| |
Collapse
|
37
|
Gene Expression Signature of BRAF Inhibitor Resistant Melanoma Spheroids. Pathol Oncol Res 2020; 26:2557-2566. [PMID: 32613561 PMCID: PMC7471197 DOI: 10.1007/s12253-020-00837-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/08/2020] [Indexed: 02/06/2023]
Abstract
In vitro cell cultures are frequently used to define the molecular background of drug resistance. The majority of currently available data have been obtained from 2D in vitro cultures, however, 3D cell culture systems (spheroids) are more likely to behave similarly to in vivo conditions. Our major aim was to compare the gene expression signature of 2D and 3D cultured BRAFV600E mutant melanoma cell lines. We successfully developed BRAF-drug resistant cell lines from paired primary/metastatic melanoma cell lines in both 2D and 3D in vitro cultures. Using Affymetrix Human Gene 1.0 ST arrays, we determined the gene expression pattern of all cell lines. Our analysis revealed 1049 genes (562 upregulated and 487 downregulated) that were differentially expressed between drug-sensitive cells grown under different cell cultures. Pathway analysis showed that the differently expressed genes were mainly associated with the cell cycle, p53, and other cancer-related pathways. The number of upregulated genes (72 genes) was remarkably fewer when comparing the resistant adherent cells to cells that grow in 3D, and were associated with cell adhesion molecules and IGF1R signalling. Only 1% of the upregulated and 5.6% of the downregulated genes were commonly altered between the sensitive and the resistant spheroids. Interestingly, we found several genes (BNIP3, RING1 and ABHD4) with inverse expression signature between sensitive and resistant spheroids, which are involved in anoikis resistance and cell cycle regulation. In summary, our study highlights gene expression alterations that might help to understand the development of acquired resistance in melanoma cells in tumour tissue.
Collapse
|
38
|
Huang D, Liu C, Sun X, Sun X, Qu Y, Tang Y, Li G, Tong T. CRL4 DCAF8 and USP11 oppositely regulate the stability of myeloid leukemia factors (MLFs). Biochem Biophys Res Commun 2020; 529:127-132. [PMID: 32703400 DOI: 10.1016/j.bbrc.2020.05.186] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 05/25/2020] [Indexed: 01/20/2023]
Abstract
Myeloid leukemia factors (MLF1 and MLF2) are proteins associated with leukemia and several other cancers. However, little is known about the regulatory mechanisms underlying the stability of these proteins. Here, we show that DDB1 and CUL4 associated factor 8 (DCAF8), which can form a functional E3 ligase complex (CRL4DCAF8), has a strong interaction with the MLF2 protein. DCAF8 could promote MLF2 degradation through the ubiquitin-proteasome pathway. In contrast, ubiquitin specific peptidase 11 (USP11) associates with MLF2, thereby increasing its stability. Since MLF1 is highly related to MLF2, we demonstrated that MLF1 also interacts with DCAF8 and USP11, suggesting that CRL4DCAF8 and USP11 may also regulate the expression of MLF1. TCGA analysis revealed that both the myeloid leukemia factors (MLF1 and MLF2) show significant differential expression in various tumors. The results of our study indicate that CRL4DCAF8 and USP11 play opposite roles in the regulation of MLF1 and MLF2, which may, in turn, affect their biological functions in various cancers.
Collapse
Affiliation(s)
- Daoyuan Huang
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, 100191, China
| | - Cheng Liu
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, 100191, China
| | - Xiwen Sun
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, 100191, China
| | - Xinpei Sun
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, 100191, China
| | - Yanan Qu
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, 100191, China
| | - Yunyi Tang
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, 100191, China
| | - Guodong Li
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, 100191, China
| | - Tanjun Tong
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, 100191, China.
| |
Collapse
|
39
|
Dai J, Huang M, Amos CI, Hung RJ, Tardon A, Andrew A, Chen C, Christiani DC, Albanes D, Rennert G, Fan J, Goodman G, Liu G, Field JK, Grankvist K, Kiemeney LA, Le Marchand L, Schabath MB, Johansson M, Aldrich MC, Johansson M, Caporaso N, Lazarus P, Lam S, Bojesen SE, Arnold S, Landi MT, Risch A, Wichmann HE, Bickeboller H, Brennan P, Shete S, Melander O, Brunnstrom H, Zienolddiny S, Woll P, Stevens V, Hu Z, Shen H. Genome-wide association study of INDELs identified four novel susceptibility loci associated with lung cancer risk. Int J Cancer 2020; 146:2855-2864. [PMID: 31577861 PMCID: PMC7101262 DOI: 10.1002/ijc.32698] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 07/23/2019] [Accepted: 08/02/2019] [Indexed: 12/24/2022]
Abstract
Genome-wide association studies (GWAS) have identified 45 susceptibility loci associated with lung cancer. Only less than SNPs, small insertions and deletions (INDELs) are the second most abundant genetic polymorphisms in the human genome. INDELs are highly associated with multiple human diseases, including lung cancer. However, limited studies with large-scale samples have been available to systematically evaluate the effects of INDELs on lung cancer risk. Here, we performed a large-scale meta-analysis to evaluate INDELs and their risk for lung cancer in 23,202 cases and 19,048 controls. Functional annotations were performed to further explore the potential function of lung cancer risk INDELs. Conditional analysis was used to clarify the relationship between INDELs and SNPs. Four new risk loci were identified in genome-wide INDEL analysis (1p13.2: rs5777156, Insertion, OR = 0.92, p = 9.10 × 10-8 ; 4q28.2: rs58404727, Deletion, OR = 1.19, p = 5.25 × 10-7 ; 12p13.31: rs71450133, Deletion, OR = 1.09, p = 8.83 × 10-7 ; and 14q22.3: rs34057993, Deletion, OR = 0.90, p = 7.64 × 10-8 ). The eQTL analysis and functional annotation suggested that INDELs might affect lung cancer susceptibility by regulating the expression of target genes. After conducting conditional analysis on potential causal SNPs, the INDELs in the new loci were still nominally significant. Our findings indicate that INDELs could be potentially functional genetic variants for lung cancer risk. Further functional experiments are needed to better understand INDEL mechanisms in carcinogenesis.
Collapse
Affiliation(s)
- Juncheng Dai
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, China
| | - Mingtao Huang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Christopher I Amos
- Department of Medicine, Epidemiology Section, Institute for Clinical and Translational Research, Baylor Medical College, Houston, TX
| | - Rayjean J Hung
- Epidemiology Division, Lunenfeld-Tanenbuaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Adonina Tardon
- Faculty of Medicine, University of Oviedo and CIBERESP, Oviedo, Spain
| | - Angeline Andrew
- Department of Neurology, Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Chu Chen
- Department of Epidemiology, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - David C Christiani
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| | - Gadi Rennert
- Technion Faculty of Medicine, Carmel Medical Center, Israel Institute of Technology, Haifa, Israel
| | - Jingyi Fan
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Gary Goodman
- Public Health Sciences Division, Swedish Cancer Institute, Seattle, WA
| | - Geoffrey Liu
- Epidemiology Division, Princess Margaret Cancer Center, Toronto, ON, Canada
| | - John K Field
- Roy Castle Lung Cancer Research Programme, Department of Molecular & Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Kjell Grankvist
- Unit of Clinical Chemistry, Department of Medical Biosciences, Umeå University, Umeå, Sweden
| | - Lambertus A Kiemeney
- Department of Health Evidence, Radboud University Medical Center, Nijmegen, Germany
| | - Loic Le Marchand
- Department of Epidemiology, University of Hawaii Cancer Center, Honolulu, HI
| | - Matthew B Schabath
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Mattias Johansson
- Genetic Epidemiology Group, International Agency for Research on Cancer, Lyon, France
| | - Melinda C Aldrich
- Department of Thoracic Surgery, Division of Epidemiology, Vanderbilt University Medical Center, Nashville, TN
| | | | - Neil Caporaso
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| | - Philip Lazarus
- Washington State University College of Pharmacy, Spokane, WA
| | - Stephan Lam
- Department of Integrative Oncology, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Stig E Bojesen
- Department of Clinical Biochemistry, Copenhagen University Hospital, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Susanne Arnold
- Markey Cancer Center, University of Kentucky, Lexington, KY
| | - Maria Teresa Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| | - Angela Risch
- Cancer Center Cluster Salzburg at PLUS, Department of Molecular Biology, University of Salzburg, Heidelberg, Austria
| | - H-Erich Wichmann
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Epidemiology, Ludwig Maximilians University, Munich, Bavaria, Germany
| | - Heike Bickeboller
- Department of Genetic Epidemiology, University Medical Center Goettingen, Goettingen, Germany
| | - Paul Brennan
- Genetic Epidemiology Group, International Agency for Research on Cancer, Lyon, France
| | - Sanjay Shete
- Department of Epidemiology, University of Texas, MD Anderson Cancer Center, Houston, TX
| | - Olle Melander
- Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Hans Brunnstrom
- Department of Clinical Sciences, Lund University, Lund, Sweden
| | | | - Penella Woll
- Academic Unit of Clinical Oncology, University of Sheffield, Sheffield, UK
| | - Victoria Stevens
- Epidemiology Research Program, American Cancer Society, Atlanta, GA
| | - Zhibin Hu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, China
| | - Hongbing Shen
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
40
|
Reddy TP, Choi DS, Anselme AC, Qian W, Chen W, Lantto J, Horak ID, Kragh M, Chang JC, Rosato RR. Simultaneous targeting of HER family pro-survival signaling with Pan-HER antibody mixture is highly effective in TNBC: a preclinical trial with PDXs. Breast Cancer Res 2020; 22:48. [PMID: 32414394 PMCID: PMC7227035 DOI: 10.1186/s13058-020-01280-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 04/15/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The human epidermal growth factor receptor (HER) family, notably EGFR, is overexpressed in most triple-negative breast cancer (TNBC) cases and provides cancer cells with compensatory signals that greatly contribute to the survival and development of resistance in response to therapy. This study investigated the effects of Pan-HER (Symphogen, Ballerup, Denmark), a novel mixture of six monoclonal antibodies directed against members of the HER family EGFR, HER2, and HER3, in a preclinical trial of TNBC patient-derived xenografts (PDXs). METHODS Fifteen low passage TNBC PDX tumor samples were transferred into the right mammary fat pad of mice for engraftment. When tumors reached an average size of 100-200 mm3, mice were randomized (n ≥ 6 per group) and treated following three 1-week cycles consisting of three times/week intraperitoneal (IP) injection of either formulation buffer (vehicle control) or Pan-HER (50 mg/kg). At the end of treatment, tumors were collected for Western blot, RNA, and immunohistochemistry analyses. RESULTS All 15 TNBC PDXs were responsive to Pan-HER treatment, showing significant reductions in tumor growth consistent with Pan-HER-mediated tumor downmodulation of EGFR and HER3 protein levels and significantly decreased activation of associated HER family signaling pathways AKT and ERK. Tumor regression was observed in five of the models, which corresponded to those PDX tumor models with the highest level of HER family activation. CONCLUSIONS The marked effect of Pan-HER in numerous HER family-dependent TNBC PDX models justifies further studies of Pan-HER in TNBC clinical trials as a potential therapeutic option.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- Cell Proliferation/drug effects
- Disease Models, Animal
- Drug Resistance, Neoplasm
- ErbB Receptors/antagonists & inhibitors
- ErbB Receptors/genetics
- ErbB Receptors/metabolism
- Female
- Humans
- Mice
- Molecular Targeted Therapy
- Mutation
- Receptor, ErbB-2/antagonists & inhibitors
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Receptor, ErbB-3/antagonists & inhibitors
- Receptor, ErbB-3/genetics
- Receptor, ErbB-3/metabolism
- Triple Negative Breast Neoplasms/drug therapy
- Triple Negative Breast Neoplasms/genetics
- Triple Negative Breast Neoplasms/metabolism
- Triple Negative Breast Neoplasms/pathology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Tejaswini P Reddy
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA
- Texas A&M Health Science Center College of Medicine, Bryan, TX, 77807, USA
| | - Dong S Choi
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Ann C Anselme
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA
- Texas A&M Health Science Center College of Medicine, Bryan, TX, 77807, USA
| | - Wei Qian
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Wen Chen
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Johan Lantto
- Symphogen A/S, Pederstrupvej 93, DK-2750, Ballerup, Denmark
| | - Ivan D Horak
- Symphogen A/S, Pederstrupvej 93, DK-2750, Ballerup, Denmark
| | - Michael Kragh
- Texas A&M Health Science Center College of Medicine, Bryan, TX, 77807, USA
| | - Jenny C Chang
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Roberto R Rosato
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA.
| |
Collapse
|
41
|
Deng Y, Wan H, Tian J, Cheng X, Rao M, Li J, Zhang H, Zhang M, Cai Y, Lu Z, Li Y, Niu S, Shen N, Chang J, Fang Z, Zhong R. CpG-methylation-based risk score predicts progression in colorectal cancer. Epigenomics 2020; 12:605-615. [DOI: 10.2217/epi-2019-0300] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aim: To identify patients with colorectal cancer (CRC) who are at a truly higher risk of progression, which is key for individualized approaches to precision therapy. Materials & methods: We developed a predictor associated with progression-free interval (PFI) using The Cancer Genome Atlas CRC methylation data. Results: The risk score was associated with PFI in the whole cohort (p < 0.001). A nomogram consisting of the risk score and other significant clinical features was generated to predict the 3- and 5-year PFI in the whole set (area under the curve: 0.79 and 0.71, respectively). Conclusion: The risk score based on 23 DNA-methylation sites may serve as the basis for improved prediction of progression in patients with CRC in future clinical practice.
Collapse
Affiliation(s)
- Yao Deng
- Department of Epidemiology & Biostatistics & Ministry of Education Key Lab of Environment & Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei 430030, PR China
| | - Hao Wan
- Department of Epidemiology & Biostatistics & Ministry of Education Key Lab of Environment & Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei 430030, PR China
| | - Jianbo Tian
- Department of Epidemiology & Biostatistics & Ministry of Education Key Lab of Environment & Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei 430030, PR China
| | - Xiang Cheng
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, PR China
| | - Meilin Rao
- Department of Epidemiology & Biostatistics & Ministry of Education Key Lab of Environment & Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei 430030, PR China
| | - Jiaoyuan Li
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, PR China
| | - Hongli Zhang
- Department of Epidemiology & Biostatistics & Ministry of Education Key Lab of Environment & Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei 430030, PR China
| | - Ming Zhang
- Department of Epidemiology & Biostatistics & Ministry of Education Key Lab of Environment & Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei 430030, PR China
| | - Yimin Cai
- Department of Epidemiology & Biostatistics & Ministry of Education Key Lab of Environment & Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei 430030, PR China
| | - Zequn Lu
- Department of Epidemiology & Biostatistics & Ministry of Education Key Lab of Environment & Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei 430030, PR China
| | - Yue Li
- Department of Epidemiology & Biostatistics & Ministry of Education Key Lab of Environment & Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei 430030, PR China
| | - Siyuan Niu
- Department of Epidemiology & Biostatistics & Ministry of Education Key Lab of Environment & Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei 430030, PR China
| | - Na Shen
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, PR China
| | - Jiang Chang
- Department of Epidemiology & Biostatistics & Ministry of Education Key Lab of Environment & Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei 430030, PR China
| | - Zemin Fang
- Division of Cardiothoracic & Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, PR China
| | - Rong Zhong
- Department of Epidemiology & Biostatistics & Ministry of Education Key Lab of Environment & Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei 430030, PR China
| |
Collapse
|
42
|
Xu Y, Ruggero D. The Role of Translation Control in Tumorigenesis and Its Therapeutic Implications. ANNUAL REVIEW OF CANCER BIOLOGY-SERIES 2020. [DOI: 10.1146/annurev-cancerbio-030419-033420] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
As a convergent mechanism downstream of most oncogenic signals, control of mRNA translation has emerged as a key driver in establishing and tuning gene expression at specific steps in cancer development. Translation control is the most energetically expensive molecular process in the cell that needs to be modulated upon adaption to limited cellular resources, such as cellular stress. It thereby serves as the Achilles’ heel for cancer cells, particularly in response to changes in the microenvironment as well as to nutrient and metabolic shifts characteristic of cancer cell growth and metastasis. In this review, we discuss emerging discoveries that reveal how cancer cells modulate the translation machinery to adapt to oncogenic stress, the mechanisms that guide mRNA translation specificity in cancer, and how this selective mode of gene regulation provides advantages for cancer progression. We also provide an overview of promising preclinical and clinical efforts aimed at targeting the unique vulnerabilities of cancer cells that rely on the remodeling of mRNA translation for their infinite growth and survival.
Collapse
Affiliation(s)
- Yichen Xu
- Department of Urology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California 94157, USA
| | - Davide Ruggero
- Department of Urology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California 94157, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California 94158, USA
| |
Collapse
|
43
|
Novel Mutation Hotspots within Non-Coding Regulatory Regions of the Chronic Lymphocytic Leukemia Genome. Sci Rep 2020; 10:2407. [PMID: 32051441 PMCID: PMC7015923 DOI: 10.1038/s41598-020-59243-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 01/27/2020] [Indexed: 01/17/2023] Open
Abstract
Mutations in non-coding DNA regions are increasingly recognized as cancer drivers. These mutations can modify gene expression in cis or by inducing high-order chormatin structure modifications with long-range effects. Previous analysis reported the detection of recurrent and functional non-coding DNA mutations in the chronic lymphocytic leukemia (CLL) genome, such as those in the 3′ untranslated region of NOTCH1 and in the PAX5 super-enhancer. In this report, we used whole genome sequencing data produced by the International Cancer Genome Consortium in order to analyze regions with previously reported regulatory activity. This approach enabled the identification of numerous recurrently mutated regions that were frequently positioned in the proximity of genes involved in immune and oncogenic pathways. By correlating these mutations with expression of their nearest genes, we detected significant transcriptional changes in genes such as PHF2 and S1PR2. More research is needed to clarify the function of these mutations in CLL, particularly those found in intergenic regions.
Collapse
|
44
|
Gowda R, Robertson BM, Iyer S, Barry J, Dinavahi SS, Robertson GP. The role of exosomes in metastasis and progression of melanoma. Cancer Treat Rev 2020; 85:101975. [PMID: 32050108 DOI: 10.1016/j.ctrv.2020.101975] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 01/16/2020] [Accepted: 01/18/2020] [Indexed: 12/21/2022]
Abstract
The mechanisms of melanoma metastasis have been the subject of extensive research for decades. Improved diagnostic and therapeutic strategies are of increasing importance for the treatment of melanoma due to its high burden of mortality in the advanced stages of the disease. Intercellular communication is a critical event for the progression of cancer. Collective evidence suggests that exosomes, small extracellular membrane vesicles released by the cells, are important facilitators of intercellular communication between the cells and the surrounding environment. Although the emerging field of exosomes is rapidly gaining traction in the scientific community, there is limited knowledge regarding the role of exosomes in melanoma. This review discusses the multifaceted role of melanoma-derived exosomes in promoting the process of metastasis by modulating the invasive and angiogenic capacity of malignant cells. The future implications of exosome research and the therapeutic potential of exosomes are also discussed.
Collapse
Affiliation(s)
- Raghavendra Gowda
- Departments of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Penn State Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Penn State Melanoma Therapeutics Program, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Foreman Foundation for Melanoma Research, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Bailey M Robertson
- Departments of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Soumya Iyer
- Departments of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Penn State Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - John Barry
- Departments of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Saketh S Dinavahi
- Departments of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Penn State Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Gavin P Robertson
- Departments of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Departments of Pathology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Departments of Dermatology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Departments of Surgery, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Penn State Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Penn State Melanoma Therapeutics Program, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Foreman Foundation for Melanoma Research, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States.
| |
Collapse
|
45
|
The role of phosphorylation of MLF2 at serine 24 in BCR-ABL leukemogenesis. Cancer Gene Ther 2019; 27:98-107. [PMID: 31831854 DOI: 10.1038/s41417-019-0152-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 11/19/2019] [Accepted: 11/22/2019] [Indexed: 01/29/2023]
Abstract
Chronic myelogenous leukemia (CML) is a myeloproliferative disorder defined by the presence of the fusion gene BCR-ABL1 in primitive hematopoietic progenitors. The myeloid leukemia factors (MLFs) were identified in the fly and human, and are involved in acute leukemia and enhancing the myeloid factor; however, the function of MLF2 in CML is poorly understood. In this study, we demonstrated that MLF2 may play an oncogenic role in CML. The expression level of MLF2 was related to the proliferation, colony-formation ability, and sensitivity to imatinib in K562 cells. Moreover, phosphorylation at serine 24, detected through Phos-tag sodium dodecyl sulfate-polyacrylamide gel electrophoresis, was required to maintain the activity of MLF2 in CML. The effects of MLF2 overexpression on the colony-formation ability in vitro and mouse survival in vivo could be alleviated by point mutation of MLF2 at serine 24. These findings uncover the oncogenic role of MLF2 through phosphorylation at serine 24 and provide a novel therapeutic target in CML.
Collapse
|
46
|
Li Y, Ren Z, Peng Y, Li K, Wang X, Huang G, Qi S, Liu Y. Classification of glioma based on prognostic alternative splicing. BMC Med Genomics 2019; 12:165. [PMID: 31729991 PMCID: PMC6858651 DOI: 10.1186/s12920-019-0603-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 10/16/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Previously developed classifications of glioma have provided enormous advantages for the diagnosis and treatment of glioma. Although the role of alternative splicing (AS) in cancer, especially in glioma, has been validated, a comprehensive analysis of AS in glioma has not yet been conducted. In this study, we aimed at classifying glioma based on prognostic AS. METHODS Using the TCGA glioblastoma (GBM) and low-grade glioma (LGG) datasets, we analyzed prognostic splicing events. Consensus clustering analysis was conducted to classified glioma samples and correlation analysis was conducted to characterize regulatory network of splicing factors and splicing events. RESULTS We analyzed prognostic splicing events and proposed novel splicing classifications across pan-glioma samples (labeled pST1-7) and across GBM samples (labeled ST1-3). Distinct splicing profiles between GBM and LGG were observed, and the primary discriminator for the pan-glioma splicing classification was tumor grade. Subtype-specific splicing events were identified; one example is AS of zinc finger proteins, which is involved in glioma prognosis. Furthermore, correlation analysis of splicing factors and splicing events identified SNRPB and CELF2 as hub splicing factors that upregulated and downregulated oncogenic AS, respectively. CONCLUSION A comprehensive analysis of AS in glioma was conducted in this study, shedding new light on glioma heterogeneity and providing new insights into glioma diagnosis and treatment.
Collapse
Affiliation(s)
- Yaomin Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Dadao Bei Street 1838#, Guangzhou, People's Republic of China
- Laboratory for Precision Neurosurgery, Nanfang hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zhonglu Ren
- College of Medical Information Engineering, Guangdong Pharmaceutical University, Guangzhou, 510515, Guangdong, China
| | - Yuping Peng
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Dadao Bei Street 1838#, Guangzhou, People's Republic of China
| | - Kaishu Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Dadao Bei Street 1838#, Guangzhou, People's Republic of China
- Laboratory for Precision Neurosurgery, Nanfang hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Xiran Wang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Dadao Bei Street 1838#, Guangzhou, People's Republic of China
- Laboratory for Precision Neurosurgery, Nanfang hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Guanglong Huang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Dadao Bei Street 1838#, Guangzhou, People's Republic of China
- Laboratory for Precision Neurosurgery, Nanfang hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Songtao Qi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Dadao Bei Street 1838#, Guangzhou, People's Republic of China.
- Laboratory for Precision Neurosurgery, Nanfang hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Yawei Liu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Dadao Bei Street 1838#, Guangzhou, People's Republic of China.
- Laboratory for Precision Neurosurgery, Nanfang hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
47
|
CDK2-mediated site-specific phosphorylation of EZH2 drives and maintains triple-negative breast cancer. Nat Commun 2019; 10:5114. [PMID: 31704972 PMCID: PMC6841924 DOI: 10.1038/s41467-019-13105-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 10/15/2019] [Indexed: 12/17/2022] Open
Abstract
Triple-negative breast cancer (TNBC), which lacks estrogen receptor α (ERα), progesterone receptor, and human epidermal growth factor receptor 2 (HER2) expression, is closely related to basal-like breast cancer. Previously, we and others report that cyclin E/cyclin-dependent kinase 2 (CDK2) phosphorylates enhancer of zeste homolog 2 (EZH2) at T416 (pT416-EZH2). Here, we show that transgenic expression of phospho-mimicking EZH2 mutant EZH2T416D in mammary glands leads to tumors with TNBC phenotype. Coexpression of EZH2T416D in mammary epithelia of HER2/Neu transgenic mice reprograms HER2-driven luminal tumors into basal-like tumors. Pharmacological inhibition of CDK2 or EZH2 allows re-expression of ERα and converts TNBC to luminal ERα-positive, rendering TNBC cells targetable by tamoxifen. Furthermore, the combination of either CDK2 or EZH2 inhibitor with tamoxifen effectively suppresses tumor growth and markedly improves the survival of the mice bearing TNBC tumors, suggesting that the mechanism-based combination therapy may be an alternative approach to treat TNBC. EZH2 phosphorylation by CDK2 promotes progression of triple-negative breast cancer (TNBC). Here, the authors show that this signaling axis downregulates ERα, and thus combinatorial blockade of CDK2 and EZH2 sensitizes TNBC cells to tamoxifen.
Collapse
|
48
|
Liu ZB, Ezzedine NE, Eterovic AK, Ensor JE, Huang HJ, Albanell J, Choi DS, Lluch A, Liu Y, Rojo F, Wong H, Martínez-Dueñas E, Guerrero-Zotano Á, Shao ZM, Darcourt JG, Mills GB, Dave B, Chang JC. Detection of breast cancer stem cell gene mutations in circulating free DNA during the evolution of metastases. Breast Cancer Res Treat 2019; 178:251-261. [PMID: 31388936 DOI: 10.1007/s10549-019-05374-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 07/21/2019] [Indexed: 12/18/2022]
Abstract
PURPOSE Limited knowledge exists on the detection of breast cancer stem cell (BCSC)-related mutations in circulating free DNA (cfDNA) from patients with advanced cancers. Identification of new cancer biomarkers may allow for earlier detection of disease progression and treatment strategy modifications. METHODS We conducted a prospective study to determine the feasibility and prognostic utility of droplet digital polymerase chain reaction (ddPCR)-based BCSC gene mutation analysis of cfDNA in patients with breast cancer. RESULTS Detection of quantitative BCSC gene mutation in cfDNA by ddPCR mirrors disease progression and thus may represent a valuable and cost-effective measure of tumor burden. We have previously shown that hematological and neurological expressed 1-like (HN1L), ribosomal protein L39 (RPL39), and myeloid leukemia factor 2 (MLF2) are novel targets for BCSC self-renewal, and targeting these genetic alterations could be useful for personalized genomic-based therapy. CONCLUSION BCSC mutation detection in cfDNA may have important implications for diagnosis, prognosis, and serial monitoring.
Collapse
Affiliation(s)
- Zhe-Bin Liu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 20032, China
- Houston Methodist Research Institute, Houston, TX, 77030, USA
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Nader E Ezzedine
- Department of Systems Biology and Institute of Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Agda K Eterovic
- Department of Systems Biology and Institute of Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Joe E Ensor
- Houston Methodist Cancer Center, 6445 Main St P21-34, Houston, TX, 77030, USA
| | - Helen J Huang
- Division of Cancer Medicine, Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Joan Albanell
- GEICAM (Spanish Breast Cancer Group), San Sebastián de los Reyes, 28703, Madrid, Spain
- Centro de Investigación Biomédica en Red de Oncología, CIBERONC-ISCIII, 28029, Madrid, Spain
- Hospital del Mar, 08003, Barcelona, Spain
| | - Dong S Choi
- Houston Methodist Research Institute, Houston, TX, 77030, USA
- Houston Methodist Cancer Center, 6445 Main St P21-34, Houston, TX, 77030, USA
| | - Ana Lluch
- GEICAM (Spanish Breast Cancer Group), San Sebastián de los Reyes, 28703, Madrid, Spain
- Centro de Investigación Biomédica en Red de Oncología, CIBERONC-ISCIII, 28029, Madrid, Spain
- Hospital Clínico Universitario de Valencia, 46010, Valencia, Spain
| | - Yi Liu
- Houston Methodist Research Institute, Houston, TX, 77030, USA
- Houston Methodist Cancer Center, 6445 Main St P21-34, Houston, TX, 77030, USA
| | - Federico Rojo
- GEICAM (Spanish Breast Cancer Group), San Sebastián de los Reyes, 28703, Madrid, Spain
- Centro de Investigación Biomédica en Red de Oncología, CIBERONC-ISCIII, 28029, Madrid, Spain
- Fundación Jiménez Díaz, 28040, Madrid, Spain
| | - Helen Wong
- Houston Methodist Research Institute, Houston, TX, 77030, USA
- Houston Methodist Cancer Center, 6445 Main St P21-34, Houston, TX, 77030, USA
| | - Eduardo Martínez-Dueñas
- GEICAM (Spanish Breast Cancer Group), San Sebastián de los Reyes, 28703, Madrid, Spain
- Hospital Provincial de Castellón, 12002, Castellón, Spain
| | - Ángel Guerrero-Zotano
- GEICAM (Spanish Breast Cancer Group), San Sebastián de los Reyes, 28703, Madrid, Spain
- Instituto Valenciano de Oncología, 46009, Valencia, Spain
| | - Zhi-Min Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 20032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jorge G Darcourt
- Houston Methodist Cancer Center, 6445 Main St P21-34, Houston, TX, 77030, USA
| | - Gordon B Mills
- Department of Systems Biology and Institute of Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Bhuvanesh Dave
- Houston Methodist Research Institute, Houston, TX, 77030, USA
- Houston Methodist Cancer Center, 6445 Main St P21-34, Houston, TX, 77030, USA
| | - Jenny C Chang
- Houston Methodist Research Institute, Houston, TX, 77030, USA.
- Houston Methodist Cancer Center, 6445 Main St P21-34, Houston, TX, 77030, USA.
| |
Collapse
|
49
|
Li J, Mai J, Hinkle L, Lin D, Zhang J, Liu X, Ramirez MR, Zu Y, Lokesh GL, Volk DE, Shen H. Tracking Biodistribution of Myeloid-Derived Cells in Murine Models of Breast Cancer. Genes (Basel) 2019; 10:genes10040297. [PMID: 31013756 PMCID: PMC6523772 DOI: 10.3390/genes10040297] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 03/29/2019] [Accepted: 04/03/2019] [Indexed: 12/24/2022] Open
Abstract
A growing tumor is constantly secreting inflammatory chemokines and cytokines that induce release of immature myeloid cells, including myeloid-derived suppressor cells (MDSCs) and macrophages, from the bone marrow. These cells not only promote tumor growth, but also prepare distant organs for tumor metastasis. On the other hand, the myeloid-derived cells also have phagocytic potential, and can serve as vehicles for drug delivery. We have previously identified thioaptamers that bind a subset of MDSCs with high affinity and specificity. In the current study, we applied one of the thioaptamers as a probe to track myeloid cell distribution in the bone, liver, spleen and tumor in multiple murine models of breast cancer including the 4T1 syngeneic model and MDA-MB-231 and SUM159 xenograft models. Information generated from this study will facilitate further understanding of tumor growth and metastasis, and predict biodistribution patterns of cell-mediated drug delivery.
Collapse
Affiliation(s)
- Jun Li
- Department of Nanomedicine, Houston Methodist Hospital Research Institute, Houston, TX 77030, USA.
- Xiangya School of Medicine, Central South University, 410008 Changsha, Hunan, China.
| | - Junhua Mai
- Department of Nanomedicine, Houston Methodist Hospital Research Institute, Houston, TX 77030, USA.
| | - Louis Hinkle
- Department of Nanomedicine, Houston Methodist Hospital Research Institute, Houston, TX 77030, USA.
| | - Daniel Lin
- Department of Nanomedicine, Houston Methodist Hospital Research Institute, Houston, TX 77030, USA.
| | - Jingxin Zhang
- Department of Nanomedicine, Houston Methodist Hospital Research Institute, Houston, TX 77030, USA.
- Xiangya School of Medicine, Central South University, 410008 Changsha, Hunan, China.
| | - Xiaoling Liu
- Department of Nanomedicine, Houston Methodist Hospital Research Institute, Houston, TX 77030, USA.
| | - Maricela R Ramirez
- Department of Nanomedicine, Houston Methodist Hospital Research Institute, Houston, TX 77030, USA.
| | - Youli Zu
- Department of Pathology and Genomic Medicine, Houston, TX 77030, USA.
| | - Ganesh L Lokesh
- Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| | - David E Volk
- Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Hospital Research Institute, Houston, TX 77030, USA.
- Cancer Center, Houston Methodist Hospital, Houston, TX 77030, USA.
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
50
|
Current Challenges in Cancer Immunotherapy: Multimodal Approaches to Improve Efficacy and Patient Response Rates. JOURNAL OF ONCOLOGY 2019; 2019:4508794. [PMID: 30941175 PMCID: PMC6420990 DOI: 10.1155/2019/4508794] [Citation(s) in RCA: 159] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 02/12/2019] [Indexed: 12/13/2022]
Abstract
Cancer immunotherapy is a promising innovative treatment for many forms of cancer, particularly melanoma. Although immunotherapy has been shown to be efficacious, patient response rates vary and, more often than not, only a small subset of the patients within a large cohort respond favourably to the treatment. This issue is particularly concerning and becomes a challenge of immunotherapy to improve the effectiveness and patient response rates. Here, we review the specific types of available immunotherapy options, their proposed mechanism(s) of action, and the reasons why the patient response to this treatment is variable. The potential favourable options to improve response rates to immunotherapy will be discussed with an emphasis on adopting a multimodal approach on the novel role that the gut microbiota may play in modulating the efficacy of cancer immunotherapy.
Collapse
|