1
|
Li Z, Qian R, Li M, Li J, Guo Y, Zhou Y, Ma C. HERC5/ISG15 Enhances Glioblastoma Stemness and Tumor Progression by mediating SERBP1protein stability. Neuromolecular Med 2025; 27:7. [PMID: 39776018 DOI: 10.1007/s12017-024-08826-w] [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: 10/28/2024] [Accepted: 12/18/2024] [Indexed: 01/30/2025]
Abstract
Glioblastoma (GBM) is the most common malignant brain tumor, and has a low survival rate and a poor prognosis. Intensive studies of pathogenic mechanisms are essential for exploring therapeutic targets for GBM. In this study, the roles played by interferon-stimulated gene 15 (ISG15), HECT, RCC1-containing protein 5 (HERC5), and SERPINE1 mRNA binding protein 1 (SERBP1) in regulating GBM cell stemness were investigated. The real-time quantitative polymerase chain reaction (qPCR), western blotting (WB), and immunohistochemistry (IHC) were used to determine the expression levels of HERC5, ISG15, and SERBP1. Cell stemness was analyzed using a cell sphere formation assay. Colony formation and cell counting kit-8 (CCK-8) assays were performed to assess cell proliferation, Transwell assays used to evaluate cell migration and invasion, and flow cytometry was used to assess cell apoptosis after treatment with temozolomide. SERBP1 stability was assessed by a CHX chase assay. A co-immunoprecipitation (Co-IP) assay verified the binding of ISG15 and HERC5 onto SERBP1. Our results showed that HERC5 and ISG15 were highly expressed in GBM. HERC5 and ISG15 promoted the cell stemness of GBM, and increased cell proliferation, sphere formation, migration, invasion, and chemoresistance. Moreover, HERC5 and ISG15 played a synergistic role in promoting the cell stemness of GBM. We also found that HERC5/ISG15 promoted the stability of SERBP1, which also promoted the cell stemness of GBM. The tumor-promoting role of HERC5 and ISG15 was also confirmed in a subcutaneous xenograft tumor model. Collectively, HERC5/ISG15 was found to regulate GBM stemness and tumor progression by mediating SERBP1 protein stability. Our present study suggests a promising therapeutic target for GBM.
Collapse
Affiliation(s)
- Zhixiao Li
- Department of Neurosurgery, Henan Provincial People's Hospital, No. 7 Weiwu Road, Zhengzhou, 450003, Henan Province, China
| | - Rongjun Qian
- Department of Neurosurgery, Henan Provincial People's Hospital, No. 7 Weiwu Road, Zhengzhou, 450003, Henan Province, China
- Department of Neurosurgery, People's Hospital of Henan University, Zhengzhou, China
- Department of Neurosurgery, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengda Li
- Department of Neurosurgery, People's Hospital of Henan University, Zhengzhou, China
| | - Juntao Li
- Department of Neurosurgery, Henan Provincial People's Hospital, No. 7 Weiwu Road, Zhengzhou, 450003, Henan Province, China
| | - Yongji Guo
- Department of Neurosurgery, People's Hospital of Henan University, Zhengzhou, China
| | - Yuanhang Zhou
- Department of Neurosurgery, People's Hospital of Henan University, Zhengzhou, China
| | - Chunxiao Ma
- Department of Neurosurgery, Henan Provincial People's Hospital, No. 7 Weiwu Road, Zhengzhou, 450003, Henan Province, China.
- Department of Neurosurgery, People's Hospital of Henan University, Zhengzhou, China.
- Department of Neurosurgery, People's Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
2
|
Du Y, Wang L, Perez-Castro L, Conacci-Sorrell M, Sieber M. Non-cell autonomous regulation of cell-cell signaling and differentiation by mitochondrial ROS. J Cell Biol 2024; 223:e202401084. [PMID: 39535785 PMCID: PMC11561560 DOI: 10.1083/jcb.202401084] [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: 01/17/2024] [Revised: 05/14/2024] [Accepted: 08/13/2024] [Indexed: 11/16/2024] Open
Abstract
Mitochondrial reactive oxygen species (ROS) function intrinsically within cells to induce cell damage, regulate transcription, and cause genome instability. However, we know little about how mitochondrial ROS production non-cell autonomously impacts cell-cell signaling. Here, we show that mitochondrial dysfunction inhibits the plasma membrane localization of cell surface receptors that drive cell-cell communication during oogenesis. Within minutes, we found that mitochondrial ROS impairs exocyst membrane binding and leads to defective endosomal recycling. This endosomal defect impairs the trafficking of receptors, such as the Notch ligand Delta, during oogenesis. Remarkably, we found that overexpressing RAB11 restores ligand trafficking and rescues the developmental defects caused by ROS production. ROS production from adjacent cells acutely initiates a transcriptional response associated with growth and migration by suppressing Notch signaling and inducing extra cellualr matrix (ECM) remodeling. Our work reveals a conserved rapid response to ROS production that links mitochondrial dysfunction to the non-cell autonomous regulation of cell-cell signaling.
Collapse
Affiliation(s)
- Yipeng Du
- Department of Physiology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Lei Wang
- Department of Physiology, UT Southwestern Medical Center, Dallas, TX, USA
| | | | | | - Matthew Sieber
- Department of Physiology, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
3
|
Zhang Q, Du Z, Zhou W, Li W, Yang Q, Yu H, Liu T. ZDHHC1 downregulates LIPG and inhibits colorectal cancer growth via IGF2BP1 Palmitoylation. Cancer Gene Ther 2024; 31:1427-1437. [PMID: 39069526 PMCID: PMC11405259 DOI: 10.1038/s41417-024-00808-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 07/05/2024] [Accepted: 07/10/2024] [Indexed: 07/30/2024]
Abstract
Alteration in lipid metabolism is recognized as a hallmark feature of colorectal cancer (CRC). Protein S-palmitoylation plays a critical role in many different cellular processes including protein-lipid interaction. Zinc Finger DHHC-Type Containing 1 (ZDHHC1, also known as ZNF377) belongs to the palmitoyl-transferase ZDHHC family, and is a potential tumor suppressor. However, our knowledge of the functional roles of ZDHHC1 in CRC is limited. We discovered that ZDHHC1 expression was downregulated in CRC tissues and that low levels of ZDHHC1 were associated with unfavorable prognosis. Functional studies showed that ZDHHC1 inhibited CRC cell proliferation and invasion in vitro and in vivo. We also found that lipase G (LIPG) is negatively regulated by ZDHHC1 and plays a key role in CRC cell growth through lipid storage. Additionally, we demonstrated that ZDHHC1 functions as a IGF2BP1-palmitoylating enzyme that induces S-palmitoylation at IGF2BP1-C337, which results in downregulated LIPG expression via m6A modification. Mechanistic investigations revealed that the ZDHHC1/IGF2BP1/LIPG signaling axis is associated with inhibition of CRC cell growth. Our study uncovers the potential role of ZDHHC1 in CRC, including inhibition of CRC growth by reducing the stability of LIPG mRNA in an m6A dependent-manner by palmitoylation of IGF2BP1.
Collapse
Affiliation(s)
- Qun Zhang
- Department of Digestive Surgical Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhouyuan Du
- Department of Digestive Surgical Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wei Zhou
- Department of Digestive Surgical Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wei Li
- Department of Digestive Surgical Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qinglin Yang
- Department of Digestive Surgical Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Haixin Yu
- Department of Digestive Surgical Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Tao Liu
- Department of Digestive Surgical Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
4
|
Davoudi P, Do DN, Rathgeber B, Colombo S, Sargolzaei M, Plastow G, Wang Z, Miar Y. Identification of consensus homozygous regions and their associations with growth and feed efficiency traits in American mink. BMC Genom Data 2024; 25:68. [PMID: 38982354 PMCID: PMC11234557 DOI: 10.1186/s12863-024-01252-8] [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: 03/15/2024] [Accepted: 07/01/2024] [Indexed: 07/11/2024] Open
Abstract
The recent chromosome-based genome assembly and the newly developed 70K single nucleotide polymorphism (SNP) array for American mink (Neogale vison) facilitate the identification of genetic variants underlying complex traits in this species. The objective of this study was to evaluate the association between consensus runs of homozygosity (ROH) with growth and feed efficiency traits in American mink. A subsample of two mink populations (n = 2,986) were genotyped using the Affymetrix Mink 70K SNP array. The identified ROH segments were included simultaneously, concatenated into consensus regions, and the ROH-based association studies were carried out with linear mixed models considering a genomic relationship matrix for 11 growth and feed efficiency traits implemented in ASReml-R version 4. In total, 298,313 ROH were identified across all individuals, with an average length and coverage of 4.16 Mb and 414.8 Mb, respectively. After merging ROH segments, 196 consensus ROH regions were detected and used for genome-wide ROH-based association analysis. Thirteen consensus ROH regions were significantly (P < 0.01) associated with growth and feed efficiency traits. Several candidate genes within the significant regions are known for their involvement in growth and body size development, including MEF2A, ADAMTS17, POU3F2, and TYRO3. In addition, we found ten consensus ROH regions, defined as ROH islands, with frequencies over 80% of the population. These islands harbored 12 annotated genes, some of which were related to immune system processes such as DTX3L, PARP9, PARP14, CD86, and HCLS1. This is the first study to explore the associations between homozygous regions with growth and feed efficiency traits in American mink. Our findings shed the light on the effects of homozygosity in the mink genome on growth and feed efficiency traits, that can be utilized in developing a sustainable breeding program for mink.
Collapse
Affiliation(s)
- Pourya Davoudi
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| | - Duy Ngoc Do
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| | - Bruce Rathgeber
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| | - Stefanie Colombo
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| | - Mehdi Sargolzaei
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada
- Select Sires Inc, Plain City, OH, USA
| | - Graham Plastow
- Department of Agricultural, Food and Nutritional Science, Livestock Gentec, University of Alberta, Edmonton, AB, Canada
| | - Zhiquan Wang
- Department of Agricultural, Food and Nutritional Science, Livestock Gentec, University of Alberta, Edmonton, AB, Canada
| | - Younes Miar
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada.
| |
Collapse
|
5
|
Huang J, Liu R, Zhang Y, Sheng X. Mechanistic analysis of endothelial lipase G promotion of the occurrence and development of cervical carcinoma by activating the phosphatidylinositol-4,5-bisphosphate 3-kinase/protein kinase B/mechanistic target of rapamycin kinase signalling pathway. J OBSTET GYNAECOL 2023; 43:2151353. [PMID: 36606668 DOI: 10.1080/01443615.2022.2151353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Lipase G, endothelial type (LIPG) is expressed abundantly in tissues with a high metabolic rate and vascularisation. Research on LIPG has focussed on metabolic syndromes. However, the role of LIPG in providing lipid precursors suggests that it might function in the metabolism of carcinoma cells. Analysis in The Cancer Genome Atlas indicated that patients with cervical carcinoma with high LIPG expression had a lower survival prognosis compared with patients with low LIPG expression. The mechanism underlying the effects of LIPG in cervical carcinoma is unclear. The present study aimed to determine the role of LIPG in cervical carcinoma and its mechanism. The results showed that the LIPG expression level was higher in cervical cancer. Downregulation of LIPG expression inhibited cell migration, invasion, proliferation, and the formation of cell colonies, but increased the rate of apoptosis. The Human papillomavirus E6 protein might reduce the expression of miR-148a-3p, relieve the inhibitory effect of miR-148a-3p on LIPG expression, and promote the progression of cervical cancer through the phosphatidylinositol-4,5-bisphosphate 3-kinase/protein kinase B/mechanistic target of rapamycin kinase signalling pathway.IMPACT STATEMENTWhat is already known on this subject? LIPG provides lipid precursors, suggesting that it might function in the metabolism of carcinoma cellsWhat do the results of this study add? LIPG might be regulated by HPV16 E6/miR-148a-3p and promote cervical carcinoma progression via the PI3K/AKT/mTOR signalling pathway.What are the implications of these finding for clinical practice and/or further research? The results indicated that novel treatment and diagnosis strategies for cervical carcinoma could be developed related to LIPG. However, the detailed relationship between LIPG and cervical carcinoma remains to be fully determined.
Collapse
Affiliation(s)
- Jing Huang
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Department of Gynecologic Oncology Research Office, Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Renci Liu
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Department of Gynecologic Oncology Research Office, Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yiwen Zhang
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Department of Gynecologic Oncology Research Office, Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiujie Sheng
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Department of Gynecologic Oncology Research Office, Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
6
|
Yi J, Wu M, Zheng Z, Zhou Q, Li X, Lu Y, Liu P. Integrated analysis of DNA methylome and transcriptome reveals SFRP1 and LIPG as potential drivers of ovarian cancer metastasis. J Gynecol Oncol 2023; 34:e71. [PMID: 37417299 PMCID: PMC10627750 DOI: 10.3802/jgo.2023.34.e71] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 04/17/2023] [Accepted: 05/13/2023] [Indexed: 07/08/2023] Open
Abstract
OBJECTIVE More than 75% of ovarian cancer patients are diagnosed at advanced stages and die of tumor cell metastasis. This study aimed to identify new epigenetic and transcriptomic alterations associated with ovarian cancer metastasis. METHODS Two cell sublines with low- and high-metastasis potentials were derived from the ovarian cancer cell line A2780. Genome-wide DNA methylome and transcriptome profiling were carried out in these two sublines by Reduced Representation Bisulfite Sequencing and RNA-seq technologies. Cell-based assays were conducted to support the clinical findings. RESULTS There are distinct DNA methylation and gene expression patterns between the two cell sublines with low- and high-metastasis potentials. Integrated analysis identified 33 methylation-induced genes potentially involved in ovarian cancer metastasis. The DNA methylation patterns of two of them (i.e., SFRP1 and LIPG) were further validated in human specimens, indicating that they were hypermethylated and downregulated in peritoneal metastatic ovarian carcinoma compared to primary ovarian carcinoma. Patients with lower SFRP1 and LIPG expression tend to have a worse prognosis. Functionally, knockdown of SFRP1 and LIPG promoted cell growth and migration, whereas their overexpression resulted in the opposite effects. In particular, knockdown of SFRP1 could phosphorylate GSK3β and increase β-catenin expression, leading to deregulated activation of the Wnt/β-catenin signaling. CONCLUSION Many systemic and important epigenetic and transcriptomic alterations occur in the progression of ovarian cancer. In particular, epigenetic silencing of SFRP1 and LIPG is a potential driver event in ovarian cancer metastasis. They can be used as prognostic biomarkers and therapeutic targets for ovarian cancer patients.
Collapse
Affiliation(s)
- Jiani Yi
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Sir Run Run Shaw Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Mengting Wu
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Department of Gynecologic Oncology, Women's Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhihong Zheng
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Department of Gynecologic Oncology, Women's Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Qing Zhou
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Department of Gynecologic Oncology, Women's Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xufan Li
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Sir Run Run Shaw Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Lu
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Department of Gynecologic Oncology, Women's Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Pengyuan Liu
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Sir Run Run Shaw Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China.
| |
Collapse
|
7
|
Yuan Y, Qin H, Li H, Shi W, Bao L, Xu S, Yin J, Zheng L. The Functional Roles of ISG15/ISGylation in Cancer. Molecules 2023; 28:molecules28031337. [PMID: 36771004 PMCID: PMC9918931 DOI: 10.3390/molecules28031337] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/11/2023] [Accepted: 01/28/2023] [Indexed: 02/01/2023] Open
Abstract
The protein ISG15 encoded by interferon-stimulated gene (ISG) 15 is the first identified member of the ubiquitin-like protein family and exists in the form of monomers and conjugated complexes. Like ubiquitin, ISG15 can mediate an ubiquitin-like modification by covalently modifying other proteins, known as ISGylation. There is growing evidence showing that both the free and conjugated ISG15 are involved in multiple key cellular processes, including autophagy, exosome secretion, DNA repair, immune regulation, and cancer occurrence and progression. In this review, we aim to further clarify the function of ISG15 and ISGylation in cancer, demonstrate the important relationship between ISG15/ISGylation and cancer, and emphasize new insights into the different roles of ISG15/ISGylation in cancer progression. This review may contribute to therapeutic intervention in cancer. However, due to the limitations of current research, the regulation of ISG15/ISGylation on cancer progression is not completely clear, thus further comprehensive and sufficient correlation studies are still needed.
Collapse
Affiliation(s)
- Yin Yuan
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Medicinal Chemistry, School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China
| | - Hai Qin
- Department of Clinical Laboratory, Guizhou Provincial Orthopedic Hospital, No. 206, Sixian Street, Baiyun District, Guiyang 550002, China
| | - Huilong Li
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Medicinal Chemistry, School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China
| | - Wanjin Shi
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Medicinal Chemistry, School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China
| | - Lichen Bao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing 210029, China
| | - Shengtao Xu
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Medicinal Chemistry, School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China
| | - Jun Yin
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Medicinal Chemistry, School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China
- Correspondence: (J.Y.); (L.Z.)
| | - Lufeng Zheng
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Medicinal Chemistry, School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China
- Correspondence: (J.Y.); (L.Z.)
| |
Collapse
|
8
|
Wang S, Chen Z, Lv H, wang C, Wei H, Yu J. LIPG is a novel prognostic biomarker and correlated with immune infiltrates in lung adenocarcinoma. J Clin Lab Anal 2022; 37:e24824. [PMID: 36572999 PMCID: PMC9833968 DOI: 10.1002/jcla.24824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/03/2022] [Accepted: 12/14/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Although many biomarkers for lung adenocarcinoma (LUAD) have been identified, their specificity and sensitivity remain unsatisfactory. Endothelial lipase gene (LIPG) plays an important role in a variety of cancers, but its role in lung adenocarcinoma remains unclear. METHODS TCGA, GEO, K-M plotter, CIBERSORT, GSEA, HPA, and GDSC were used to analyze LIPG in LUAD. Data analysis was mainly achieved by R 4.0.3. RESULTS The expression of LIPG in LUAD tissues was higher than that in adjacent normal tissues, especially in women, patients aged >65 years, and those with lymph node metastasis. High expression predicted a poor prognosis. The results of enrichment analysis suggest that LIPG may exert profound effects on the development of LUAD through multiple stages of lipid metabolism and immune system regulation. In addition, LIPG expression was significantly correlated with the expression levels of multiple immune checkpoint genes and the abundance of multiple immune infiltrates, including the activated memory CD4 T cell, M1 macrophage, neutrophil, plasma cells, and T follicular helper (Tfh) cells in the LUAD microenvironment content. At the same time, patients with high LIPG expression respond well to a variety of antitumor drugs and have a low rate of drug resistance. CONCLUSIONS LIPG is a prognostic marker and is associated with lipid metabolism and immune infiltration in LUAD.
Collapse
Affiliation(s)
- Shan Wang
- Cancer Center, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
| | - Zhaoxin Chen
- Cancer Center, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
| | - Hongwei Lv
- Cancer Center, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
| | - Cong wang
- Cancer Center, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
| | - Huamin Wei
- Department of Traditional Chinese Medicine, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
| | - Jing Yu
- Cancer Center, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
9
|
The diverse repertoire of ISG15: more intricate than initially thought. Exp Mol Med 2022; 54:1779-1792. [PMID: 36319753 PMCID: PMC9722776 DOI: 10.1038/s12276-022-00872-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 08/03/2022] [Accepted: 08/08/2022] [Indexed: 11/05/2022] Open
Abstract
ISG15, the product of interferon (IFN)-stimulated gene 15, is the first identified ubiquitin-like protein (UBL), which plays multifaceted roles not only as a free intracellular or extracellular molecule but also as a post-translational modifier in the process of ISG15 conjugation (ISGylation). ISG15 has only been identified in vertebrates, indicating that the functions of ISG15 and its conjugation are restricted to higher eukaryotes and have evolved with IFN signaling. Despite the highlighted complexity of ISG15 and ISGylation, it has been suggested that ISG15 and ISGylation profoundly impact a variety of cellular processes, including protein translation, autophagy, exosome secretion, cytokine secretion, cytoskeleton dynamics, DNA damage response, telomere shortening, and immune modulation, which emphasizes the necessity of reassessing ISG15 and ISGylation. However, the underlying mechanisms and molecular consequences of ISG15 and ISGylation remain poorly defined, largely due to a lack of knowledge on the ISG15 target repertoire. In this review, we provide a comprehensive overview of the mechanistic understanding and molecular consequences of ISG15 and ISGylation. We also highlight new insights into the roles of ISG15 and ISGylation not only in physiology but also in the pathogenesis of various human diseases, especially in cancer, which could contribute to therapeutic intervention in human diseases.
Collapse
|
10
|
A Proof-of-Concept Inhibitor of Endothelial Lipase Suppresses Triple-Negative Breast Cancer Cells by Hijacking the Mitochondrial Function. Cancers (Basel) 2022; 14:cancers14153763. [PMID: 35954428 PMCID: PMC9367514 DOI: 10.3390/cancers14153763] [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: 07/06/2022] [Revised: 07/24/2022] [Accepted: 07/30/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Endothelial lipase (EL/LIPG) is a key regulator of tumor cell metabolism. In triple-negative breast cancer (TNBC) cells, we find that the expression of LIPG is associated with long non-coding RNA DANCR and positively correlates with gene signatures of mitochondrial metabolism-oxidative phosphorylation (OXPHOS). DANCR binds to LIPG, which enables tumor cells to maintain the expression. Importantly, LIPG knockdown inhibits OXPHOS and TNBC tumor formation. Finally, our study identifies a natural compound, the LIPG inhibitor cynaroside, which provides a new therapeutic strategy against TNBC. Abstract Triple-negative breast cancer (TNBC) cells reprogram their metabolism to provide metabolic flexibility for tumor cell growth and survival in the tumor microenvironment. While our previous findings indicated that endothelial lipase (EL/LIPG) is a hallmark of TNBC, the precise mechanism through which LIPG instigates TNBC metabolism remains undefined. Here, we report that the expression of LIPG is associated with long non-coding RNA DANCR and positively correlates with gene signatures of mitochondrial metabolism-oxidative phosphorylation (OXPHOS). DANCR binds to LIPG, enabling tumor cells to maintain LIPG protein stability and OXPHOS. As one mechanism of LIPG in the regulation of tumor cell oxidative metabolism, LIPG mediates histone deacetylase 6 (HDAC6) and histone acetylation, which contribute to changes in IL-6 and fatty acid synthesis gene expression. Finally, aided by a relaxed docking approach, we discovered a new LIPG inhibitor, cynaroside, that effectively suppressed the enzyme activity and DANCR in TNBC cells. Treatment with cynaroside inhibited the OXPHOS phenotype of TNBC cells, which severely impaired tumor formation. Taken together, our study provides mechanistic insights into the LIPG modulation of mitochondrial metabolism in TNBC and a proof-of-concept that targeting LIPG is a promising new therapeutic strategy for the treatment of TNBC.
Collapse
|
11
|
Gilson Sena IF, Fernandes LL, Lorandi LL, Santana TV, Cintra L, Lima IF, Iwai LK, Kramer JM, Birbrair A, Heller D. Identification of early biomarkers in saliva in genetically engineered mouse model C(3)1-TAg of breast cancer. Sci Rep 2022; 12:11544. [PMID: 35798767 PMCID: PMC9263110 DOI: 10.1038/s41598-022-14514-1] [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: 01/13/2022] [Accepted: 06/08/2022] [Indexed: 11/09/2022] Open
Abstract
Breast cancer is one of leading causes of death worldwide in the female population. Deaths from breast cancer could be reduced significantly through earlier and more efficient detection of the disease. Saliva, an oral fluid that contains an abundance of protein biomarkers, has been recognized as a promising diagnostic biofluid that is easy to isolate through non-invasive techniques. Assays on saliva can be performed rapidly and are cost-effective. Therefore, our work aimed to identify salivary biomarkers present in the initial stages of breast cancer, where cell alterations are not yet detectable by histopathological analysis. Using state-of-the-art techniques, we employed a transgenic mouse model of mammary cancer to identify molecular changes in precancerous stage breast cancer through protein analysis in saliva. Through corroborative molecular approaches, we established that proteins related to metabolic changes, inflammatory process and cell matrix degradation are detected in saliva at the onset of tumor development. Our work demonstrated that salivary protein profiles can be used to identify cellular changes associated with precancerous stage breast cancer through non-invasive means even prior to biopsy-evident disease.
Collapse
Affiliation(s)
| | | | | | | | | | - Ismael Feitosa Lima
- Laboratory of Applied Toxicology, Center of Toxins, Immune-Response and Cell Signaling (LETA/CeTICS), Instituto Butantan, São Paulo, Brazil
| | - Leo Kei Iwai
- Laboratory of Applied Toxicology, Center of Toxins, Immune-Response and Cell Signaling (LETA/CeTICS), Instituto Butantan, São Paulo, Brazil
| | - Jill M Kramer
- Department of Oral Biology, School of Dental Medicine, The University of Buffalo, State University of New York, Buffalo, NY, USA
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil. .,Department of Dermatology, Medical Sciences Center, University of Wisconsin-Madison, Rm 4385, 1300 University Avenue, Madison, WI, 53706, USA. .,Department of Radiology, Columbia University Medical Center, New York, NY, USA.
| | - Débora Heller
- Post Graduate Program in Dentistry, Cruzeiro do Sul University, São Paulo, Brazil. .,Hospital Israelita Albert Einstein, São Paulo, Brazil. .,Department of Periodontology, University of Texas Health Science Center San Antonio, San Antonio, TX, USA.
| |
Collapse
|
12
|
Mirzalieva O, Juncker M, Schwartzenburg J, Desai S. ISG15 and ISGylation in Human Diseases. Cells 2022; 11:cells11030538. [PMID: 35159348 PMCID: PMC8834048 DOI: 10.3390/cells11030538] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/18/2022] [Accepted: 01/25/2022] [Indexed: 12/04/2022] Open
Abstract
Type I Interferons (IFNs) induce the expression of >500 genes, which are collectively called ISGs (IFN-stimulated genes). One of the earliest ISGs induced by IFNs is ISG15 (Interferon-Stimulated Gene 15). Free ISG15 protein synthesized from the ISG15 gene is post-translationally conjugated to cellular proteins and is also secreted by cells into the extracellular milieu. ISG15 comprises two ubiquitin-like domains (UBL1 and UBL2), each of which bears a striking similarity to ubiquitin, accounting for its earlier name ubiquitin cross-reactive protein (UCRP). Like ubiquitin, ISG15 harbors a characteristic β-grasp fold in both UBL domains. UBL2 domain has a conserved C-terminal Gly-Gly motif through which cellular proteins are appended via an enzymatic cascade similar to ubiquitylation called ISGylation. ISG15 protein is minimally expressed under physiological conditions. However, its IFN-dependent expression is aberrantly elevated or compromised in various human diseases, including multiple types of cancer, neurodegenerative disorders (Ataxia Telangiectasia and Amyotrophic Lateral Sclerosis), inflammatory diseases (Mendelian Susceptibility to Mycobacterial Disease (MSMD), bacteriopathy and viropathy), and in the lumbar spinal cords of veterans exposed to Traumatic Brain Injury (TBI). ISG15 and ISGylation have both inhibitory and/or stimulatory roles in the etiology and pathogenesis of human diseases. Thus, ISG15 is considered a “double-edged sword” for human diseases in which its expression is elevated. Because of the roles of ISG15 and ISGylation in cancer cell proliferation, migration, and metastasis, conferring anti-cancer drug sensitivity to tumor cells, and its elevated expression in cancer, neurodegenerative disorders, and veterans exposed to TBI, both ISG15 and ISGylation are now considered diagnostic/prognostic biomarkers and therapeutic targets for these ailments. In the current review, we shall cover the exciting journey of ISG15, spanning three decades from the bench to the bedside.
Collapse
Affiliation(s)
| | | | | | - Shyamal Desai
- Correspondence: ; Tel.: +1-504-568-4388; Fax: +1-504-568-2093
| |
Collapse
|
13
|
Construction and validation of a metabolic gene-associated prognostic model for cervical carcinoma and the role on tumor microenvironment and immunity. Aging (Albany NY) 2021; 13:25072-25088. [PMID: 34852326 PMCID: PMC8714137 DOI: 10.18632/aging.203723] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 10/25/2021] [Indexed: 12/14/2022]
Abstract
Metabolic reprogramming is a common feature of tumor cells and is associated with tumorigenesis and progression. In this study, a metabolic gene-associated prognostic model (MGPM) was constructed using multiple bioinformatics analysis methods in cervical carcinoma (CC) tissues from The Cancer Genome Atlas (TCGA) database, which comprised fifteen differentially expressed metabolic genes (DEMGs). Patients were divided into a high-risk group with shorter overall survival (OS) and a low-risk group with better survival. Receiver operating characteristic (ROC) curve analysis showed that the MGPM precisely predicted the 1-, 3- and 5-year survival of CC patients. As expected, MGPM exhibited a favorable prognostic significance in the training and testing datasets of TCGA. And the clinicopathological parameters including stage, tumor (T) and metastasis (M) classifications had significant differences in low- and high-risk groups, which further demonstrated the MGPM had a favorite prognostic prediction ability. Additionally, patients with low-ESTMATEScore had a shorter OS and when those combined with high-risk scores presented a worse prognosis. Through “CIBERSORT” package and Wilcoxon rank-sum test, patients in the high-risk group with a poor prognosis showed lower levels of infiltration of T cell CD8 (P < 0.001), T cells memory activated (P = 0.010) and mast cells resting (P < 0.001), and higher levels of mast cells activated (P < 0.001), and we also found these patients had a worse response for immunosuppressive therapy. These findings demonstrate that MGPM accurately predicts survival outcomes in CC patients, which will be helpful for further optimizing immunotherapies for cancer by reprogramming its cell metabolism.
Collapse
|
14
|
Song F, Zhang Y, Pan Z, Hu X, Yi Y, Zheng X, Wei H, Huang P. Identification of novel key genes associated with the metastasis of prostate cancer based on bioinformatics prediction and validation. Cancer Cell Int 2021; 21:559. [PMID: 34696780 PMCID: PMC8547030 DOI: 10.1186/s12935-021-02258-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 10/11/2021] [Indexed: 11/22/2022] Open
Abstract
Background Metastatic prostate cancer (PCa) is a lethal tumor. However, the molecular mechanisms underlying PCa progression have not been fully elucidated. Methods Transcriptome expression profiling and clinical information on primary and metastatic PCa samples were obtained from TCGA. R software was used to screen the DEGs, and LASSO logistical regression method was utilized to identify the pivotal PCa metastasis-related DEGs. The transcriptional expression levels of the key genes were analyzed using the UALCAN database, and the corresponding protein expression were validated by Immunohistochemistry (IHC). Survival analysis of the key genes was performed using the GEPIA database. Wound healing assay and Transwell assay were conducted to determine whether knockdown of the key genes influence the migration and invasion abilities of PCa cells (22Rv1 and PC3). GSEA was performed to predict key genes-mediated signaling pathways for the development of PCa. Western blotting was used to evaluate the expression changes of E-cadherin, Twist1, and Vimentin in PCa cells with the key genes silencing. An in vivo mouse metastatic model for PCa was also generated to verify the important role of ISG15 and CST2 in PCa metastasis. Results A comparison between primary and metastatic PCa tissues was conducted, and 19 DEGs were screened. Among these, three key genes were identified that might be closely associated with PCa progression according to the LASSO logistical analysis, namely ISG15, DNAH8, and CST2. Further functional experiments revealed that knockdown of ISG15 and CST2 suppressed wound healing, migration, and invasion of PCa cells. To explore the molecular mechanism of ISG15 and CST2 in the development of PCa, GSEA was performed, and it was found that both genes play crucial roles in cell adhesion molecules, extracellular matrix-receptor interaction, and focal adhesion. Western blotting results exhibited that inhibiting ISG15 and CST2 led to increase the expression of E-cadherin and decrease the expression of Twist1 and Vimentin. Additionally, the metastatic in vivo study demonstrated that both PC3 and 22Rv1 cells expressing with luciferase-shISG15 and luciferase-shCST2 had significantly lower detectable bioluminescence than that in the control PCa cells. Conclusion ISG15 and CST2 may participate in PCa metastasis by regulating the epithelial-mesenchymal transition (EMT) signaling pathway. These findings may help to better understand the pathogenetic mechanisms governing PCa and provide promising therapeutic targets for metastatic PCa therapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02258-3.
Collapse
Affiliation(s)
- Feifeng Song
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.,Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, 310014, Zhejiang, China
| | - Yiwen Zhang
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.,Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, 310014, Zhejiang, China
| | - Zongfu Pan
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.,Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, 310014, Zhejiang, China
| | - Xiaoping Hu
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Yaodong Yi
- Laboratory of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xiaochun Zheng
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Haibin Wei
- Department of Urology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Ping Huang
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China. .,Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, 310014, Zhejiang, China.
| |
Collapse
|
15
|
Manupati K, Yeeravalli R, Kaushik K, Singh D, Mehra B, Gangane N, Gupta A, Goswami K, Das A. Activation of CD44-Lipoprotein lipase axis in breast cancer stem cells promotes tumorigenesis. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166228. [PMID: 34311079 DOI: 10.1016/j.bbadis.2021.166228] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/25/2021] [Accepted: 07/20/2021] [Indexed: 01/16/2023]
Abstract
Breast cancer stem cells (CSCs) are distinct CD44+-subpopulations that are involved in metastasis and chemoresistance. However, the underlying molecular mechanism of CD44 in breast CSCs-mediated tumorigenesis remains elusive. We observed high CD44 expression in advanced-stage clinical breast tumor samples. CD44 activation in breast CSCs sorted from various triple negative breast cancer (TNBC) cell lines induced proliferation, migration, invasion, mammosphere formation that were reversed in presence of inhibitor, 4-methyl umbelliferone or CD44 silencing. CD44 activation in breast CSCs induced Src, Akt, and nuclear translocation of pSTAT3. PCR arrays revealed differential expression of a metabolic gene, Lipoprotein lipase (LPL), and transcription factor, SNAI3. Differential transcriptional regulation of LPL by pSTAT3 and SNAI3 was confirmed by promoter-reporter and chromatin immunoprecipitation analysis. Orthotopic xenograft murine breast tumor model revealed high tumorigenicity of CD24-/CD44+-breast CSCs as compared with CD24+-breast cancer cells. Furthermore, stable breast CSCs-CD44 shRNA and/or intratumoral administration of Tetrahydrolipstatin (LPL inhibitor) abrogated tumor progression and neoangiogenesis. Thus, LPL serves as a potential target for an efficacious therapeutics against aggressive breast cancer.
Collapse
Affiliation(s)
- Kanakaraju Manupati
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad, TS 500 007, India; Academy of Science and Innovative Research (AcSIR), Ghaziabad, UP 201 002, India
| | - Ragini Yeeravalli
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad, TS 500 007, India; Academy of Science and Innovative Research (AcSIR), Ghaziabad, UP 201 002, India
| | - Komal Kaushik
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad, TS 500 007, India; Academy of Science and Innovative Research (AcSIR), Ghaziabad, UP 201 002, India
| | - Digvijay Singh
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad, TS 500 007, India; Academy of Science and Innovative Research (AcSIR), Ghaziabad, UP 201 002, India
| | - Bhupendra Mehra
- Department of Surgery, Mahatma Gandhi Institute of Medical Sciences, Sewagram, Wardha, Maharashtra 442 102, India
| | - Nitin Gangane
- Department of Pathology, Mahatma Gandhi Institute of Medical Sciences, Sewagram, Wardha, Maharashtra 442 102, India
| | - Anupama Gupta
- Department of Pathology, Mahatma Gandhi Institute of Medical Sciences, Sewagram, Wardha, Maharashtra 442 102, India
| | - Kalyan Goswami
- Department of Biochemistry, Mahatma Gandhi Institute of Medical Sciences, Sewagram, Wardha, Maharashtra 442 102, India
| | - Amitava Das
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad, TS 500 007, India; Academy of Science and Innovative Research (AcSIR), Ghaziabad, UP 201 002, India.
| |
Collapse
|
16
|
Gago-Dominguez M, Redondo CM, Calaza M, Matabuena M, Bermudez MA, Perez-Fernandez R, Torres-Español M, Carracedo Á, Castelao JE. LIPG endothelial lipase and breast cancer risk by subtypes. Sci Rep 2021; 11:10436. [PMID: 34001944 PMCID: PMC8129130 DOI: 10.1038/s41598-021-89669-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 04/21/2021] [Indexed: 11/09/2022] Open
Abstract
Experimental data showed that endothelial lipase (LIPG) is a crucial player in breast cancer. However, very limited data exists on the role of LIPG on the risk of breast cancer in humans. We examined the LIPG-breast cancer association within our population-based case-control study from Galicia, Spain, BREOGAN (BREast Oncology GAlicia Network). Plasma LIPG and/or OxLDL were measured on 114 breast cancer cases and 82 controls from our case-control study, and were included in the present study. The risk of breast cancer increased with increasing levels of LIPG (multivariable OR for the highest category (95% CI) 2.52 (1.11-5.81), P-trend = 0.037). The LIPG-breast cancer association was restricted to Pre-menopausal breast cancer (Multivariable OR for the highest LIPG category (95% CI) 4.76 (0.94-28.77), P-trend = 0.06, and 1.79 (0.61-5.29), P-trend = 0.372, for Pre-menopausal and Post-menopausal breast cancer, respectively). The LIPG-breast cancer association was restricted to Luminal A breast cancers (Multivariable OR for the highest LIPG category (95% CI) 3.70 (1.42-10.16), P-trend = 0.015, and 2.05 (0.63-7.22), P-trend = 0.311, for Luminal A and non-Luminal A breast cancers, respectively). Subset analysis only based on HER2 receptor indicated that the LIPG-breast cancer relationship was restricted to HER2-negative breast cancers (Multivariable OR for the highest LIPG category (95% CI) 4.39 (1.70-12.03), P-trend = 0.012, and 1.10 (0.28-4.32), P-trend = 0.745, for HER2-negative and HER2-positive tumors, respectively). The LIPG-breast cancer association was restricted to women with high total cholesterol levels (Multivariable OR for the highest LIPG category (95% CI) 6.30 (2.13-20.05), P-trend = 0.018, and 0.65 (0.11-3.28), P-trend = 0.786, among women with high and low cholesterol levels, respectively). The LIPG-breast cancer association was also restricted to non-postpartum breast cancer (Multivariable OR for the highest LIPG category (95% CI) 3.83 (1.37-11.39), P-trend = 0.003, and 2.35 (0.16-63.65), P-trend = 0.396, for non-postpartum and postpartum breast cancer, respectively), although we lacked precision. The LIPG-breast cancer association was more pronounced among grades II and III than grade I breast cancers (Multivariable ORs for the highest category of LIPG (95% CI) 2.73 (1.02-7.69), P-trend = 0.057, and 1.90 (0.61-6.21), P-trend = 0.170, for grades II and III, and grade I breast cancers, respectively). No association was detected for OxLDL levels and breast cancer (Multivariable OR for the highest versus the lowest category (95% CI) 1.56 (0.56-4.32), P-trend = 0.457).
Collapse
Affiliation(s)
- Manuela Gago-Dominguez
- Galician Public Foundation of Genomic Medicine (FPGMX), Servicio Galego de Saúde (SERGAS), Santiago de Compostela, Spain.
- Genomic Medicine Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Centro en Red de Enfermedades Raras (CIBERER), University of Santiago de Compostela, Santiago de Compostela, Spain.
- Galician Public Foundation of Genomic Medicine (FPGMX), Genomic Medicine Group, International Cancer Genetics and Epidemiology Group, Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain.
| | - Carmen M Redondo
- Oncology and Genetics Unit, Instituto de Investigación Sanitaria Galicia Sur, Vigo, Spain
| | - Manuel Calaza
- Conselleria de Educación, Xunta de Galicia, Santiago de Compostela, Spain
| | - Marcos Matabuena
- Centro de Investigación en Tecnoloxías da Información (CiTIUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Maria A Bermudez
- Department of Biology, Faculty of Science, University of A Coruña, A Coruña, Spain
| | - Roman Perez-Fernandez
- Department of Physiology and Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - María Torres-Español
- Galician Public Foundation of Genomic Medicine (FPGMX), Servicio Galego de Saúde (SERGAS), Santiago de Compostela, Spain
- Genomic Medicine Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Centro en Red de Enfermedades Raras (CIBERER), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Ángel Carracedo
- Galician Public Foundation of Genomic Medicine (FPGMX), Servicio Galego de Saúde (SERGAS), Santiago de Compostela, Spain
- Genomic Medicine Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Centro en Red de Enfermedades Raras (CIBERER), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - J Esteban Castelao
- Oncology and Genetics Unit, Instituto de Investigación Sanitaria Galicia Sur, Vigo, Spain
| |
Collapse
|
17
|
Zhang ET, Hannibal RL, Badillo Rivera KM, Song JHT, McGowan K, Zhu X, Meinhardt G, Knöfler M, Pollheimer J, Urban AE, Folkins AK, Lyell DJ, Baker JC. PRG2 and AQPEP are misexpressed in fetal membranes in placenta previa and percreta†. Biol Reprod 2021; 105:244-257. [PMID: 33982062 DOI: 10.1093/biolre/ioab068] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 03/03/2021] [Accepted: 04/07/2021] [Indexed: 11/13/2022] Open
Abstract
The obstetrical conditions placenta accreta spectrum (PAS) and placenta previa are a significant source of pregnancy-associated morbidity and mortality, yet the specific molecular and cellular underpinnings of these conditions are not known. In this study, we identified misregulated gene expression patterns in tissues from placenta previa and percreta (the most extreme form of PAS) compared with control cases. By comparing this gene set with existing placental single-cell and bulk RNA-Seq datasets, we show that the upregulated genes predominantly mark extravillous trophoblasts. We performed immunofluorescence on several candidate molecules and found that PRG2 and AQPEP protein levels are upregulated in both the fetal membranes and the placental disk in both conditions. While this increased AQPEP expression remains restricted to trophoblasts, PRG2 is mislocalized and is found throughout the fetal membranes. Using a larger patient cohort with a diverse set of gestationally aged-matched controls, we validated PRG2 as a marker for both previa and PAS and AQPEP as a marker for only previa in the fetal membranes. Our findings suggest that the extraembryonic tissues surrounding the conceptus, including both the fetal membranes and the placental disk, harbor a signature of previa and PAS that is characteristic of EVTs and that may reflect increased trophoblast invasiveness.
Collapse
Affiliation(s)
- Elisa T Zhang
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Roberta L Hannibal
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Janet H T Song
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Kelly McGowan
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Xiaowei Zhu
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.,Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Gudrun Meinhardt
- Department of Obstetrics and Gynecology, Reproductive Biology Unit, Medical University of Vienna, Vienna, Austria
| | - Martin Knöfler
- Department of Obstetrics and Gynecology, Reproductive Biology Unit, Medical University of Vienna, Vienna, Austria
| | - Jürgen Pollheimer
- Department of Obstetrics and Gynecology, Reproductive Biology Unit, Medical University of Vienna, Vienna, Austria
| | - Alexander E Urban
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.,Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Ann K Folkins
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Deirdre J Lyell
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Julie C Baker
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.,Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
18
|
Egan G, Khan DH, Lee JB, Mirali S, Zhang L, Schimmer AD. Mitochondrial and Metabolic Pathways Regulate Nuclear Gene Expression to Control Differentiation, Stem Cell Function, and Immune Response in Leukemia. Cancer Discov 2021; 11:1052-1066. [PMID: 33504581 DOI: 10.1158/2159-8290.cd-20-1227] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/16/2020] [Accepted: 11/24/2020] [Indexed: 11/16/2022]
Abstract
Mitochondria are involved in many biological processes including cellular homeostasis, energy generation, and apoptosis. Moreover, mitochondrial and metabolic pathways are interconnected with gene expression to regulate cellular functions such as cell growth, survival, differentiation, and immune recognition. Metabolites and mitochondrial enzymes regulate chromatin-modifying enzymes, chromatin remodeling, and transcription regulators. Deregulation of mitochondrial pathways and metabolism leads to alterations in gene expression that promote cancer development, progression, and evasion of the immune system. This review highlights how mitochondrial and metabolic pathways function as a central mediator to control gene expression, specifically on stem cell functions, differentiation, and immune response in leukemia. SIGNIFICANCE: Emerging evidence demonstrates that mitochondrial and metabolic pathways influence gene expression to promote tumor development, progression, and immune evasion. These data highlight new areas of cancer biology and potential new therapeutic strategies.
Collapse
Affiliation(s)
- Grace Egan
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Dilshad H Khan
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Jong Bok Lee
- Toronto General Hospital Research Institute, Toronto, Ontario, Canada
| | - Sara Mirali
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Li Zhang
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.,Toronto General Hospital Research Institute, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathology, University of Toronto, Toronto, Ontario, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Aaron D Schimmer
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada. .,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
19
|
Kung CP, Cottrell KA, Ryu S, Bramel ER, Kladney RD, Bao EA, Freeman EC, Sabloak T, Maggi L, Weber JD. Evaluating the therapeutic potential of ADAR1 inhibition for triple-negative breast cancer. Oncogene 2021; 40:189-202. [PMID: 33110236 PMCID: PMC7796950 DOI: 10.1038/s41388-020-01515-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 09/28/2020] [Accepted: 10/06/2020] [Indexed: 11/09/2022]
Abstract
Triple-negative breast cancer (TNBC) is the deadliest form of breast cancer. Unlike other types of breast cancer that can be effectively treated by targeted therapies, no such targeted therapy exists for all TNBC patients. The ADAR1 enzyme carries out A-to-I editing of RNA to prevent sensing of endogenous double-stranded RNAs. ADAR1 is highly expressed in breast cancer including TNBC. Here, we demonstrate that expression of ADAR1, specifically its p150 isoform, is required for the survival of TNBC cell lines. In TNBC cells, knockdown of ADAR1 attenuates proliferation and tumorigenesis. Moreover, ADAR1 knockdown leads to robust translational repression. ADAR1-dependent TNBC cell lines also exhibit elevated IFN stimulated gene expression. IFNAR1 reduction significantly rescued the proliferative defects of ADAR1 loss. These findings establish ADAR1 as a novel therapeutic target for TNBC tumors.
Collapse
Affiliation(s)
- Che-Pei Kung
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Kyle A Cottrell
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Sua Ryu
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Emily R Bramel
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Raleigh D Kladney
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Emily A Bao
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Eric C Freeman
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Thwisha Sabloak
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Leonard Maggi
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jason D Weber
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Cell Biology and Physiology, Siteman Cancer Center, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
20
|
Ye H, Minhajuddin M, Krug A, Pei S, Chou CH, Culp-Hill R, Ponder J, De Bloois E, Schniedewind B, Amaya ML, Inguva A, Stevens BM, Pollyea DA, Christians U, Grimes HL, D'Alessandro A, Jordan CT. The Hepatic Microenvironment Uniquely Protects Leukemia Cells through Induction of Growth and Survival Pathways Mediated by LIPG. Cancer Discov 2020; 11:500-519. [PMID: 33028621 DOI: 10.1158/2159-8290.cd-20-0318] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 08/11/2020] [Accepted: 10/02/2020] [Indexed: 12/19/2022]
Abstract
Due to the disseminated nature of leukemia, malignant cells are exposed to many different tissue microenvironments, including a variety of extramedullary sites. In the present study, we demonstrate that leukemic cells residing in the liver display unique biological properties and also contribute to systemic changes that influence physiologic responses to chemotherapy. Specifically, the liver microenvironment induces metabolic adaptations via upregulating expression of endothelial lipase in leukemia cells, which not only stimulates tumor cell proliferation through polyunsaturated fatty acid-mediated pathways, but also promotes survival by stabilizing antiapoptotic proteins. Additionally, hepatic infiltration and tissue damage caused by malignant cells induces release of liver-derived enzymes capable of degrading chemotherapy drugs, an event that further protects leukemia cells from conventional therapies. Together, these studies demonstrate a unique role for liver in modulating the pathogenesis of leukemic disease and suggest that the hepatic microenvironment may protect leukemia cells from chemotherapeutic challenge. SIGNIFICANCE: The studies presented herein demonstrate that the liver provides a microenvironment in which leukemia cells acquire unique metabolic properties. The adaptations that occur in the liver confer increased resistance to chemotherapy. Therefore, we propose that therapies designed to overcome liver-specific metabolic changes will yield improved outcomes for patients with leukemia.This article is highlighted in the In This Issue feature, p. 211.
Collapse
Affiliation(s)
- Haobin Ye
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| | - Mohammad Minhajuddin
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Anna Krug
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Shanshan Pei
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Chih-Hsing Chou
- Division of Immunobiology, Cincinnati Children's Hospital, Cincinnati, Ohio
| | - Rachel Culp-Hill
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jessica Ponder
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Erik De Bloois
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Björn Schniedewind
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Maria L Amaya
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Anagha Inguva
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Brett M Stevens
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Daniel A Pollyea
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Uwe Christians
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - H Leighton Grimes
- Division of Immunobiology, Cincinnati Children's Hospital, Cincinnati, Ohio
| | - Angelo D'Alessandro
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado.,Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Craig T Jordan
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
21
|
Tecalco-Cruz AC, Ramírez-Jarquín JO, Cruz-Ramos E. Regulation and action of interferon-stimulated gene 15 in breast cancer cells. Hum Cell 2020; 33:954-962. [DOI: 10.1007/s13577-020-00414-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 08/09/2020] [Indexed: 12/24/2022]
|
22
|
Zhang Y, Zhu X, Qiao X, Gu X, Xue J, Han Y, Sun L, Cui M, Liu C. LIPH promotes metastasis by enriching stem-like cells in triple-negative breast cancer. J Cell Mol Med 2020; 24:9125-9134. [PMID: 32618099 PMCID: PMC7417731 DOI: 10.1111/jcmm.15549] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 05/05/2020] [Accepted: 06/07/2020] [Indexed: 11/30/2022] Open
Abstract
Lipase member H (LIPH), a novel member of the triglyceride lipase family. The clinical implications of its expression in breast cancer are still unclear. Therefore, in this study, we investigated the associations between LIPH and the tumorigenic behaviours of 144 triple-negative breast cancer (TNBC) patients. The ratio and mammosphere-forming ability of CD44+/CD24- stem-like cells were tested. The role of LIPH in breast cancer cell migration and invasion was also evaluated. In addition, the effect of LIPH silencing on mitochondrial respiration was determined using the Seahorse assay. Finally, the effect of LIPH silencing on protein expression was determined via tandem mass tag-based spectrometry and Western blotting. We found that LIPH expression was associated with metastasis in lymph nodes and distant organs (P = 0.025), resulting in poor survival among breast cancer patients (P = 0.027). LIPH knockdown significantly decreased both the ratio of CD44+ /CD24- stem-like cells and their mammosphere-forming ability. LIPH silencing promoted apoptosis, arrested cell cycle in the G2/M phase, mitigated the oxidation-related oxygen consumption rate in the mitochondria, and reduced metabolism. LIPH inhibited adhesion between tumour cells and enhanced the epithelial-mesenchymal transition. Tandem mass spectrometric analysis presented 68 proteins were differentially expressed in LIPH-silenced cells and LIPH-mediated modulation of tumour cell adhesion depended on integrin-related CAPN2 and paxillin signalling. Overall, our findings provided strong evidence that LIPH up-regulation promoted metastasis and the stemness of TNBC cells. Therefore, targeting LIPH is a potentially viable strategy for preventing metastasis in TNBC.
Collapse
Affiliation(s)
- Yixiao Zhang
- Cancer Stem Cell and Translational Medicine LaboratoryDepartment of Breast SurgeryShengjing Hospital of China Medical UniversityShenyangChina
| | - Xudong Zhu
- Cancer Stem Cell and Translational Medicine LaboratoryDepartment of Breast SurgeryShengjing Hospital of China Medical UniversityShenyangChina
| | - Xinbo Qiao
- Cancer Stem Cell and Translational Medicine LaboratoryDepartment of Breast SurgeryShengjing Hospital of China Medical UniversityShenyangChina
| | - Xi Gu
- Cancer Stem Cell and Translational Medicine LaboratoryDepartment of Breast SurgeryShengjing Hospital of China Medical UniversityShenyangChina
| | - Jinqi Xue
- Cancer Stem Cell and Translational Medicine LaboratoryDepartment of Breast SurgeryShengjing Hospital of China Medical UniversityShenyangChina
| | - Yanshuo Han
- Cancer Stem Cell and Translational Medicine LaboratoryDepartment of Breast SurgeryShengjing Hospital of China Medical UniversityShenyangChina
| | - Lisha Sun
- Cancer Stem Cell and Translational Medicine LaboratoryDepartment of Breast SurgeryShengjing Hospital of China Medical UniversityShenyangChina
| | - Meizi Cui
- Department of Cadre WardThe First Hospital of Jilin UniversityChangchunChina
| | - Caigang Liu
- Cancer Stem Cell and Translational Medicine LaboratoryDepartment of Breast SurgeryShengjing Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
23
|
Azimi A, Lo K, Kim J, Fernandez-Penas P. Investigating proteome changes between primary and metastatic cutaneous squamous cell carcinoma using SWATH mass spectrometry. J Dermatol Sci 2020; 99:119-127. [PMID: 32651104 DOI: 10.1016/j.jdermsci.2020.06.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/14/2020] [Accepted: 06/28/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Cutaneous squamous cell carcinoma (cSCC) is a common malignancy worldwide and the first as the cause of death from keratinocytic carcinomas. Around 5% of primary cSCCs metastasize, leading to a 5-year survival rate of only 11 %. OBJECTIVE This paper aims to investigate the proteome profile of primary and metastatic cSCC lesions for the identification of potential diagnostic biomarkers and molecular alterations. METHODS Liquid chromatography coupled with SWATH-MS workflow was used to analyse the proteome profile of formalin-fixed and paraffin-embedded samples of primary (n = 20) and metastatic cSCC (n = 25) lesions. Statistical and bioinformatics analysis was performed to identify differentially abundant proteins and molecular alterations between the lesions. RESULTS A total of 5037 proteins were identified across the samples of which 19 proteins including ISG15, APOA1 and MARCKS with roles in metastasis were increased and 11 proteins including DMKN, APCS and CST6 decreased in metastatic cSCC lesions relative to the primary phenotypes (adj. p-value <0.05). The proteomic data separated the lesions based on their histopathological diagnosis. Bioinformatics analysis revealed that cell migration, cell survival and immune response are likely activated, and apoptosis is inhibited in metastatic cSCC lesions, indicating increased lesion complexity as the disease progresses from primary to the metastatic phenotype. Two samples were reclassified after PCA analysis. CONCLUSION SWATH-MS analysis identified important molecular changes between primary and metastatic cSCC samples. Exploring these findings further will allow their translation into the clinic for improved tumour diagnosis, staging and therapeutic intervention.
Collapse
Affiliation(s)
- Ali Azimi
- Centre for Translational Skin Research, Faculty of Medicine and Health, The University of Sydney, Westmead, New South Wales, Australia; Department of Dermatology, Westmead Hospital, Westmead, New South Wales, Australia
| | - Kitty Lo
- School of Mathematics and Statistics, The University of Sydney, Camperdown, New South Wales, Australia
| | - Jennifer Kim
- Department of Tissue Pathology and Diagnostic Oncology, Institute of Clinical Pathology and Medical Research (ICPMR) Westmead Hospital, Westmead, New South Wales, Australia
| | - Pablo Fernandez-Penas
- Centre for Translational Skin Research, Faculty of Medicine and Health, The University of Sydney, Westmead, New South Wales, Australia; Department of Dermatology, Westmead Hospital, Westmead, New South Wales, Australia.
| |
Collapse
|
24
|
Ríos-Castro E, Souza GHMF, Delgadillo-Álvarez DM, Ramírez-Reyes L, Torres-Huerta AL, Velasco-Suárez A, Cruz-Cruz C, Hernández-Hernández JM, Tapia-Ramírez J. Quantitative Proteomic Analysis of MARC-145 Cells Infected with a Mexican Porcine Reproductive and Respiratory Syndrome Virus Strain Using a Label-Free Based DIA approach. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2020; 31:1302-1312. [PMID: 32379441 DOI: 10.1021/jasms.0c00134] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is an infectious disease characterized by severe reproductive failure in sows, acute respiratory disorders in growing pigs, and high mortality in piglets. The causative agent of this syndrome is the PRRS virus (PRRSV), an RNA virus belonging to the Arteriviridae family. To date, several quantitative approaches of proteomics have been applied to analyze the gene expression profiles during PRRSV infection in PAMs and MARC-145 cells, and few proteins have been consistent among independent studies, probably due to the differences in the levels of virulence of different PRRSV strains used and/or due to analytical conditions. In this study, total proteins isolated from noninfected and infected MARC-145 cells with a Mexican PRRSV strain were relatively quantified using label-free based DIA approach in combination with ion-mobility separation. As a result, 1456 quantified proteins were found to be shared between the control and infected samples. Afterward, these proteins were filtered, and 699 of them were considered without change. Also, 17 proteins were up-regulated and 19 proteins were down-regulated during the PRSSV infection. Bioinformatic analysis revealed that many of the differentially expressed proteins are involved in processes like antigen processing, presentation of antigens, response to viruses, response to IFNs, and innate immune response, among others. The present work is the first one which provides a detailed proteomic analysis through label-free based DIA approach in MARC-145 cells during the infection with a Mexican PRRSV strain.
Collapse
Affiliation(s)
- Emmanuel Ríos-Castro
- Unidad de Genómica, Proteómica y Metabolómica (UGPM), LaNSE, Cinvestav-IPN, Ciudad de México C.P. 07360, México
| | | | | | - Lorena Ramírez-Reyes
- Unidad de Genómica, Proteómica y Metabolómica (UGPM), LaNSE, Cinvestav-IPN, Ciudad de México C.P. 07360, México
| | - Ana Laura Torres-Huerta
- Unidad de Desarrollo e Innovación (UDI), LaNSE, Cinvestav-IPN, Ciudad de México, C.P. 07360, México
| | - Andrea Velasco-Suárez
- Unidad de Genómica, Proteómica y Metabolómica (UGPM), LaNSE, Cinvestav-IPN, Ciudad de México C.P. 07360, México
| | - Carlos Cruz-Cruz
- Departamento de Genética y Biologı́a Molecular, Cinvestav-IPN, Ciudad de México, C.P. 07360, México
| | | | - José Tapia-Ramírez
- Departamento de Genética y Biologı́a Molecular, Cinvestav-IPN, Ciudad de México, C.P. 07360, México
| |
Collapse
|
25
|
Inhibition of LIPG phospholipase activity suppresses tumor formation of human basal-like triple-negative breast cancer. Sci Rep 2020; 10:8911. [PMID: 32488004 PMCID: PMC7265491 DOI: 10.1038/s41598-020-65400-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 05/04/2020] [Indexed: 01/10/2023] Open
Abstract
The endothelial lipase LIPG possesses serine phospholipase activity and is involved in lipoprotein metabolism. Our previous studies have revealed that LIPG overexpression is required for tumor formation and metastasis of human basal-like triple-negative breast cancer (TNBC). We also demonstrated that LIPG differentially regulates TNBC malignancy through its enzymatic and non-enzymatic functions. The present studies were aimed at determining how XEN445, a specific inhibitor targeting LIPG phospholipase activity, impacts on TNBC tumor formation and malignant features. We established a cell-based LIPG enzymatic assay system to measure the inhibitory effect of XEN445 on LIPG phospholipase activity and determine its IC50. We found that XEN445 preferentially inhibited the proliferation of LIPG-expressing TNBC cells but not LIPG-negative luminal breast cancer cells. XEN445 inhibited the self-renewal of cancer stem cells (CSCs) in vitro and TNBC tumor formation in vivo. However, XEN445 had no inhibitory effect on the invasiveness and CSC stemness of TNBC cells. Our studies suggest that targeting both LIPG enzymatic and non-enzymatic functions is an important strategy for the treatment of TNBC.
Collapse
|
26
|
Hu H, Wang F, Wang M, Liu Y, Wu H, Chen X, Lin Q. FAM83A is amplified and promotes tumorigenicity in non-small cell lung cancer via ERK and PI3K/Akt/mTOR pathways. Int J Med Sci 2020; 17:807-814. [PMID: 32218702 PMCID: PMC7085261 DOI: 10.7150/ijms.33992] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 02/25/2020] [Indexed: 12/12/2022] Open
Abstract
Family with sequence similarity 83A (FAM83A) is a newly-found over-expressed oncogene in several types of cancers and associates with poor prognosis. However, the role that FAM83A may play in the carcinogenesis of non-small cell lung cancer (NSCLC) still needs to be defined. The present study aimed to investigate the function of FAM83A in NSCLC progression and to investigate the possible mechanism. Analysis of Gene Expression Omnibus (GEO) database and rt-PCR showed up-regulated expression of FAM83A in NSCLC. GEO and the Cancer Genome Atlas (TCGA) data analysis revealed that high expression level of FAM83A in NSCLC was associated with poor prognosis. In vitro experiments showed that depleting FAM83A by siRNA/shRNA significantly inhibited cell proliferation and induced cell apoptosis. Cell motility was also retarded after silencing FAM83A, as demonstrated by Transwell assay. FAM83A depletion in A549 cells also inhibited subcutaneous tumor growth and lung metastasis in vivo. Western blotting showed that silencing FAM83A decreased the phosphorylation of ERK and PI3K/Akt/mTOR. On the other hand, overexpressing FAM83A in vitro enhanced cell proliferation and invasiveness, which was repressed by PI3K inhibitor and ERK inhibitor separately. Taken together, our study suggests that FAM83A promotes tumorigenesis of NSCLC at least partly via ERK and PI3K/Akt/mTOR pathways, making it a promising therapeutic target.
Collapse
Affiliation(s)
- Haiyang Hu
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Hongkou District, Shanghai 200080, China
| | - Fajiu Wang
- Department of Thoracic Surgery, Hwa Mei Hospital, University of Chinese Academy of Sciences, No. 41 Xibei Road, Ningbo 315010, China
| | - Muyun Wang
- Department of Geriatric Respiratory and Critical Care, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, China
| | - Yuanyuan Liu
- Department of Otorhinolaryngology Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Hongkou District, Shanghai 200080, China
| | - Han Wu
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Hongkou District, Shanghai 200080, China
| | - Xi Chen
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Hongkou District, Shanghai 200080, China
| | - Qiang Lin
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Hongkou District, Shanghai 200080, China
| |
Collapse
|
27
|
Guo M, Sinha S, Wang SM. Coupled Genome-Wide DNA Methylation and Transcription Analysis Identified Rich Biomarkers and Drug Targets in Triple-Negative Breast Cancer. Cancers (Basel) 2019; 11:E1724. [PMID: 31690011 PMCID: PMC6896154 DOI: 10.3390/cancers11111724] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 10/30/2019] [Indexed: 02/02/2023] Open
Abstract
Triple-negative breast cancer (TNBC) has poor clinical prognosis. Lack of TNBC-specific biomarkers prevents active clinical intervention. We reasoned that TNBC must have its specific signature due to the lack of three key receptors to distinguish TNBC from other types of breast cancer. We also reasoned that coupling methylation and gene expression as a single unit may increase the specificity for the detected TNBC signatures. We further reasoned that choosing the proper controls may be critical to increasing the sensitivity to identify TNBC-specific signatures. Furthermore, we also considered that specific drugs could target the detected TNBC-specific signatures. We developed a system to identify potential TNBC signatures. It consisted of (1) coupling methylation and expression changes in TNBC to identify the methylation-regulated signature genes for TNBC; (2) using TPBC (triple-positive breast cancer) as the control to detect TNBC-specific signature genes; (3) searching in the drug database to identify those targeting TNBC signature genes. Using this system, we identified 114 genes with both altered methylation and expression, and 356 existing drugs targeting 10 of the 114 genes. Through docking and molecular dynamics simulation, we determined the structural basis between sapropterin, a drug used in the treatment of tetrahydrobiopterin deficiency, and PTGS2, a TNBC signature gene involved in the conversion of arachidonic acid to prostaglandins. Our study reveals the existence of rich TNBC-specific signatures, and many can be drug target and biomarker candidates for clinical applications.
Collapse
Affiliation(s)
- Maoni Guo
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau 999078, China.
| | - Siddharth Sinha
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau 999078, China.
| | - San Ming Wang
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau 999078, China.
| |
Collapse
|
28
|
Leonhardt G. LIPG supports adaption to oxidative stress. EXCLI JOURNAL 2019; 18:499-500. [PMID: 31423129 PMCID: PMC6694707 DOI: 10.17179/excli2019-1555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 06/26/2019] [Indexed: 11/18/2022]
Affiliation(s)
- Gregor Leonhardt
- Leibniz Research Centre for Working Environment and Human Factors
| |
Collapse
|
29
|
Cadenas C, Vosbeck S, Edlund K, Grgas K, Madjar K, Hellwig B, Adawy A, Glotzbach A, Stewart JD, Lesjak MS, Franckenstein D, Claus M, Hayen H, Schriewer A, Gianmoena K, Thaler S, Schmidt M, Micke P, Pontén F, Mardinoglu A, Zhang C, Käfferlein HU, Watzl C, Frank S, Rahnenführer J, Marchan R, Hengstler JG. LIPG-promoted lipid storage mediates adaptation to oxidative stress in breast cancer. Int J Cancer 2019; 145:901-915. [PMID: 30653260 PMCID: PMC6618071 DOI: 10.1002/ijc.32138] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 12/19/2018] [Indexed: 12/16/2022]
Abstract
Endothelial lipase (LIPG) is a cell surface associated lipase that displays phospholipase A1 activity towards phosphatidylcholine present in high‐density lipoproteins (HDL). LIPG was recently reported to be expressed in breast cancer and to support proliferation, tumourigenicity and metastasis. Here we show that severe oxidative stress leading to AMPK activation triggers LIPG upregulation, resulting in intracellular lipid droplet accumulation in breast cancer cells, which supports survival. Neutralizing oxidative stress abrogated LIPG upregulation and the concomitant lipid storage. In human breast cancer, high LIPG expression was observed in a limited subset of tumours and was significantly associated with shorter metastasis‐free survival in node‐negative, untreated patients. Moreover, expression of PLIN2 and TXNRD1 in these tumours indicated a link to lipid storage and oxidative stress. Altogether, our findings reveal a previously unrecognized role for LIPG in enabling oxidative stress‐induced lipid droplet accumulation in tumour cells that protects against oxidative stress, and thus supports tumour progression. What's new? Endothelial lipase (LIPG), a cell surface‐associated lipase with multifaceted roles, is expressed on breast cancer cells, but its molecular function and clinical relevance remain unknown. Here the authors uncover a link between oxidative stress and LIPG upregulation and show that high LIPG expression is associated with shorter metastasis‐free survival in women with node‐negative breast cancer. The authors speculate that LIPG may favor metastasis by enabling stress adaptation through lipid droplet formation and protection of mitochondria.
Collapse
Affiliation(s)
- Cristina Cadenas
- Department of Toxicology, Leibniz-Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Dortmund, Germany
| | - Sonja Vosbeck
- Department of Toxicology, Leibniz-Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Dortmund, Germany
| | - Karolina Edlund
- Department of Toxicology, Leibniz-Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Dortmund, Germany
| | - Katharina Grgas
- Department of Toxicology, Leibniz-Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Dortmund, Germany
| | - Katrin Madjar
- Department of Statistics, TU Dortmund University, Dortmund, Germany
| | - Birte Hellwig
- Department of Statistics, TU Dortmund University, Dortmund, Germany
| | - Alshaimaa Adawy
- Department of Toxicology, Leibniz-Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Dortmund, Germany
| | - Annika Glotzbach
- Department of Toxicology, Leibniz-Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Dortmund, Germany
| | - Joanna D Stewart
- Department of Toxicology, Leibniz-Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Dortmund, Germany
| | - Michaela S Lesjak
- Department of Toxicology, Leibniz-Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Dortmund, Germany
| | - Dennis Franckenstein
- Department of Toxicology, Leibniz-Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Dortmund, Germany
| | - Maren Claus
- Department of Immunology, Leibniz-Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Dortmund, Germany
| | - Heiko Hayen
- Department of Analytical Chemistry, Institute of Inorganic and Analytical Chemistry, University of Münster, Münster, Germany
| | - Alexander Schriewer
- Department of Analytical Chemistry, Institute of Inorganic and Analytical Chemistry, University of Münster, Münster, Germany
| | - Kathrin Gianmoena
- Department of Toxicology, Leibniz-Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Dortmund, Germany
| | - Sonja Thaler
- European Center for Angioscience (ECAS), Medical Faculty Mannheim of the University of Heidelberg, Tridomus C, Mannheim, Germany
| | - Marcus Schmidt
- Department of Obstetrics and Gynecology, University Hospital Mainz, Mainz, Germany
| | - Patrick Micke
- Department of Immunology Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Fredrik Pontén
- Department of Immunology Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Cheng Zhang
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Heiko U Käfferlein
- Center of Toxicology, Institute for Prevention and Occupational Medicine of the German Social Accident Insurance (IPA), Institute of the Ruhr University Bochum, Bochum, Germany
| | - Carsten Watzl
- Department of Toxicology, Leibniz-Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Dortmund, Germany
| | - Saša Frank
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | | | - Rosemarie Marchan
- Department of Toxicology, Leibniz-Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Dortmund, Germany
| | - Jan G Hengstler
- Department of Toxicology, Leibniz-Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Dortmund, Germany
| |
Collapse
|
30
|
Han HG, Moon HW, Jeon YJ. ISG15 in cancer: Beyond ubiquitin-like protein. Cancer Lett 2018; 438:52-62. [DOI: 10.1016/j.canlet.2018.09.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 09/06/2018] [Indexed: 01/08/2023]
|