1
|
Conger KO, Chidley C, Ozgurses ME, Zhao H, Kim Y, Semina SE, Burns P, Rawat V, Lietuvninkas L, Sheldon R, Ben-Sahra I, Frasor J, Sorger PK, DeNicola GM, Coloff JL. ASCT2 is a major contributor to serine uptake in cancer cells. Cell Rep 2024; 43:114552. [PMID: 39068660 PMCID: PMC11406281 DOI: 10.1016/j.celrep.2024.114552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 05/23/2024] [Accepted: 07/12/2024] [Indexed: 07/30/2024] Open
Abstract
The non-essential amino acid serine is a critical nutrient for cancer cells due to its diverse biosynthetic functions. While some tumors can synthesize serine de novo, others are auxotrophic and therefore reliant on serine uptake. Importantly, despite several transporters being known to be capable of transporting serine, the transporters that mediate serine uptake in cancer cells are not known. Here, we characterize the amino acid transporter ASCT2 (SLC1A5) as a major contributor to serine uptake in cancer cells. ASCT2 is well known as a glutamine transporter in cancer, and our work demonstrates that serine and glutamine compete for uptake through ASCT2. We further show that ASCT2-mediated serine uptake is essential for purine nucleotide biosynthesis and that estrogen receptor α (ERα) promotes serine uptake by directly activating SLC1A5 transcription. Collectively, our work defines an additional important role for ASCT2 as a serine transporter in cancer and evaluates ASCT2 as a potential therapeutic target.
Collapse
Affiliation(s)
- Kelly O Conger
- Department of Physiology and Biophysics, University of Illinois Cancer Center, University of Illinois College of Medicine, Chicago, IL, USA
| | - Christopher Chidley
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
| | - Mete Emir Ozgurses
- Department of Physiology and Biophysics, University of Illinois Cancer Center, University of Illinois College of Medicine, Chicago, IL, USA
| | - Huiping Zhao
- Department of Physiology and Biophysics, University of Illinois Cancer Center, University of Illinois College of Medicine, Chicago, IL, USA
| | - Yumi Kim
- Department of Cancer Metabolism and Physiology, H. Lee. Moffitt Cancer Center, Tampa, FL, USA
| | - Svetlana E Semina
- Department of Physiology and Biophysics, University of Illinois Cancer Center, University of Illinois College of Medicine, Chicago, IL, USA
| | - Philippa Burns
- Department of Physiology and Biophysics, University of Illinois Cancer Center, University of Illinois College of Medicine, Chicago, IL, USA
| | - Vipin Rawat
- Department of Physiology and Biophysics, University of Illinois Cancer Center, University of Illinois College of Medicine, Chicago, IL, USA
| | - Lina Lietuvninkas
- Department of Physiology and Biophysics, University of Illinois Cancer Center, University of Illinois College of Medicine, Chicago, IL, USA
| | - Ryan Sheldon
- Metabolic and Nutritional Programming, Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, MI, USA
| | - Issam Ben-Sahra
- Robert H. Lurie Cancer Center, Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, IL, USA
| | - Jonna Frasor
- Department of Physiology and Biophysics, University of Illinois Cancer Center, University of Illinois College of Medicine, Chicago, IL, USA
| | - Peter K Sorger
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA; Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Gina M DeNicola
- Department of Cancer Metabolism and Physiology, H. Lee. Moffitt Cancer Center, Tampa, FL, USA
| | - Jonathan L Coloff
- Department of Physiology and Biophysics, University of Illinois Cancer Center, University of Illinois College of Medicine, Chicago, IL, USA.
| |
Collapse
|
2
|
Thakur C, Qiu Y, Bi Z, Wang Z, Chen F. MDIG in Breast Cancer Progression and Metastasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1465:1-14. [PMID: 39586990 DOI: 10.1007/978-3-031-66686-5_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Breast cancer is the most diagnosed cancer among women worldwide, and metastasis remains the major cause of breast cancer-related mortality and is associated with poor patient outcomes. Among breast cancers, triple-negative breast cancers have the worst prognosis owing to their highly aggressive and metastasizing attributes and hence have limited therapeutic options. Here, we have presented our research on an environmentally regulated gene named mdig and its role in the pathogenesis of breast cancer and metastasis. Through global proteomics, chromatin immunoprecipitation sequencing, and a mouse model of orthotopic xenograft, our studies established mdig as an anti-metastasis modulator in breast cancer with its influence on the methylation of DNA and histone proteins, thereby regulating the expression of genes implicated in epithelial-mesenchymal transitional, cell motility, and metastasis.
Collapse
Affiliation(s)
- Chitra Thakur
- Stony Brook Cancer Center and Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA.
| | - Yiran Qiu
- Stony Brook Cancer Center and Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Zhuoyue Bi
- Stony Brook Cancer Center and Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Ziwei Wang
- Stony Brook Cancer Center and Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Fei Chen
- Stony Brook Cancer Center and Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
3
|
Conger KO, Chidley C, Ozgurses ME, Zhao H, Kim Y, Semina SE, Burns P, Rawat V, Sheldon R, Ben-Sahra I, Frasor J, Sorger PK, DeNicola GM, Coloff JL. ASCT2 is the primary serine transporter in cancer cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.09.561530. [PMID: 37873453 PMCID: PMC10592681 DOI: 10.1101/2023.10.09.561530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
The non-essential amino acid serine is a critical nutrient for cancer cells due to its diverse biosynthetic functions. While some tumors can synthesize serine de novo, others are auxotrophic for serine and therefore reliant on the uptake of exogenous serine. Importantly, however, the transporter(s) that mediate serine uptake in cancer cells are not known. Here, we characterize the amino acid transporter ASCT2 (coded for by the gene SLC1A5) as the primary serine transporter in cancer cells. ASCT2 is well-known as a glutamine transporter in cancer, and our work demonstrates that serine and glutamine compete for uptake through ASCT2. We further show that ASCT2-mediated serine uptake is essential for purine nucleotide biosynthesis and that ERα promotes serine uptake by directly activating SLC1A5 transcription. Together, our work defines an additional important role for ASCT2 as a serine transporter in cancer and evaluates ASCT2 as a potential therapeutic target in serine metabolism.
Collapse
Affiliation(s)
- Kelly O. Conger
- Department of Physiology and Biophysics, University of Illinois Cancer Center, University of Illinois College of Medicine, Chicago, IL, USA
| | - Christopher Chidley
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
| | - Mete Emir Ozgurses
- Department of Physiology and Biophysics, University of Illinois Cancer Center, University of Illinois College of Medicine, Chicago, IL, USA
| | - Huiping Zhao
- Department of Physiology and Biophysics, University of Illinois Cancer Center, University of Illinois College of Medicine, Chicago, IL, USA
| | - Yumi Kim
- Department of Cancer Metabolism and Physiology, H. Lee. Moffitt Cancer Center, Tampa, FL, USA
| | - Svetlana E. Semina
- Department of Physiology and Biophysics, University of Illinois Cancer Center, University of Illinois College of Medicine, Chicago, IL, USA
| | - Philippa Burns
- Department of Physiology and Biophysics, University of Illinois Cancer Center, University of Illinois College of Medicine, Chicago, IL, USA
| | - Vipin Rawat
- Department of Physiology and Biophysics, University of Illinois Cancer Center, University of Illinois College of Medicine, Chicago, IL, USA
| | - Ryan Sheldon
- Metabolic and Nutritional Programming, Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, MI, USA
| | - Issam Ben-Sahra
- Robert H. Lurie Cancer Center, Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, IL, USA
| | - Jonna Frasor
- Department of Physiology and Biophysics, University of Illinois Cancer Center, University of Illinois College of Medicine, Chicago, IL, USA
| | - Peter K. Sorger
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Gina M. DeNicola
- Department of Cancer Metabolism and Physiology, H. Lee. Moffitt Cancer Center, Tampa, FL, USA
| | - Jonathan L. Coloff
- Department of Physiology and Biophysics, University of Illinois Cancer Center, University of Illinois College of Medicine, Chicago, IL, USA
| |
Collapse
|
4
|
Zhao Z, Khurana A, Antony F, Young JW, Hewton KG, Brough Z, Zhong T, Parker SJ, Duong van Hoa F. A Peptidisc-Based Survey of the Plasma Membrane Proteome of a Mammalian Cell. Mol Cell Proteomics 2023; 22:100588. [PMID: 37295717 PMCID: PMC10416069 DOI: 10.1016/j.mcpro.2023.100588] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 05/05/2023] [Accepted: 06/05/2023] [Indexed: 06/12/2023] Open
Abstract
Membrane proteins play critical roles at the cell surface and their misfunction is a hallmark of many human diseases. A precise evaluation of the plasma membrane proteome is therefore essential for cell biology and for discovering novel biomarkers and therapeutic targets. However, the low abundance of this proteome relative to soluble proteins makes it difficult to characterize, even with the most advanced proteomics technologies. Here, we apply the peptidisc membrane mimetic to purify the cell membrane proteome. Using the HeLa cell line as a reference, we capture 500 different integral membrane proteins, with half annotated to the plasma membrane. Notably, the peptidisc library is enriched with several ABC, SLC, GPCR, CD, and cell adhesion molecules that generally exist at low to very low copy numbers in the cell. We extend the method to compare two pancreatic cell lines, Panc-1 and hPSC. Here we observe a striking difference in the relative abundance of the cell surface cancer markers L1CAM, ANPEP, ITGB4, and CD70. We also identify two novel SLC transporters, SLC30A1 and SLC12A7, that are highly present in the Panc-1 cell only. The peptidisc library thus emerges as an effective way to survey and compare the membrane proteome of mammalian cells. Furthermore, since the method stabilizes membrane proteins in a water-soluble state, members of the library, here SLC12A7, can be specifically isolated.
Collapse
Affiliation(s)
- Zhiyu Zhao
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Arshdeep Khurana
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Frank Antony
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - John W Young
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Keeley G Hewton
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada; British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Zora Brough
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Tianshuang Zhong
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Seth J Parker
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada; British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Centre for Molecular Medicine and Therapeutics, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Franck Duong van Hoa
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
5
|
Thakur C, Qiu Y, Zhang Q, Carruthers NJ, Yu M, Bi Z, Fu Y, Wadgaonkar P, Almutairy B, Seno A, Stemmer PM, Chen F. Deletion of mdig enhances H3K36me3 and metastatic potential of the triple negative breast cancer cells. iScience 2022; 25:105057. [PMID: 36124233 PMCID: PMC9482110 DOI: 10.1016/j.isci.2022.105057] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 07/06/2022] [Accepted: 08/26/2022] [Indexed: 11/25/2022] Open
Abstract
In this report, we provide evidence showing diminished expression of the mineral dust-induced gene (mdig), a previously identified oncogenic gene, in human triple negative breast cancer (TNBC). Using a mouse model of orthotopic xenograft of the TNBC MDA-MB-231 cells, we demonstrate that mdig promotes the growth of primary tumors but inhibits metastasis of these cells in vivo. Knockout of mdig resulted in an enhancement of H3K36me3 in the genome and upregulation of some X chromosome-linked genes for cell motility, invasion, and metastasis. Silencing MAGED2, one of the most upregulated and H3K36me3-enriched genes resulted from mdig depletion, can partially reverse the invasive migration of the mdig knockout cells. The anti-metastatic and inhibitory role of mdig on H3K36me3 was cross-validated in another cell line, A549 lung cancer cells. Together, our data suggest that mdig is antagonist against H3K36me3 that enforces expression of genes, such as MAGED2, for cell invasion and metastasis. Loss of mdig expression in TNBC and metastatic breast cancer Knockout of mdig enforces metastasis of the TNBC cells Mdig antagonizes H3K36me3 that promotes expression of X-linked metastatic genes Silencing MAGED2 reduces invasive migration of the mdig knockout cells
Collapse
Affiliation(s)
- Chitra Thakur
- Stony Brook Cancer Center and Department of Pathology, Renaissance School of Medicine, Stony Brook University, Lauterbur Drive, Stony Brook, NY 11794, USA
| | - Yiran Qiu
- Stony Brook Cancer Center and Department of Pathology, Renaissance School of Medicine, Stony Brook University, Lauterbur Drive, Stony Brook, NY 11794, USA
| | - Qian Zhang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USA
| | - Nicholas J Carruthers
- Institute of Environmental Health Sciences, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Miaomiao Yu
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USA.,Cancer Hospital of China Medical University, 44 Xiaoheyan Road, Dadong District, Shenyang, 110042 Liaoning Province, China
| | - Zhuoyue Bi
- Stony Brook Cancer Center and Department of Pathology, Renaissance School of Medicine, Stony Brook University, Lauterbur Drive, Stony Brook, NY 11794, USA
| | - Yao Fu
- Stony Brook Cancer Center and Department of Pathology, Renaissance School of Medicine, Stony Brook University, Lauterbur Drive, Stony Brook, NY 11794, USA
| | - Priya Wadgaonkar
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USA
| | - Bandar Almutairy
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USA.,College of Pharmacy, Al-Dawadmi Campus, Shaqra University, P.O. Box 11961, Riyadh, Saudi Arabia
| | - Akimasa Seno
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USA.,Faculty of Engineering, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan
| | - Paul M Stemmer
- Institute of Environmental Health Sciences, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Fei Chen
- Stony Brook Cancer Center and Department of Pathology, Renaissance School of Medicine, Stony Brook University, Lauterbur Drive, Stony Brook, NY 11794, USA.,Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USA
| |
Collapse
|
6
|
Zhao J, Liu B, Li X. A transcription factor signature predicts the survival of patients with adrenocortical carcinoma. PeerJ 2021; 9:e12433. [PMID: 34966575 PMCID: PMC8667743 DOI: 10.7717/peerj.12433] [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] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 10/13/2021] [Indexed: 12/26/2022] Open
Abstract
Background Adrenocortical carcinoma (ACC) is a rare endocrine cancer that manifests as abdominal masses and excessive steroid hormone levels and is associated with poor clinical outcomes. Transcription factors (TFs) deregulation is found to be involved in adrenocortical tumorigenesis and cancer progression. This study aimed to construct a TF-based prognostic signature for the prediction of survival of ACC patients. Methods The gene expression profile and clinical information for ACC patients were downloaded from The Cancer Genome Atlas (TCGA, training set) and Gene Expression Omnibus (GEO, validation set) datasets after obtained 1,639 human TFs from a previously published study. The univariate Cox regression analysis was applied to identify the survival-related TFs and the LASSO Cox regression was conducted to construct the TF signature based on these survival-associated TFs candidates. Then, multivariate analysis was used to reveal the independent prognostic factors. Furthermore, Gene Set Enrichment Analysis (GSEA) was performed to analyze the significance of the TFs constituting the prognostic signature. Results LASSO Cox regression and multivariate Cox regression identified a 13-TF prognostic signature comprised of CREB3L3, NR0B1, CENPA, FOXM1, E2F2, MYBL2, HOXC11, ZIC2, ZNF282, DNMT1, TCF3, ELK4, and KLF6. The risk score based on the TF signature could classify patients into low- and high-risk groups. Kaplan-Meier analyses showed that patients in the high-risk group had significantly shorter overall survival (OS) compared to the low-risk patients. Receiver operating characteristic (ROC) curves showed that the prognostic signature predicted the OS of ACC patients with good sensitivity and specificity both in the training set (AUC > 0.9) and the validation set (AUC > 0.7). Furthermore, the TF-risk score was an independent prognostic factor. Conclusions Taken together, we identified a 13-TF prognostic marker to predict OS in ACC patients.
Collapse
Affiliation(s)
- Jianyu Zhao
- Department of Endocrinology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Bo Liu
- Department of Endocrinology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Xiaoping Li
- Department of Pediatrics Endocrinology, The First Hospital of Jilin University, Changchun, Jilin, China, Jilin, China
| |
Collapse
|
7
|
Dong C, Rao N, Du W, Gao F, Lv X, Wang G, Zhang J. mRBioM: An Algorithm for the Identification of Potential mRNA Biomarkers From Complete Transcriptomic Profiles of Gastric Adenocarcinoma. Front Genet 2021; 12:679612. [PMID: 34386038 PMCID: PMC8354214 DOI: 10.3389/fgene.2021.679612] [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] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 05/06/2021] [Indexed: 12/09/2022] Open
Abstract
Purpose In this work, an algorithm named mRBioM was developed for the identification of potential mRNA biomarkers (PmBs) from complete transcriptomic RNA profiles of gastric adenocarcinoma (GA). Methods mRBioM initially extracts differentially expressed (DE) RNAs (mRNAs, miRNAs, and lncRNAs). Next, mRBioM calculates the total information amount of each DE mRNA based on the coexpression network, including three types of RNAs and the protein-protein interaction network encoded by DE mRNAs. Finally, PmBs were identified according to the variation trend of total information amount of all DE mRNAs. Four PmB-based classifiers without learning and with learning were designed to discriminate the sample types to confirm the reliability of PmBs identified by mRBioM. PmB-based survival analysis was performed. Finally, three other cancer datasets were used to confirm the generalization ability of mRBioM. Results mRBioM identified 55 PmBs (41 upregulated and 14 downregulated) related to GA. The list included thirteen PmBs that have been verified as biomarkers or potential therapeutic targets of gastric cancer, and some PmBs were newly identified. Most PmBs were primarily enriched in the pathways closely related to the occurrence and development of gastric cancer. Cancer-related factors without learning achieved sensitivity, specificity, and accuracy of 0.90, 1, and 0.90, respectively, in the classification of the GA and control samples. Average accuracy, sensitivity, and specificity of the three classifiers with machine learning ranged within 0.94–0.98, 0.94–0.97, and 0.97–1, respectively. The prognostic risk score model constructed by 4 PmBs was able to correctly and significantly (∗∗∗p < 0.001) classify 269 GA patients into the high-risk (n = 134) and low-risk (n = 135) groups. GA equivalent classification performance was achieved using the complete transcriptomic RNA profiles of colon adenocarcinoma, lung adenocarcinoma, and hepatocellular carcinoma using PmBs identified by mRBioM. Conclusions GA-related PmBs have high specificity and sensitivity and strong prognostic risk prediction. MRBioM has also good generalization. These PmBs may have good application prospects for early diagnosis of GA and may help to elucidate the mechanism governing the occurrence and development of GA. Additionally, mRBioM is expected to be applied for the identification of other cancer-related biomarkers.
Collapse
Affiliation(s)
- Changlong Dong
- Center for Informational Biology, School of Life Sciences and Technology, University of Electronic Science and Technology of China, Chengdu, China.,School of Life Sciences and Technology, University of Electronic Science and Technology of China, Chengdu, China.,Key Laboratory for NeuroInformation of the Ministry of Education, University of Electronic Science and Technology of China, Chengdu, China
| | - Nini Rao
- Center for Informational Biology, School of Life Sciences and Technology, University of Electronic Science and Technology of China, Chengdu, China.,School of Life Sciences and Technology, University of Electronic Science and Technology of China, Chengdu, China.,Key Laboratory for NeuroInformation of the Ministry of Education, University of Electronic Science and Technology of China, Chengdu, China
| | - Wenju Du
- Center for Informational Biology, School of Life Sciences and Technology, University of Electronic Science and Technology of China, Chengdu, China.,School of Life Sciences and Technology, University of Electronic Science and Technology of China, Chengdu, China.,Key Laboratory for NeuroInformation of the Ministry of Education, University of Electronic Science and Technology of China, Chengdu, China
| | - Fenglin Gao
- Center for Informational Biology, School of Life Sciences and Technology, University of Electronic Science and Technology of China, Chengdu, China.,School of Life Sciences and Technology, University of Electronic Science and Technology of China, Chengdu, China.,Key Laboratory for NeuroInformation of the Ministry of Education, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaoqin Lv
- Center for Informational Biology, School of Life Sciences and Technology, University of Electronic Science and Technology of China, Chengdu, China.,School of Life Sciences and Technology, University of Electronic Science and Technology of China, Chengdu, China.,Key Laboratory for NeuroInformation of the Ministry of Education, University of Electronic Science and Technology of China, Chengdu, China
| | - Guangbin Wang
- Center for Informational Biology, School of Life Sciences and Technology, University of Electronic Science and Technology of China, Chengdu, China.,School of Life Sciences and Technology, University of Electronic Science and Technology of China, Chengdu, China.,Key Laboratory for NeuroInformation of the Ministry of Education, University of Electronic Science and Technology of China, Chengdu, China
| | - Junpeng Zhang
- Center for Informational Biology, School of Life Sciences and Technology, University of Electronic Science and Technology of China, Chengdu, China.,School of Life Sciences and Technology, University of Electronic Science and Technology of China, Chengdu, China.,Key Laboratory for NeuroInformation of the Ministry of Education, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
8
|
Pizzagalli MD, Bensimon A, Superti‐Furga G. A guide to plasma membrane solute carrier proteins. FEBS J 2021; 288:2784-2835. [PMID: 32810346 PMCID: PMC8246967 DOI: 10.1111/febs.15531] [Citation(s) in RCA: 241] [Impact Index Per Article: 60.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 08/07/2020] [Accepted: 08/17/2020] [Indexed: 12/13/2022]
Abstract
This review aims to serve as an introduction to the solute carrier proteins (SLC) superfamily of transporter proteins and their roles in human cells. The SLC superfamily currently includes 458 transport proteins in 65 families that carry a wide variety of substances across cellular membranes. While members of this superfamily are found throughout cellular organelles, this review focuses on transporters expressed at the plasma membrane. At the cell surface, SLC proteins may be viewed as gatekeepers of the cellular milieu, dynamically responding to different metabolic states. With altered metabolism being one of the hallmarks of cancer, we also briefly review the roles that surface SLC proteins play in the development and progression of cancer through their influence on regulating metabolism and environmental conditions.
Collapse
Affiliation(s)
- Mattia D. Pizzagalli
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Ariel Bensimon
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Giulio Superti‐Furga
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
- Center for Physiology and PharmacologyMedical University of ViennaAustria
| |
Collapse
|
9
|
Mizdrak M, Tičinović Kurir T, Božić J. The Role of Biomarkers in Adrenocortical Carcinoma: A Review of Current Evidence and Future Perspectives. Biomedicines 2021; 9:174. [PMID: 33578890 PMCID: PMC7916711 DOI: 10.3390/biomedicines9020174] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/07/2021] [Accepted: 02/08/2021] [Indexed: 12/18/2022] Open
Abstract
Adrenocortical carcinoma (ACC) is a rare endocrine malignancy arising from the adrenal cortex often with unexpected biological behavior. It can occur at any age, with two peaks of incidence: in the first and between fifth and seventh decades of life. Although ACC are mostly hormonally active, precursors and metabolites, rather than end products of steroidogenesis are produced by dedifferentiated and immature malignant cells. Distinguishing the etiology of adrenal mass, between benign adenomas, which are quite frequent in general population, and malignant carcinomas with dismal prognosis is often unfeasible. Even after pathohistological analysis, diagnosis of adrenocortical carcinomas is not always straightforward and represents a great challenge for experienced and multidisciplinary expert teams. No single imaging method, hormonal work-up or immunohistochemical labelling can definitively prove the diagnosis of ACC. Over several decades' great efforts have been made in finding novel reliable and available diagnostic and prognostic factors including steroid metabolome profiling or target gene identification. Despite these achievements, the 5-year mortality rate still accounts for approximately 75% to 90%, ACC is frequently diagnosed in advanced stages and therapeutic options are unfortunately limited. Therefore, imperative is to identify new biological markers that can predict patient prognosis and provide new therapeutic options.
Collapse
Affiliation(s)
- Maja Mizdrak
- Department of Nephrology and Hemodialysis, University Hospital of Split, 21000 Split, Croatia;
- Department of Pathophysiology, University of Split School of Medicine, 21000 Split, Croatia;
| | - Tina Tičinović Kurir
- Department of Pathophysiology, University of Split School of Medicine, 21000 Split, Croatia;
- Department of Endocrinology, Diabetes and Metabolic Disorders, University Hospital of Split, 21000 Split, Croatia
| | - Joško Božić
- Department of Pathophysiology, University of Split School of Medicine, 21000 Split, Croatia;
| |
Collapse
|
10
|
El Khoury LY, Fu S, Hlady RA, Wagner RT, Wang L, Eckel-Passow JE, Castle EP, Stanton ML, Thompson RH, Parker AS, Ho TH, Robertson KD. Identification of DNA methylation signatures associated with poor outcome in lower-risk Stage, Size, Grade and Necrosis (SSIGN) score clear cell renal cell cancer. Clin Epigenetics 2021; 13:12. [PMID: 33461589 PMCID: PMC7814746 DOI: 10.1186/s13148-020-00998-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 12/21/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Despite using prognostic algorithms and standard surveillance guidelines, 17% of patients initially diagnosed with low risk clear cell renal cell carcinoma (ccRCC) ultimately relapse and die of recurrent disease, indicating additional molecular parameters are needed for improved prognosis. RESULTS To address the gap in ccRCC prognostication in the lower risk population, we performed a genome-wide analysis for methylation signatures capable of distinguishing recurrent and non-recurrent ccRCCs within the subgroup classified as 'low risk' by the Mayo Clinic Stage, Size, Grade, and Necrosis score (SSIGN 0-3). This approach revealed that recurrent patients have globally hypermethylated tumors and differ in methylation significantly at 5929 CpGs. Differentially methylated CpGs (DMCpGs) were enriched in regulatory regions and genes modulating cell growth and invasion. A subset of DMCpGs stratified low SSIGN groups into high and low risk of recurrence in independent data sets, indicating that DNA methylation enhances the prognostic power of the SSIGN score. CONCLUSIONS This study reports a global DNA hypermethylation in tumors of recurrent ccRCC patients. Furthermore, DMCpGs were capable of discriminating between aggressive and less aggressive tumors, in addition to SSIGN score. Therefore, DNA methylation presents itself as a potentially strong biomarker to further improve prognostic power in patients with low risk SSIGN score (0-3).
Collapse
Affiliation(s)
- Louis Y El Khoury
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA.,Center for Individualized Medicine, Epigenomics Program, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Shuang Fu
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA.,Center for Individualized Medicine, Epigenomics Program, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.,Hematology Laboratory, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ryan A Hlady
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA.,Center for Individualized Medicine, Epigenomics Program, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Ryan T Wagner
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA.,Center for Individualized Medicine, Epigenomics Program, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Liguo Wang
- Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic, Rochester, MN, USA
| | - Jeanette E Eckel-Passow
- Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic, Rochester, MN, USA
| | - Erik P Castle
- Department of Urology, Mayo Clinic, Phoenix, AZ, USA
| | - Melissa L Stanton
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Phoenix, AZ, USA
| | | | - Alexander S Parker
- Office of Research Affairs, University of Florida, Jacksonville, FL, USA
| | - Thai H Ho
- Division of Hematology and Medical Oncology, Mayo Clinic, 13400 E. Shea Blvd, Scottsdale, AZ, 85259, USA.
| | - Keith D Robertson
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA. .,Center for Individualized Medicine, Epigenomics Program, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
11
|
Li SL, Li ZF, Cao QW, Wang WZ. SLC12A8 plays a key role in bladder cancer progression and EMT. Open Med (Wars) 2020; 16:58-67. [PMID: 33364434 PMCID: PMC7739378 DOI: 10.1515/med-2021-0013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 09/02/2020] [Accepted: 09/22/2020] [Indexed: 12/11/2022] Open
Abstract
Bladder cancer is the most common malignant tumor of the urinary system. The intention of the present research is to explore the prognostic value and biological function of solute carrier family 12 member 8 (SLC12A8) in bladder cancer. The analysis based on the TCGA and ONCOMINE database revealed that the expression of SLC12A8 in bladder cancer was notably increased compared with the normal group. SLC12A8 expression was notably correlated with the age, pathological stage, T-stage, and lymph node metastasis of bladder cancer patients. Moreover, the patients’ overall survival was notably shorter in the high SLC12A8 group. Compared with the control, SLC12A8 upregulation enhanced the proliferative, invasive, and migratory capacities of bladder cancer cells and promoted the expression of epithelial–mesenchymal transition (EMT) protein markers including β-catenin, vimentin, snail, and slug, while reduced the expression of E-cadherin. In the case of downregulated SLC12A8 expression, the proliferative, invasive, and migratory capacities of bladder cancer cells and the expression of EMT protein markers presented the opposite trend. This study demonstrated that SLC12A8 was highly correlated with oncogenesis and progression of bladder cancer, indicating that SLC12A8 may be a meaningful biomarker for initial diagnosis and early treatment of bladder cancer.
Collapse
Affiliation(s)
- Shun-Lai Li
- The Fifth People's Hospital of Jinan, Department of Urology, No. 24297, Jingshi Road, Huaiyin District, Jinan, Shandong, China
| | - Zheng-Feng Li
- The Fifth People's Hospital of Jinan, Department of Urology, No. 24297, Jingshi Road, Huaiyin District, Jinan, Shandong, China
| | - Qing-Wei Cao
- Shandong Provincial Hospital, Department of Urology, No. 9677, Jingshi Road, Lixia District, Jinan, Shandon, China
| | - Wen-Zhen Wang
- The Fifth People's Hospital of Jinan, Department of Urology, No. 24297, Jingshi Road, Huaiyin District, Jinan, Shandong, China
| |
Collapse
|
12
|
Li L, Xia J, Cui R, Kong B. Solute carrier family 12 member 8 impacts the biological behaviors of breast carcinoma cells by activating TLR/NLR signaling pathway. Cytotechnology 2020; 73:23-34. [PMID: 33505111 DOI: 10.1007/s10616-020-00439-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 11/04/2020] [Indexed: 12/21/2022] Open
Abstract
This study aimed to understand the expression of solute carrier family 12 member 8 (SLC12A8) in breast carcinoma and its biological functions, as well as its effect on the Toll-like receptor /NOD-like receptor (TLR/NLR) signaling pathway. The expression of SLC12A8 was analyzed using the public RNA sequencing dataset from TCGA database and the two datasets from Oncomine database. The former dataset was also used to evaluate the prognostic value of SLC12A8 in breast carcinoma. Real-time qPCR and western blot were applied to measure relative expression of SLC12A8. Functionally, the effect of SLC12A8 on the cells proliferation and motion was studied using cell counting kit 8 and Transwell assays respectively. Mechanistic studies were conducted using Gene Set Enrichment Analysis (GSEA) and confirmed by western blot. As a result, SLC12A8 was upregulated in breast carcinoma, and high levels of SLC12A8 led to a poorer prognosis and can be regarded as an independent prognosticator for patients with breast carcinoma. Functional experiments demonstrated that SLC12A8-knockdown suppressed while SLC12A8-overexpression elevated the viability, invasiveness and motility of breast carcinoma cells. Furthermore, GSEA indicated that high SLC12A8 was positively correlated with TLR/NLR signaling pathway. Silencing SLC12A8 significantly reduced the protein expression of TLR/NLR-related markers, whereas overexpression of SLC12A8 caused an elevation on the protein expression of these markers. All these data suggested that SLC12A8 plays a promoting effect on the cells viability, invasiveness and motility in breast carcinoma by activating TLR/NLR signaling pathway.
Collapse
Affiliation(s)
- LinWei Li
- Department of Breast Disease Center, The Affiliated Hospital of Qingdao University, No. 59 of Haier Road, Shandong 266000 Qingdao, China
| | - Jing Xia
- Department of Breast Surgery, Qingdao Central Hospital, Qingdao, 266000 Shandong China
| | - RuTing Cui
- Department of Breast Disease Center, The Affiliated Hospital of Qingdao University, No. 59 of Haier Road, Shandong 266000 Qingdao, China
| | - Bin Kong
- Department of Breast Disease Center, The Affiliated Hospital of Qingdao University, No. 59 of Haier Road, Shandong 266000 Qingdao, China
| |
Collapse
|
13
|
Brown TC, Nicolson NG, Man J, Gibson CE, Stenman A, Juhlin CC, Korah R, Carling T. Recurrent Amplification of the Osmotic Stress Transcription Factor NFAT5 in Adrenocortical Carcinoma. J Endocr Soc 2020; 4:bvaa060. [PMID: 32587934 PMCID: PMC7304660 DOI: 10.1210/jendso/bvaa060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/20/2020] [Indexed: 11/19/2022] Open
Abstract
Tumorigenesis requires mitigation of osmotic stress and the transcription factor nuclear factor of activated T cells 5 (NFAT5) coordinates this response by inducing transcellular transport of ions and osmolytes. NFAT5 modulates in vitro behavior in several cancer types, but a potential role of NFAT5 in adrenocortical carcinoma (ACC) has not been studied. A discovery cohort of 28 ACCs was selected for analysis. Coverage depth analysis of whole-exome sequencing reads assessed NFAT5 copy number alterations in 19 ACCs. Quantitative real-time PCR measured NFAT5 mRNA expression levels in 11 ACCs and 23 adrenocortical adenomas. Immunohistochemistry investigated protein expression in representative adrenal samples. The Cancer Genome Atlas database was analyzed to corroborate NFAT5 findings from the discovery cohort and to test whether NFAT5 expression correlated with ion/osmolyte channel and regulatory protein expression patterns in ACC. NFAT5 was amplified in 10 ACCs (52.6%) and clustered in the top 6% of all amplified genes. mRNA expression levels were 5-fold higher compared with adrenocortical adenomas (P < 0.0001) and NFAT5 overexpression had a sensitivity and specificity of 81.8% and 82.7%, respectively, for malignancy. Increased protein expression and nuclear localization occurred in representative ACCs. The Cancer Genome Atlas analysis demonstrated concomitant NFAT5 amplification and overexpression (P < 0.0001) that correlated with increased expression of sodium/myo-inositol transporter SLC5A3 (r2 = 0.237, P < 0.0001) and 14 other regulatory proteins (P < 0.05) previously shown to interact with NFAT5. Amplification and overexpression of NFAT5 and associated osmotic stress response related genes may play an important role adrenocortical tumorigenesis.
Collapse
Affiliation(s)
- Taylor C Brown
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Norman G Nicolson
- Department of Surgery & Yale Endocrine Neoplasia Laboratory, Yale University School of Medicine, New Haven, Connecticut
| | - Jianliang Man
- Department of Surgery & Yale Endocrine Neoplasia Laboratory, Yale University School of Medicine, New Haven, Connecticut
| | - Courtney E Gibson
- Department of Surgery & Yale Endocrine Neoplasia Laboratory, Yale University School of Medicine, New Haven, Connecticut
| | - Adam Stenman
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - C Christofer Juhlin
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.,Department of Pathology and Cytology, Karolinska University Hospital, Stockholm, Sweden
| | - Reju Korah
- Department of Surgery & Yale Endocrine Neoplasia Laboratory, Yale University School of Medicine, New Haven, Connecticut
| | - Tobias Carling
- Department of Surgery & Yale Endocrine Neoplasia Laboratory, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
14
|
Lanni JS, Peal D, Ekstrom L, Chen H, Stanclift C, Bowen ME, Mercado A, Gamba G, Kahle KT, Harris MP. Integrated K+ channel and K+Cl- cotransporter functions are required for the coordination of size and proportion during development. Dev Biol 2019; 456:164-178. [PMID: 31472116 PMCID: PMC7235970 DOI: 10.1016/j.ydbio.2019.08.016] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 08/07/2019] [Accepted: 08/23/2019] [Indexed: 10/26/2022]
Abstract
The coordination of growth during development establishes proportionality within and among the different anatomic structures of organisms. Innate memory of this proportionality is preserved, as shown in the ability of regenerating structures to return to their original size. Although the regulation of this coordination is incompletely understood, mutant analyses of zebrafish with long-finned phenotypes have uncovered important roles for bioelectric signaling in modulating growth and size of the fins and barbs. To date, long-finned mutants identified are caused by hypermorphic mutations, leaving unresolved whether such signaling is required for normal development. We isolated a new zebrafish mutant, schleier, with proportional overgrowth phenotypes caused by a missense mutation and loss of function in the K+-Cl- cotransporter Kcc4a. Creation of dominant negative Kcc4a in wild-type fish leads to loss of growth restriction in fins and barbs, supporting a requirement for Kcc4a in regulation of proportion. Epistasis experiments suggest that Kcc4a and the two-pore potassium channel Kcnk5b both contribute to a common bioelectrical signaling response in the fin. These data suggest that an integrated bioelectric signaling pathway is required for the coordination of size and proportion during development.
Collapse
Affiliation(s)
| | - David Peal
- Department of Genetics, Harvard Medical School, Boston, MA, 02124, USA; Department of Orthopaedic Research, Boston Children's Hospital, Boston, MA, 02124, USA
| | - Laura Ekstrom
- Department of Biology, Wheaton College, Norton, MA, 02766, USA
| | - Haining Chen
- Department of Biology, Wheaton College, Norton, MA, 02766, USA
| | | | - Margot E Bowen
- Department of Genetics, Harvard Medical School, Boston, MA, 02124, USA; Department of Orthopaedic Research, Boston Children's Hospital, Boston, MA, 02124, USA
| | | | - Gerardo Gamba
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México and Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico; Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico
| | - Kristopher T Kahle
- Departments of Neurosurgery, Pediatrics, and Cellular & Molecular Physiology, and NIH-Rockefeller Center for Mendelian Genomics, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Matthew P Harris
- Department of Genetics, Harvard Medical School, Boston, MA, 02124, USA; Department of Orthopaedic Research, Boston Children's Hospital, Boston, MA, 02124, USA
| |
Collapse
|
15
|
Brown TC, Nicolson NG, Stenman A, Juhlin CC, Gibson CE, Callender GG, Korah R, Carling T. Insulin-Like Growth Factor and SLC12A7 Dysregulation: A Novel Signaling Hallmark of Non-Functional Adrenocortical Carcinoma. J Am Coll Surg 2019; 229:305-315. [PMID: 31034883 DOI: 10.1016/j.jamcollsurg.2019.04.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 04/11/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND Insulin-like growth factor (IGF) dysregulation and gene copy number variations (CNV) are hallmarks of adrenocortical carcinoma (ACC). The contribution of IGF CNVs in adrenal carcinogenesis has not been studied previously. In addition, studies demonstrating an association between SLC12A7 gene amplifications and enhanced metastatic behavior in ACC, as well as reported IGF-SLC12A7 signaling interactions in other cancers, suggest a potential IGF-SLC12A7 signaling circuitry in ACC. Here we investigate the potential complicity of IGF-SLC12A7 signaling in ACC. STUDY DESIGN Insulin-like growth factor CNVs were determined by whole-exome sequencing analysis in an exploratory cohort of ACC. Quantitative polymerase chain reaction methods determined IGF1 and IGF2 expression levels and were evaluated for correlation with SLC12A7 expression and tumor characteristics. Insulin-like growth factor CNVs and expression patterns were compared with The Cancer Genome Atlas. In vitro studies determined the relationship of IGF and SLC12A7 co-expression in 2 ACC cell lines, SW-13 and NCI-H295R. Immunohistochemistry assessed IGF1 receptor (IGF1R) activation. RESULTS The IGF1 gene was amplified in 9 of 19 ACC samples, similar to findings in The Cancer Genome Atlas database. The IGF1 overexpression was observed in 5 samples and was associated with SLC12A7 overexpression and non-functional, early-stage tumors (p < 0.05). In contrast, IGF2 overexpression was associated with larger tumors (p < 0.05). In vitro IGF treatment of ACC cell lines did not stimulate SLC12A7 expression, and endogenous overexpression and silencing of SLC12A7 significantly altered IGF1 and IGF1R expression without impacting other IGFs. The IGF1R activation was associated with IGF1 overexpression in ACC tumor samples. CONCLUSIONS These findings indicate that IGF1 overexpression, caused in part by gene amplifications, is correlated with SLC12A7 overexpression in non-functional, early-stage ACCs, suggesting a potentially targeted IGF1-SLC12A7 therapeutic opportunity for these tumors.
Collapse
Affiliation(s)
- Taylor C Brown
- Department of Surgery and Yale Endocrine Neoplasia Laboratory, Yale University School of Medicine, New Haven, CT
| | - Norman G Nicolson
- Department of Surgery and Yale Endocrine Neoplasia Laboratory, Yale University School of Medicine, New Haven, CT
| | - Adam Stenman
- Department of Oncology-Pathology, Karolinska University Hospital, Stockholm, Sweden
| | - C Christofer Juhlin
- Department of Oncology-Pathology, Karolinska University Hospital, Stockholm, Sweden; Department of Pathology and Cytology, Karolinska University Hospital, Stockholm, Sweden
| | - Courtney E Gibson
- Department of Surgery and Yale Endocrine Neoplasia Laboratory, Yale University School of Medicine, New Haven, CT
| | - Glenda G Callender
- Department of Surgery and Yale Endocrine Neoplasia Laboratory, Yale University School of Medicine, New Haven, CT
| | - Reju Korah
- Department of Surgery and Yale Endocrine Neoplasia Laboratory, Yale University School of Medicine, New Haven, CT
| | - Tobias Carling
- Department of Surgery and Yale Endocrine Neoplasia Laboratory, Yale University School of Medicine, New Haven, CT.
| |
Collapse
|
16
|
McCabe MJ, Pinese M, Chan CL, Sheriff N, Thompson TJ, Grady J, Wong M, Gauthier MEA, Puttick C, Gayevskiy V, Hajdu E, Wong SQ, Barrett W, Earls P, Lukeis R, Cheng YY, Lin RCY, Thomas DM, Watkins DN, Dinger ME, McCormack AI, Cowley MJ. Genomic stratification and liquid biopsy in a rare adrenocortical carcinoma (ACC) case, with dual lung metastases. Cold Spring Harb Mol Case Stud 2019; 5:mcs.a003764. [PMID: 30936196 PMCID: PMC6549567 DOI: 10.1101/mcs.a003764] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 02/11/2019] [Indexed: 12/22/2022] Open
Abstract
Adrenocortical carcinoma is a rare malignancy with a poor prognosis and few treatment options. Molecular characterization of this cancer remains limited. We present a case of an adrenocortical carcinoma (ACC) in a 37-yr-old female, with dual lung metastases identified 1 yr following commencement of adjuvant mitotane therapy. As standard therapeutic regimens are often unsuccessful in ACC, we undertook a comprehensive genomic study into this case to identify treatment options and monitor disease progress. We performed targeted and whole-genome sequencing of germline, primary tumor, and both metastatic tumors from this patient and monitored recurrence over 2 years using liquid biopsy for ctDNA and steroid hormone measurements. Sequencing revealed the primary and metastatic tumors were hyperhaploid, with extensive loss of heterozygosity but few structural rearrangements. Loss-of-function mutations were identified in MSH2, TP53, RB1, and PTEN, resulting in tumors with mismatch repair signatures and microsatellite instability. At the cellular level, tumors were populated by mitochondria-rich oncocytes. Longitudinal ctDNA mutation and hormone profiles were unable to detect micrometastatic disease, consistent with clinical indicators of disease remission. The molecular signatures in our ACC case suggested immunotherapy in the event of disease progression; however, the patient remains free of cancer. The extensive molecular analysis presented here could be applied to other rare and/or poorly stratified cancers to identify novel or repurpose existing therapeutic options, thereby broadly improving diagnoses, treatments, and prognoses.
Collapse
Affiliation(s)
- Mark J McCabe
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia.,Hormones and Cancer Group, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia.,Faculty of Medicine, St Vincent's Clinical School, UNSW Australia, Sydney, New South Wales 2010, Australia
| | - Mark Pinese
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia.,Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - Chia-Ling Chan
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - Nisa Sheriff
- Hormones and Cancer Group, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia.,Department of Endocrinology, St Vincent's Hospital, Sydney, New South Wales 2010, Australia
| | - Tanya J Thompson
- Hormones and Cancer Group, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - John Grady
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - Marie Wong
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - Marie-Emilie A Gauthier
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - Clare Puttick
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - Velimir Gayevskiy
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - Elektra Hajdu
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - Stephen Q Wong
- Molecular and Translational Genomics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - Wade Barrett
- Department of Anatomical Pathology, St Vincent's Hospital, Sydney, New South Wales 2010, Australia
| | - Peter Earls
- Department of Anatomical Pathology, St Vincent's Hospital, Sydney, New South Wales 2010, Australia
| | - Robyn Lukeis
- Department of Anatomical Pathology, St Vincent's Hospital, Sydney, New South Wales 2010, Australia
| | - Yuen Y Cheng
- Asbestos Diseases Research Institute, The University of Sydney, Sydney, New South Wales 2139, Australia
| | - Ruby C Y Lin
- Asbestos Diseases Research Institute, The University of Sydney, Sydney, New South Wales 2139, Australia.,Centre for Infectious Diseases and Microbiology, The Westmead Institute for Medical Research, Westmead, New South Wales 2145, Australia
| | - David M Thomas
- Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - D Neil Watkins
- Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - Marcel E Dinger
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia.,Faculty of Medicine, St Vincent's Clinical School, UNSW Australia, Sydney, New South Wales 2010, Australia
| | - Ann I McCormack
- Hormones and Cancer Group, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia.,Faculty of Medicine, St Vincent's Clinical School, UNSW Australia, Sydney, New South Wales 2010, Australia.,Department of Endocrinology, St Vincent's Hospital, Sydney, New South Wales 2010, Australia
| | - Mark J Cowley
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia.,Faculty of Medicine, St Vincent's Clinical School, UNSW Australia, Sydney, New South Wales 2010, Australia.,Computational Biology Group, Children's Cancer Institute, Kensington, New South Wales 2031, Australia
| |
Collapse
|