1
|
Yuan J, Yin C, Peng H, Fang G, Mo B, Qin X, Chen Y, Wang Z, Yu Y, Wang Y, Wang Q. NDRG1 regulates iron metabolism and inhibits pathological cardiac hypertrophy. Can J Cardiol 2024:S0828-282X(24)01029-8. [PMID: 39427843 DOI: 10.1016/j.cjca.2024.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 09/26/2024] [Accepted: 10/14/2024] [Indexed: 10/22/2024] Open
Abstract
BACKGROUND Cardiac pathological hypertrophy, a pathological physiological alteration in many cardiovascular diseases, can progress to heart failure. The cellular biology underlying myocardial hypertrophy remains to be fully elucidated. While NDRG1 has been reported to participate in cellular proliferation, differentiation, and cellular stress responses, its role in cardiac diseases remains unexplored. Here, we investigated the role of NDRG1 in pathological hypertrophy. METHOD Cardiomyocyte-specific-NDRG1 knockout (KO) transgenic mice and NDRG1-AAV9 were used in mice. Angiotensin II (AngII) stimulation were applied to induce hypertrophy. Histological, molecular, and RNA-sequencing analyses were performed, and ferroptosis markers and iron levels were studied. We used co-IP and application of iron chelator to further studied the mechanisms of NDRG1 in cardiac hypertrophy. RESULTS We found that NDRG1 expression is decreased in pathological hypertrophy induced by AngII stimulation. Conditional knockout of NDRG1 in mouse cardiomyocytes led to progressive cardiac hypertrophy and heart failure. Cardiomyocyte-specific overexpression of NDRG1 via AAV9 significantly reversed AngII-induced ventricular hypertrophy and fibrosis. Mechanistically, NDRG1-deficient cardiomyocytes exhibited iron overload and increased ferroptosis, accompanied by elevated levels of reactive oxygen species (ROS) and lipid peroxidation. Subsequently, we confirmed the involvement of NDRG1 in regulating ferroptosis and iron metabolism in myocardial cells. Finally, we identified an interaction between NDRG1 and transferrin in cells. The iron chelator Dp44mT effectively reduced myocardial iron overload and ventricular remodeling induced by NDRG1 deficiency. CONCLUSION These findings highlight NDRG1's critical role in iron metabolism and ferroptosis in cardiomyocytes, suggesting that NDRG1 or iron metabolism may serve as therapeutic targets for cardiac hypertrophy.
Collapse
Affiliation(s)
- Jiali Yuan
- Department of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Chengye Yin
- Department of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Hong Peng
- Department of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Guojian Fang
- Department of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Binfeng Mo
- Department of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Xiji Qin
- Department of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Yuhan Chen
- Department of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Zhengshuai Wang
- Department of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Yichi Yu
- Department of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Yuepeng Wang
- Department of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China.
| | - Qunshan Wang
- Department of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China.
| |
Collapse
|
2
|
Jadhav SB, Vondrackova M, Potomova P, Sandoval-Acuña C, Smigova J, Klanicova K, Rosel D, Brabek J, Stursa J, Werner L, Truksa J. NDRG1 acts as an oncogene in triple-negative breast cancer and its loss sensitizes cells to mitochondrial iron chelation. Front Pharmacol 2024; 15:1422369. [PMID: 38983911 PMCID: PMC11231402 DOI: 10.3389/fphar.2024.1422369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 06/06/2024] [Indexed: 07/11/2024] Open
Abstract
Multiple studies indicate that iron chelators enhance their anti-cancer properties by inducing NDRG1, a known tumor and metastasis suppressor. However, the exact role of NDRG1 remains controversial, as newer studies have shown that NDRG1 can also act as an oncogene. Our group recently introduced mitochondrially targeted iron chelators deferoxamine (mitoDFO) and deferasirox (mitoDFX) as effective anti-cancer agents. In this study, we evaluated the ability of these modified chelators to induce NDRG1 and the role of NDRG1 in breast cancer. We demonstrated that both compounds specifically increase NDRG1 without inducing other NDRG family members. We have documented that the effect of mitochondrially targeted chelators is at least partially mediated by GSK3α/β, leading to phosphorylation of NDRG1 at Thr346 and to a lesser extent on Ser330. Loss of NDRG1 increases cell death induced by mitoDFX. Notably, MDA-MB-231 cells lacking NDRG1 exhibit reduced extracellular acidification rate and grow slower than parental cells, while the opposite is true for ER+ MCF7 cells. Moreover, overexpression of full-length NDRG1 and the N-terminally truncated isoform (59112) significantly reduced sensitivity towards mitoDFX in ER+ cells. Furthermore, cells overexpressing full-length NDRG1 exhibited a significantly accelerated tumor formation, while its N-terminally truncated isoforms showed significantly impaired capacity to form tumors. Thus, overexpression of full-length NDRG1 promotes tumor growth in highly aggressive triple-negative breast cancer.
Collapse
Affiliation(s)
- Sukanya B. Jadhav
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Centre, Vestec, Czechia
- Faculty of Sciences, Charles University, Prague, Czechia
| | - Michaela Vondrackova
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Centre, Vestec, Czechia
- Faculty of Sciences, Charles University, Prague, Czechia
- Faculty of Sciences, BIOCEV Research Centre, Charles University, Vestec, Czechia
| | - Petra Potomova
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Centre, Vestec, Czechia
- Faculty of Sciences, Charles University, Prague, Czechia
| | - Cristian Sandoval-Acuña
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Centre, Vestec, Czechia
| | - Jana Smigova
- Faculty of Sciences, BIOCEV Research Centre, Charles University, Vestec, Czechia
| | - Kristyna Klanicova
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Centre, Vestec, Czechia
| | - Daniel Rosel
- Faculty of Sciences, Charles University, Prague, Czechia
- Faculty of Sciences, BIOCEV Research Centre, Charles University, Vestec, Czechia
| | - Jan Brabek
- Faculty of Sciences, Charles University, Prague, Czechia
- Faculty of Sciences, BIOCEV Research Centre, Charles University, Vestec, Czechia
| | - Jan Stursa
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Centre, Vestec, Czechia
| | - Lukas Werner
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Centre, Vestec, Czechia
| | - Jaroslav Truksa
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Centre, Vestec, Czechia
| |
Collapse
|
3
|
Yang YP, Bai M, Cheng YX, Feng X, Zhang YY, Zhang YY, Liu MY, Duan YQ. Based on the prognosis model of immunogenes, the prognosis model was constructed to predict the invasion of immune genes and immune cells related to primary liver cancer and its experimental validation. Heliyon 2024; 10:e27362. [PMID: 38560168 PMCID: PMC10980948 DOI: 10.1016/j.heliyon.2024.e27362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 02/28/2024] [Accepted: 02/28/2024] [Indexed: 04/04/2024] Open
Abstract
Background Primary liver cancer (PLC) is a prevalent malignancy of the digestive system characterized by insidious symptom onset and a generally poor prognosis. Recent studies have highlighted a significant correlation between the initiation and prognosis of liver cancer and the immune function of PLC patients. Purpose Revealing the expression of PLC-related immune genes and the characteristics of immune cell infiltration provides assistance for the analysis of clinical pathological parameters and prognosis of PLC patients. Methods PLC-related differentially expressed genes (DEGs) with a median absolute deviation (MAD > 0.5) were identified from TCGA and GEO databases. These DEGs were intersected with immune-related genes (IRGs) from the ImmPort database to obtain PLC-related IRGs. The method of constructing a prognostic model through immune-related gene pairs (IRGPs) is used to obtain IRGPs and conduct the selection of central immune genes. The central immune genes obtained from the selection of IRGPs are validated in PLC. Subsequently, the relative proportions of 22 types of immune cells in different immune risk groups are evaluated, and the differential characteristics of PLC-related immune cells are verified through animal experiments. Results Through database screening and the construction of an IRGP prognosis model, 84 pairs of IRGPs (P < 0.001) were ultimately obtained. Analysis of these 84 IRGPs revealed 11 central immune genes related to PLC, showing differential expression in liver cancer tissues compared to normal liver tissues. Results from the CiberSort platform indicate differential expression of immune cells such as naive B cells, macrophages, and neutrophils in different immune risk groups. Animal experiments demonstrated altered immune cell proportions in H22 tumor-bearing mice, validating findings from peripheral blood and spleen homogenate analyses. Conclusion Our study successfully predicted and validated PLC-related IRGs and immune cells, suggesting their potential as prognostic indicators and therapeutic targets for PLC.
Collapse
Affiliation(s)
- Yu-Ping Yang
- Gansu University of Traditional Chinese Medicine, College of Basic Medical Sciences, Lanzhou, 730000, PR China
| | - Min Bai
- Gansu University of Traditional Chinese Medicine, College of Basic Medical Sciences, Lanzhou, 730000, PR China
| | - Yin-Xia Cheng
- Ningxia Medical University, College of Traditional Chinese Medicine, Yinchuan, 750000, PR China
| | - Xin Feng
- Gansu University of Traditional Chinese Medicine, College of Basic Medical Sciences, Lanzhou, 730000, PR China
| | - Yan-Ying Zhang
- Gansu University of Traditional Chinese Medicine, College of Basic Medical Sciences, Lanzhou, 730000, PR China
| | - Yuan-Yuan Zhang
- Gansu University of Traditional Chinese Medicine, College of Basic Medical Sciences, Lanzhou, 730000, PR China
| | - Meng-Ya Liu
- Gansu University of Traditional Chinese Medicine, College of Basic Medical Sciences, Lanzhou, 730000, PR China
| | - Yong-Qiang Duan
- Ningxia Medical University, College of Traditional Chinese Medicine, Yinchuan, 750000, PR China
| |
Collapse
|
4
|
Villodre ES, Nguyen APN, Debeb BG. NDRGs in Breast Cancer: A Review and In Silico Analysis. Cancers (Basel) 2024; 16:1342. [PMID: 38611020 PMCID: PMC11011033 DOI: 10.3390/cancers16071342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
The N-myc downstream regulated gene family (NDRGs) includes four members: NDRG1, NDRG2, NDRG3, and NDRG4. These members exhibit 53-65% amino acid identity. The role of NDRGs in tumor growth and metastasis appears to be tumor- and context-dependent. While many studies have reported that these family members have tumor suppressive roles, recent studies have demonstrated that NDRGs, particularly NDRG1 and NDRG2, function as oncogenes, promoting tumor growth and metastasis. Additionally, NDRGs are involved in regulating different signaling pathways and exhibit diverse cellular functions in breast cancers. In this review, we comprehensively outline the oncogenic and tumor suppressor roles of the NDRG family members in breast cancer, examining evidence from in vitro and in vivo breast cancer models as well as tumor tissues from breast cancer patients. We also present analyses of publicly available genomic and transcriptomic data from multiple independent cohorts of breast cancer patients.
Collapse
Affiliation(s)
- Emilly S. Villodre
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (E.S.V.); (A.P.N.N.)
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research Program, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Anh P. N. Nguyen
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (E.S.V.); (A.P.N.N.)
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research Program, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Bisrat G. Debeb
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (E.S.V.); (A.P.N.N.)
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research Program, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
5
|
Xu D, Hu Z, Wang K, Hu S, Zhou Y, Zhang S, Chen Y, Pan T. Why does HER2-positive breast cancer metastasize to the brain and what can we do about it? Crit Rev Oncol Hematol 2024; 195:104269. [PMID: 38272149 DOI: 10.1016/j.critrevonc.2024.104269] [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: 04/07/2023] [Revised: 12/18/2023] [Accepted: 01/19/2024] [Indexed: 01/27/2024] Open
Abstract
Breast cancer is the most frequent malignancy in women. However, in the middle and late stages, some people develop distant metastases, which considerably lower the quality of life and life expectancy. The brain is one of the sites where metastasis frequently happens. According to epidemiological research, brain metastases occur at a late stage in 30-50% of patients with HER2-positive breast cancer, resulting in a poor prognosis. Additionally, few treatments are available for HER2-positive brain metastatic breast cancer, and the mortality rate is remarkable owing to the complexity of the brain's anatomical structure and physiological function. In this review, we described the stages of the brain metastasis of breast cancer, the relationship between the microenvironment and metastatic cancer cells, and the unique molecular and cellular mechanisms. It involves cancer cells migrating, invading, and adhering to the brain; penetrating the blood-brain barrier; interacting with brain cells; and activating signal pathways once inside the brain. Finally, we reviewed current clinically used treatment approaches for brain metastasis in HER2-positive breast cancer; summarized the traditional treatment, targeted treatment, immunotherapy, and other treatment modalities; compared the benefits and drawbacks of each approach; discussed treatment challenges; and emphasized the importance of identifying potential targets to improve patient survival rates and comprehend brain metastasis in breast cancer.
Collapse
Affiliation(s)
- Dongyan Xu
- Department of Breast Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Zhengfang Hu
- Beijing Tian Tan Hospital, Capital Medical University, Beijing 100050, China
| | - Kaiyue Wang
- Department of Breast Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Shiyao Hu
- Department of Breast Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Yunxiang Zhou
- Department of Breast Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Shizhen Zhang
- Department of Breast Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Yiding Chen
- Department of Breast Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Tao Pan
- Department of Breast Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China.
| |
Collapse
|
6
|
Liu X, Liu S, Yang Y, Cai H, Zheng R, Zhang Y, Li X, Fan F, Liu H, Li S. Animal models of brain and spinal cord metastases of NSCLC established using a brain stereotactic instrument. Heliyon 2024; 10:e24809. [PMID: 38318004 PMCID: PMC10838758 DOI: 10.1016/j.heliyon.2024.e24809] [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: 06/17/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 02/07/2024] Open
Abstract
Objective Animal models of brain and spinal cord metastases of non-small cell lung cancer were established through the intracranial injection of PC-9 Luc cells with a brain stereotaxic device. This method provides a reliable modeling method for studying brain and spinal cord metastases of non-small cell lung cancer. Methods PC-9 Luc cells at logarithmic growth stage were injected into the skulls of 5-week-old BALB/c nude mice at different cell volumes (30 × 104, 80 × 104) and different locations (using anterior fontanel as a location point, 1 mm from the coronal suture, and 1.5 mm from the sagittal suture on the right upper and right lower side of the skull). After 1 week of cell inoculation, fluorescence signals of tumor cells in the brain and spinal were detected using the IVIS Xenogen Imaging system. After 4 weeks, brain and spinal tissues from the nude mice were harvested. Following paraffin-embedded sectioning, HE staining was performed on the tissues. Results The fluorescence signals revealed that both brain and spinal cord metastasis occurred in the mice where the cells were injected at the lower right side of the skull. There was only brain metastasis in the nude mice injected with 30 × 104 cells at the upper right side of the skull. Both brain and spinal cord metastasis occurred in the nude mice injected with 80 × 104 cells. The HE staining revealed that both brain and spinal cord metastasis occurred in the mice injected with different amounts of PC-9 Luc cells, consistent with the results detected using the IVIS Xenogen Imaging system, thereby demonstrating the reliability of detecting fluorescent signals in vivo to determine tumor growth. Conclusion It is a reliable method to establish the animal model of brain and spinal cord metastases of non-small cell lung cancer by injecting different quantities of cells from different positions with a brain stereotaxic device. The IVIS Xenogen Imaging system has high reliability in detecting the fluorescence signals of brain and spinal cord metastatic tumors.
Collapse
Affiliation(s)
- Xuerou Liu
- School of Pharmacy, Bengbu Medical University, Bengbu, China
| | - Shiyao Liu
- School of Pharmacy, Bengbu Medical University, Bengbu, China
| | - Yumei Yang
- School of Pharmacy, Bengbu Medical University, Bengbu, China
| | - Hui Cai
- School of Pharmacy, Bengbu Medical University, Bengbu, China
| | - Ruijie Zheng
- School of Pharmacy, Bengbu Medical University, Bengbu, China
| | - Yaoshuai Zhang
- School of Pharmacy, Bengbu Medical University, Bengbu, China
| | - Xian Li
- School of Pharmacy, Bengbu Medical University, Bengbu, China
- Anhui Province Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, China
| | - Fangtian Fan
- School of Pharmacy, Bengbu Medical University, Bengbu, China
- Anhui Province Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, China
| | - Hao Liu
- School of Pharmacy, Bengbu Medical University, Bengbu, China
- Anhui Province Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, China
| | - Shanshan Li
- School of Pharmacy, Bengbu Medical University, Bengbu, China
- Anhui Province Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, China
| |
Collapse
|
7
|
Iwase T, Wang X, Thi Hanh Phi L, Sridhar N, Ueno NT, Lee J. Advances in targets in inflammatory breast cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 384:125-152. [PMID: 38637096 DOI: 10.1016/bs.ircmb.2023.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Affiliation(s)
- Toshiaki Iwase
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; University of Hawaii Cancer Center, Honolulu, HI, United States.
| | - Xiaoping Wang
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Lan Thi Hanh Phi
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nithya Sridhar
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Naoto T Ueno
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; University of Hawaii Cancer Center, Honolulu, HI, United States
| | - Jangsoon Lee
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
8
|
Bin Y, Wei S, Chen R, Zhang H, Ren J, Liu P, Xin Z, Zhang T, Yang H, Wang K, Feng Z, Sun X, Chen Z, Zhang H. Dclre1c-Mutation-Induced Immunocompromised Mice Are a Novel Model for Human Xenograft Research. Biomolecules 2024; 14:180. [PMID: 38397417 PMCID: PMC10887050 DOI: 10.3390/biom14020180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/20/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Severe combined immunodeficient (SCID) mice serve as a critical model for human xenotransplantation studies, yet they often suffer from low engraftment rates and susceptibility to graft-versus-host disease (GVHD). Moreover, certain SCID strains demonstrate 'immune leakage', underscoring the need for novel model development. Here, we introduce an SCID mouse model with a targeted disruption of the dclre1c gene, encoding Artemis, which is essential for V(D)J recombination and DNA repair during T cell receptor (TCR) and B cell receptor (BCR) assembly. Artemis deficiency precipitates a profound immunodeficiency syndrome, marked by radiosensitivity and compromised T and B lymphocyte functionality. Utilizing CRISPR/Cas9-mediated gene editing, we generated dclre1c-deficient mice with an NOD genetic background. These mice exhibited a radiosensitive SCID phenotype, with pronounced DNA damage and defective thymic, splenic and lymph node development, culminating in reduced T and B lymphocyte populations. Notably, both cell lines and patient-derived tumor xenografts were successfully engrafted into these mice. Furthermore, the human immune system was effectively rebuilt following peripheral blood mononuclear cells (PBMCs) transplantation. The dclre1c-knockout NOD mice described herein represent a promising addition to the armamentarium of models for xenotransplantation, offering a valuable platform for advancing human immunobiological research.
Collapse
Affiliation(s)
- Yixiao Bin
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an 710032, China; (Y.B.); (R.C.); (J.R.); (P.L.); (Z.X.); (T.Z.); (H.Y.); (K.W.); (Z.F.); (X.S.)
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi’an 710032, China
- School of Basic Medical Sciences, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Sanhua Wei
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, Tang Du Hospital, Fourth Military Medical University, Xi’an 710038, China;
| | - Ruo Chen
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an 710032, China; (Y.B.); (R.C.); (J.R.); (P.L.); (Z.X.); (T.Z.); (H.Y.); (K.W.); (Z.F.); (X.S.)
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi’an 710032, China
| | - Haowei Zhang
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi’an 710032, China;
| | - Jing Ren
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an 710032, China; (Y.B.); (R.C.); (J.R.); (P.L.); (Z.X.); (T.Z.); (H.Y.); (K.W.); (Z.F.); (X.S.)
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi’an 710032, China
- School of Basic Medical Sciences, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Peijuan Liu
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an 710032, China; (Y.B.); (R.C.); (J.R.); (P.L.); (Z.X.); (T.Z.); (H.Y.); (K.W.); (Z.F.); (X.S.)
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi’an 710032, China
| | - Zhiqian Xin
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an 710032, China; (Y.B.); (R.C.); (J.R.); (P.L.); (Z.X.); (T.Z.); (H.Y.); (K.W.); (Z.F.); (X.S.)
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi’an 710032, China
| | - Tianjiao Zhang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an 710032, China; (Y.B.); (R.C.); (J.R.); (P.L.); (Z.X.); (T.Z.); (H.Y.); (K.W.); (Z.F.); (X.S.)
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi’an 710032, China
| | - Haijiao Yang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an 710032, China; (Y.B.); (R.C.); (J.R.); (P.L.); (Z.X.); (T.Z.); (H.Y.); (K.W.); (Z.F.); (X.S.)
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi’an 710032, China
| | - Ke Wang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an 710032, China; (Y.B.); (R.C.); (J.R.); (P.L.); (Z.X.); (T.Z.); (H.Y.); (K.W.); (Z.F.); (X.S.)
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi’an 710032, China
| | - Zhuan Feng
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an 710032, China; (Y.B.); (R.C.); (J.R.); (P.L.); (Z.X.); (T.Z.); (H.Y.); (K.W.); (Z.F.); (X.S.)
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi’an 710032, China
| | - Xiuxuan Sun
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an 710032, China; (Y.B.); (R.C.); (J.R.); (P.L.); (Z.X.); (T.Z.); (H.Y.); (K.W.); (Z.F.); (X.S.)
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi’an 710032, China
| | - Zhinan Chen
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an 710032, China; (Y.B.); (R.C.); (J.R.); (P.L.); (Z.X.); (T.Z.); (H.Y.); (K.W.); (Z.F.); (X.S.)
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi’an 710032, China
| | - Hai Zhang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an 710032, China; (Y.B.); (R.C.); (J.R.); (P.L.); (Z.X.); (T.Z.); (H.Y.); (K.W.); (Z.F.); (X.S.)
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi’an 710032, China
| |
Collapse
|
9
|
Fares J, Petrosyan E, Kanojia D, Dmello C, Cordero A, Duffy JT, Yeeravalli R, Sahani MH, Zhang P, Rashidi A, Arrieta VA, Ulasov I, Ahmed AU, Miska J, Balyasnikova IV, James CD, Sonabend AM, Heimberger AB, Lesniak MS. Metixene is an incomplete autophagy inducer in preclinical models of metastatic cancer and brain metastases. J Clin Invest 2023; 133:e161142. [PMID: 37847564 PMCID: PMC10721147 DOI: 10.1172/jci161142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 10/12/2023] [Indexed: 10/18/2023] Open
Abstract
A paucity of chemotherapeutic options for metastatic brain cancer limits patient survival and portends poor clinical outcomes. Using a CNS small-molecule inhibitor library of 320 agents known to be blood-brain barrier permeable and approved by the FDA, we interrogated breast cancer brain metastasis vulnerabilities to identify an effective agent. Metixene, an antiparkinsonian drug, was identified as a top therapeutic agent that was capable of decreasing cellular viability and inducing cell death across different metastatic breast cancer subtypes. This agent significantly reduced mammary tumor size in orthotopic xenograft assays and improved survival in an intracardiac model of multiorgan site metastases. Metixene further extended survival in mice bearing intracranial xenografts and in an intracarotid mouse model of multiple brain metastases. Functional analysis revealed that metixene induced incomplete autophagy through N-Myc downstream regulated 1 (NDRG1) phosphorylation, thereby leading to caspase-mediated apoptosis in both primary and brain-metastatic cells, regardless of cancer subtype or origin. CRISPR/Cas9 KO of NDRG1 led to autophagy completion and reversal of the metixene apoptotic effect. Metixene is a promising therapeutic agent against metastatic brain cancer, with minimal reported side effects in humans, which merits consideration for clinical translation.
Collapse
Affiliation(s)
- Jawad Fares
- Department of Neurological Surgery, and
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Edgar Petrosyan
- Department of Neurological Surgery, and
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Deepak Kanojia
- Department of Neurological Surgery, and
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Crismita Dmello
- Department of Neurological Surgery, and
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Alex Cordero
- Department of Neurological Surgery, and
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Joseph T. Duffy
- Department of Neurological Surgery, and
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ragini Yeeravalli
- Department of Neurological Surgery, and
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Mayurbhai H. Sahani
- Dr. Vikram Sarabhai Institute of Cell and Molecular Biology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Peng Zhang
- Department of Neurological Surgery, and
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Aida Rashidi
- Department of Neurological Surgery, and
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Victor A. Arrieta
- Department of Neurological Surgery, and
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ilya Ulasov
- Department of Neurological Surgery, and
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Atique U. Ahmed
- Department of Neurological Surgery, and
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Jason Miska
- Department of Neurological Surgery, and
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Irina V. Balyasnikova
- Department of Neurological Surgery, and
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - C. David James
- Department of Neurological Surgery, and
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Adam M. Sonabend
- Department of Neurological Surgery, and
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Amy B. Heimberger
- Department of Neurological Surgery, and
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Maciej S. Lesniak
- Department of Neurological Surgery, and
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
10
|
He Y, Wang X. Identifying biomarkers associated with immunotherapy response in melanoma by multi-omics analysis. Comput Biol Med 2023; 167:107591. [PMID: 37875043 DOI: 10.1016/j.compbiomed.2023.107591] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/26/2023] [Accepted: 10/17/2023] [Indexed: 10/26/2023]
Abstract
Despite immune checkpoint inhibitors (ICIs) have shown the greatest success in melanoma treatment, only a subset of melanoma patients responds well to ICIs. Thus, identifying predictive biomarkers for immunotherapy response is crucial. In this study, we took complementary advantages of immunotherapy data and The Cancer Genome Atlas (TCGA) multi-omics data to explore the predictive biomarkers for the response to immunotherapy in melanoma. We first predicted responsive and non-responsive melanomas in the TCGA skin cutaneous melanoma (SKCM) cohort based on both somatic mutation and transcriptome datasets which involved immunotherapy data for melanoma. This method identified 170 responsive and 56 non-responsive melanomas in TCGA-SKCM. Based on the TCGA-SKCM data, we performed a comprehensive comparison of multi-omics molecular features between responsive and non-responsive melanomas. We identified the molecular features significantly associated with immunotherapy response in melanoma at the genome, transcriptome, epigenome, and proteome levels, respectively. Our analysis confirmed certain immunotherapy response-associated biomarkers, such as tumor mutation burden (TMB), copy number alteration (CNA), intratumor heterogeneity (ITH), PD-L1 expression, and tumor immunity. Moreover, we identified some novel molecular features associated with immunotherapy response: (1) the activation of mast cells and dendritic cells correlating negatively with immunotherapy response; (2) the enrichment of many oncogenic pathways correlating positively with immunotherapy response, such as JAK-STAT, RAS, MAPK, HIF-1, PI3K-Akt, and VEGF pathways; and (3) a number of microRNAs and proteins whose expression correlates with immunotherapy response. In addition, the mTOR signaling pathway has a negative association with immunotherapy response. The novel biomarkers have potential predictive values in immunotherapy response and warrant further investigation.
Collapse
Affiliation(s)
- Yin He
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China; Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China; Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiaosheng Wang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China; Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China; Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
11
|
Xu L, Wang S, Zhang D, Wu Y, Shan J, Zhu H, Wang C, Wang Q. Machine learning- and WGCNA-mediated double analysis based on genes associated with disulfidptosis, cuproptosis and ferroptosis for the construction and validation of the prognostic model for breast cancer. J Cancer Res Clin Oncol 2023; 149:16511-16523. [PMID: 37712959 DOI: 10.1007/s00432-023-05378-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 08/30/2023] [Indexed: 09/16/2023]
Abstract
BACKGROUND Disulfidptosis, a recently discovered cellular death mechanism, has not been extensively studied in relation to breast cancer (BC). Specifically, no previous research has integrated disulfidptosis-related genes (DRGs), cuproptosis-related genes (CRGs), and ferroptosis-related genes (FRGs) to construct a prognostic signature for BC. METHODS DRGs, CRGs and FRGs with prognostic potential were identified through Cox regression analysis. A predictive model was constructed by intersecting the core genes obtained from unsupervised cluster analysis and weighted correlation network analysis (WGCNA). Differences in chemotherapy drug sensitivity, immune checkpoint levels were analyzed according to different risk score groups. The expression of the core disulfidptosis gene, SLC7A11, was analyzed using immunofluorescence. RESULTS Single-cell RNA sequencing analysis revealed differential expression of DRGs in the BC tumor microenvironment. We developed a prognostic model, consisting of six genes, based on machine learning which included unsupervised cluster analysis and Lasso-Cox analysis. An internal training set and a validation set, both derived from the Cancer Genome Atlas-Breast Cancer (TCGA-BRCA) database, GSE20685 and GSE42568 as external validation sets all verified the model's validity. The low-risk group exhibited increased sensitivity to paclitaxel. Additionally, the high-risk group demonstrated significantly higher expression of tumor mutation burden and microsatellite instability compared to the low-risk group. A nomogram confirmed that the risk score can be an independent risk factor for BC. Notably, our findings highlighted the impact of SLC7A11 on the BC tumor microenvironment. Immunofluorescence analysis revealed significantly higher expression of SLC7A11 in BC tissues compared to paracancerous tissues. CONCLUSION Multiplex analysis based on DRGs, CRGs and FRGs correlated strongly with BC, providing new insights for developing clinical prognostic tools and designing immunotherapy regimens for BC patients.
Collapse
Affiliation(s)
- Lijun Xu
- Department of General Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Shanshan Wang
- Department of General Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Dan Zhang
- Department of General Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Yunxi Wu
- Department of General Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Jiali Shan
- Department of General Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Huixia Zhu
- Department of Biochemistry, Medical College, Nantong University, Nantong, 226001, China
| | - Chongyu Wang
- Department of Medicine, Xinglin College, Nantong University, Nantong, 226007, China
| | - Qingqing Wang
- Department of General Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China.
| |
Collapse
|
12
|
Kotepui K, Kotepui M, Majima HJ, Tangpong J. Association between NDRG1 protein expression and aggressive features of breast cancer: a systematic review and meta-analysis. BMC Cancer 2023; 23:1003. [PMID: 37858101 PMCID: PMC10585795 DOI: 10.1186/s12885-023-11517-7] [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: 05/30/2023] [Accepted: 10/11/2023] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND N-myc downstream-regulated gene-1 (NDRG1) is well-described as a potent metastasis suppressor, but its role in human breast cancer remains controversial and unclear. Therefore, the present study utilized a systematic review and meta-analysis approach to synthesize the association between NDRG1 protein expression and the aggressive characteristics of breast cancer. METHODS The protocol for the systematic review and meta-analysis was registered on the PROSPERO website (CRD42023414814). Relevant articles were searched for in PubMed, Scopus, Embase, MEDLINE, and Ovid between March 30, 2023, and May 5, 2023. The included studies were critically evaluated using the Joanna Briggs Institute critical appraisal tools. The results from individual studies were qualitatively synthesized using textual narrative synthesis. Using a random-effects model, the pooled log odds ratio of effect estimate was used to look at the link between NDRG1 protein expression and aggressive features of breast cancer, such as tumor grade, tumor stage, metastasis to the axillary lymph nodes, and hormonal receptor status. RESULTS A total of 1423 articles were retrieved from the electronic database search, and six studies that met the eligibility criteria were included for synthesis. There was an association between the expression of NDRG1 protein and the status of the axillary lymph nodes (P = 0.01, log Odds Ratio (OR): 0.59, 95% Confidence Interval (CI): 0.13-1.05, I2: 24.24%, 292 breast cancer cases with positive axillary lymph nodes and 229 breast cancer cases with negative axillary lymph nodes, 4 studies). NDRG1 protein expression and human epidermal growth factor receptor 2 (Her2) status were found to have a negative relationship (P = 0.01, log OR: -0.76, 95% CI: -1.32-(-0.20), I2: 32.42%, 197 breast cancer cases with Her2 positive and 272 breast cancer cases with Her2 negative, 3 studies). No correlation was found between NDRG1 protein expression and tumor grade (P = 0.10), estrogen receptor (ER) status (P = 0.57), or progesterone receptor (PR) status (P = 0.41). CONCLUSION The study concluded that increased NDRG1 protein expression was associated with increased metastasis of the tumor to the axillary lymph node. Additionally, increased NDRG1 protein expression was observed in Her2-negative breast cancer, suggesting its role in both less aggressive and more aggressive behavior depending on breast cancer subtypes. Based on the findings of the meta-analysis, an increase in NDRG1 protein expression was associated with aggressive characteristics of breast cancer.
Collapse
Affiliation(s)
- Kwuntida Kotepui
- Medical Technology, School of Allied Health Sciences, Walailak University, Tha Sala, Nakhon Si Thammarat, Thailand
| | - Manas Kotepui
- Medical Technology, School of Allied Health Sciences, Walailak University, Tha Sala, Nakhon Si Thammarat, Thailand.
| | - Hideyuki J Majima
- Medical Technology, School of Allied Health Sciences, Walailak University, Tha Sala, Nakhon Si Thammarat, Thailand
| | - Jitbanjong Tangpong
- Medical Technology, School of Allied Health Sciences, Walailak University, Tha Sala, Nakhon Si Thammarat, Thailand
| |
Collapse
|
13
|
Wang YY, Zhou YQ, Xie JX, Zhang X, Wang SC, Li Q, Hu LP, Jiang SH, Yi SQ, Xu J, Cao H, Zhao EH, Li J. MAOA suppresses the growth of gastric cancer by interacting with NDRG1 and regulating the Warburg effect through the PI3K/AKT/mTOR pathway. Cell Oncol (Dordr) 2023; 46:1429-1444. [PMID: 37249744 DOI: 10.1007/s13402-023-00821-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2023] [Indexed: 05/31/2023] Open
Abstract
OBJECTIVE Previous studies have indicated that neurotransmitters play important roles in the occurrence and development of gastric cancer. MAOA is an important catecholamine neurotransmitter-degrading enzyme involved in the degradation of norepinephrine, epinephrine and serotonin. To find a potential therapeutic target for the treatment of gastric cancer, the biological functions of MAOA and the underlying mechanism in gastric cancer need to be explored. METHODS The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO) datasets, Kaplan‒Meier (KM) plotter were used to identify the differentially expressed genes, which mainly involved the degradation and synthesis enzymes of neurotransmitters in gastric cancer. We also investigated the expression pattern of MAOA in human and mouse tissues and cell lines by immunohistochemistry and Western blotting analysis. Western blotting, quantitative real-time PCR, enzyme-linked immunosorbent assay (ELISA) and a Seahorse experiment were used to identify the molecular mechanism of cancer cell glycolysis. MAOA expression and patient survival were analysed in the Ren Ji cohort, and univariate and multivariate analyses were performed based on the clinicopathological characteristics of the above samples. RESULTS MAOA expression was significantly downregulated in gastric cancer tissue and associated with poor patient prognosis. Moreover, the expression level of MAOA in gastric cancer tissue had a close negative correlation with the SUXmax value of PET-CT in patients. MAOA suppressed tumour growth and glycolysis and promoted cancer cell apoptosis. We also reported that MAOA can interact with NDRG1 and regulate glycolysis through suppression of the PI3K/Akt/mTOR pathway. MAOA expression may serve as an independent prognostic factor in gastric cancer patients. CONCLUSIONS MAOA attenuated glycolysis and inhibited the progression of gastric cancer through the PI3K/Akt/mTOR pathway. Loss of function or downregulation of MAOA can facilitate gastric cancer progression. Overexpression of MAOA and inhibition of the PI3K/Akt/mTOR pathway may provide a potential method for gastric cancer treatment in clinical therapeutic regimens.
Collapse
Affiliation(s)
- Yang-Yang Wang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Yao-Qi Zhou
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Jia-Xuan Xie
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Xiang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Shu-Chang Wang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Qing Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Li-Peng Hu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Shu-Heng Jiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Shuang-Qin Yi
- Department of Frontier Health Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Hachioji, Japan
| | - Jia Xu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China.
| | - Hui Cao
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China.
| | - En-Hao Zhao
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China.
| | - Jun Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China.
| |
Collapse
|
14
|
Montazeri Aliabadi H, Manda A, Sidgal R, Chung C. Targeting Breast Cancer: The Familiar, the Emerging, and the Uncharted Territories. Biomolecules 2023; 13:1306. [PMID: 37759706 PMCID: PMC10526846 DOI: 10.3390/biom13091306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/16/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
Breast cancer became the most diagnosed cancer in the world in 2020. Chemotherapy is still the leading clinical strategy in breast cancer treatment, followed by hormone therapy (mostly used in hormone receptor-positive types). However, with our ever-expanding knowledge of signaling pathways in cancer biology, new molecular targets are identified for potential novel molecularly targeted drugs in breast cancer treatment. While this has resulted in the approval of a few molecularly targeted drugs by the FDA (including drugs targeting immune checkpoints), a wide array of signaling pathways seem to be still underexplored. Also, while combinatorial treatments have become common practice in clinics, the majority of these approaches seem to combine molecularly targeted drugs with chemotherapeutic agents. In this manuscript, we start by analyzing the list of FDA-approved molecularly targeted drugs for breast cancer to evaluate where molecular targeting stands in breast cancer treatment today. We will then provide an overview of other options currently under clinical trial or being investigated in pre-clinical studies.
Collapse
Affiliation(s)
- Hamidreza Montazeri Aliabadi
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA
| | | | | | | |
Collapse
|
15
|
Cheng Q, Ning S, Zhu L, Zhang C, Jiang S, Hao Y, Zhu J. NDRG1 facilitates self-renewal of liver cancer stem cells by preventing EpCAM ubiquitination. Br J Cancer 2023; 129:237-248. [PMID: 37165202 PMCID: PMC10338678 DOI: 10.1038/s41416-023-02278-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/25/2023] [Accepted: 04/12/2023] [Indexed: 05/12/2023] Open
Abstract
BACKGROUND Portal vein tumour thrombus (PVTT) is the main pathway of HCC intrahepatic metastasis and is responsible for the poor prognosis of patients with HCC. However, the molecular mechanisms underlying PVTT vascular metastases have not been fully elucidated. METHODS NDRG1 expression was assessed by immunohistochemistry and immunoblotting in clinical specimens obtained from curative surgery. The functional relevance of NDRG1 was evaluated using sphere formation and animal models of tumorigenicity and metastasis. The relationship between NDRG1 and EpCAM was explored using molecular biological techniques. RESULTS NDRG1 protein was upregulated in HCC samples compared to non-tumorous tissues. Furthermore, NDRG1 expression was enhanced in the PVTT samples. Our functional study showed that NDRG1 was required for the self-renewal of tumour-initiating/cancer stem cells (CSCs). In addition, NDRG1 knockdown inhibited the proliferation and migration of PVTT-1 cells in vitro and in vivo. NDRG1 was found to stabilise the functional tumour-initiating cell marker EpCAM through protein-protein interactions and inhibition of EpCAM ubiquitination. CONCLUSION Our findings suggest that NDRG1 enhances CSCs expansion, PVTT formation and growth capability through the regulation of EpCAM stability. NDRG1 may be a promising target for the treatment of patients with HCC and PVTT.
Collapse
Affiliation(s)
- Qian Cheng
- Department of Hepatobiliary Surgery, Beijing Key Laboratory of HCC and Liver Cirrhosis, Peking University People's Hospital, 100044, Beijing, China.
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, 100101, Beijing, China.
| | - Shanglei Ning
- Department of General Surgery, Qilu Hospital of Shandong University, 250012, Shandong, China
| | - Lei Zhu
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, 100101, Beijing, China
| | - Changlu Zhang
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, 100101, Beijing, China
| | - Shaodong Jiang
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, 100101, Beijing, China
| | - Yajing Hao
- Key Laboratory of RNA Biology, Center for Big Data Research in Health, Institute of Biophysics, Chinese Academy of Sciences, 100101, Beijing, China
| | - Jiye Zhu
- Department of Hepatobiliary Surgery, Beijing Key Laboratory of HCC and Liver Cirrhosis, Peking University People's Hospital, 100044, Beijing, China.
| |
Collapse
|
16
|
Chang J, Lo ZHY, Alenizi S, Kovacevic Z. Re-Shaping the Pancreatic Cancer Tumor Microenvironment: A New Role for the Metastasis Suppressor NDRG1. Cancers (Basel) 2023; 15:2779. [PMID: 37345116 DOI: 10.3390/cancers15102779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/01/2023] [Accepted: 05/05/2023] [Indexed: 06/23/2023] Open
Abstract
Pancreatic cancer (PaC) is a highly aggressive disease, with poor response to current treatments and 5-year survival rates of 10-15%. PaC progression is facilitated by its interaction with the complex and multifaceted tumor microenvironment (TME). In the TME, cancer cells and surrounding stromal cells constantly communicate with each other via the secretion and uptake of factors including cytokines, chemokines, growth factors, metabolites, and extracellular vesicles (EVs), reshaping the landscape of PaC. Recent studies demonstrated that the metastasis suppressor N-myc downstream regulated 1 (NDRG1) not only inhibits oncogenic signaling pathways in PaC cells but also alters the communication between PaC cells and the surrounding stroma. In fact, NDRG1 was found to influence the secretome of PaC cells, alter cancer cell metabolism, and interfere with intracellular trafficking and intercellular communication between PaC cells and surrounding fibroblasts. This review will present recent advancements in understanding the role of NDRG1 in PaC progression, with a focus on how this molecule influences PaC-stroma communication and its potential for re-shaping the PaC TME.
Collapse
Affiliation(s)
- Jiawei Chang
- School of Medical Sciences, Faculty of Medicine & Health, University of Sydney, Sydney 2006, Australia
- Department of Physiology, School of Biomedical Sciences, Faculty of Medicine & Health, University of NSW, Sydney 2052, Australia
| | - Zoe H Y Lo
- School of Medical Sciences, Faculty of Medicine & Health, University of Sydney, Sydney 2006, Australia
| | - Shafi Alenizi
- School of Medical Sciences, Faculty of Medicine & Health, University of Sydney, Sydney 2006, Australia
| | - Zaklina Kovacevic
- School of Medical Sciences, Faculty of Medicine & Health, University of Sydney, Sydney 2006, Australia
- Department of Physiology, School of Biomedical Sciences, Faculty of Medicine & Health, University of NSW, Sydney 2052, Australia
| |
Collapse
|
17
|
Xiong S, Tan X, Wu X, Wan A, Zhang G, Wang C, Liang Y, Zhang Y. Molecular landscape and emerging therapeutic strategies in breast
cancer brain metastasis. Ther Adv Med Oncol 2023; 15:17588359231165976. [PMID: 37034479 PMCID: PMC10074632 DOI: 10.1177/17588359231165976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 03/06/2023] [Indexed: 04/07/2023] Open
Abstract
Breast cancer (BC) is the most commonly diagnosed cancer worldwide. Advanced BC
with brain metastasis (BM) is a major cause of mortality with no specific or
effective treatment. Therefore, better knowledge of the cellular and molecular
mechanisms underlying breast cancer brain metastasis (BCBM) is crucial for
developing novel therapeutic strategies and improving clinical outcomes. In this
review, we focused on the latest advances and discuss the contribution of the
molecular subtype of BC, the brain microenvironment, exosomes, miRNAs/lncRNAs,
and genetic background in BCBM. The blood–brain barrier and blood–tumor barrier
create challenges to brain drug delivery, and we specifically review novel
approaches to bypass these barriers. Furthermore, we discuss the potential
application of immunotherapies and genetic editing techniques based on
CRISPR/Cas9 technology in treating BCBM. Emerging techniques and research
findings continuously shape our views of BCBM and contribute to improvements in
precision therapies and clinical outcomes.
Collapse
Affiliation(s)
- Siyi Xiong
- Breast and Thyroid Surgery, Southwest Hospital,
Army Medical University, Chongqing, China
| | - Xuanni Tan
- Breast and Thyroid Surgery, Southwest Hospital,
Army Medical University, Chongqing, China
| | - Xiujuan Wu
- Breast and Thyroid Surgery, Southwest Hospital,
Army Medical University, Chongqing, China
| | - Andi Wan
- Breast and Thyroid Surgery, Southwest Hospital,
Army Medical University, Chongqing, China
| | - Guozhi Zhang
- Breast and Thyroid Surgery, Southwest Hospital,
Army Medical University, Chongqing, China
| | - Cheng Wang
- Breast and Thyroid Surgery, Southwest Hospital,
Army Medical University, Chongqing, China
| | - Yan Liang
- Breast and Thyroid Surgery, Southwest Hospital,
Army Medical University, 30 Gaotanyan, Shapingba, China Chongqing 400038,
China
| | | |
Collapse
|
18
|
Zhao X, Richardson DR. The role of the NDRG1 in the pathogenesis and treatment of breast cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:188871. [PMID: 36841367 DOI: 10.1016/j.bbcan.2023.188871] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 02/18/2023] [Accepted: 02/19/2023] [Indexed: 02/26/2023]
Abstract
Breast cancer (BC) is the leading cause of cancer death in women. This disease is heterogeneous, with clinical subtypes being estrogen receptor-α (ER-α) positive, having human epidermal growth factor receptor 2 (HER2) overexpression, or being triple-negative for ER-α, progesterone receptor, and HER2 (TNBC). The ER-α positive and HER2 overexpressing tumors can be treated with agents targeting these proteins, including tamoxifen and pertuzumab, respectively. Despite these treatments, resistance and metastasis are problematic, while TNBC is challenging to treat due to the lack of suitable targets. Many studies examining BC and other tumors indicate a role for N-myc downstream-regulated gene-1 (NDRG1) as a metastasis suppressor. The ability of NDRG1 to inhibit metastasis is due, in part, to the inhibition of the initial step in metastasis, namely the epithelial-to-mesenchymal transition. Paradoxically, there are also reports of NDRG1 playing a pro-oncogenic role in BC pathogenesis. The oncogenic effects of NDRG1 in BC have been reported to relate to lipid metabolism or the mTOR signaling pathway. The molecular mechanism(s) of how NDRG1 regulates the activity of multiple signaling pathways remains unclear. Therapeutic strategies that up-regulate NDRG1 have been developed and include agents of the di-2-pyridylketone thiosemicarbazone class. These compounds target oncogenic drivers in BC cells, suppressing the expression of multiple key hormone receptors including ER-α, progesterone receptor, androgen receptor, and prolactin receptor, and can also overcome tamoxifen resistance. Considering the varying role of NDRG1 in BC pathogenesis, further studies are required to examine what subset of BC patients would benefit from pharmacopeia that up-regulate NDRG1.
Collapse
Affiliation(s)
- Xiao Zhao
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland 4111, Australia
| | - Des R Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland 4111, Australia; Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| |
Collapse
|
19
|
Cell-Type-Specific Signalling Networks Impacted by Prostate Epithelial-Stromal Intercellular Communication. Cancers (Basel) 2023; 15:cancers15030699. [PMID: 36765657 PMCID: PMC9913520 DOI: 10.3390/cancers15030699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/14/2023] [Accepted: 01/20/2023] [Indexed: 01/25/2023] Open
Abstract
Prostate cancer is the second most common cause of cancer death in males. A greater understanding of cell signalling events that occur within the prostate cancer tumour microenvironment (TME), for example, between cancer-associated fibroblasts (CAFs) and prostate epithelial or cancer cells, may identify novel biomarkers and more effective therapeutic strategies for this disease. To address this, we used cell-type-specific labelling with amino acid precursors (CTAP) to define cell-type-specific (phospho)proteomic changes that occur when prostate epithelial cells are co-cultured with normal patient-derived prostate fibroblasts (NPFs) versus matched CAFs. We report significant differences in the response of BPH-1 benign prostate epithelial cells to CAF versus NPF co-culture. Pathway analysis of proteomic changes identified significant upregulation of focal adhesion and cytoskeleton networks, and downregulation of metabolism pathways, in BPH-1 cells cultured with CAFs. In addition, co-cultured CAFs exhibited alterations in stress, DNA damage, and cytoskeletal networks. Functional validation of one of the top differentially-regulated proteins in BPH-1 cells upon CAF co-culture, transglutaminase-2 (TGM2), demonstrated that knockdown of this protein significantly reduced the proliferation and migration of prostate epithelial cells. Overall, this study provides novel insights into intercellular communication in the prostate cancer TME that may be exploited to improve patient management.
Collapse
|
20
|
Bassiouni R, Idowu MO, Gibbs LD, Robila V, Grizzard PJ, Webb MG, Song J, Noriega A, Craig DW, Carpten JD. Spatial Transcriptomic Analysis of a Diverse Patient Cohort Reveals a Conserved Architecture in Triple-Negative Breast Cancer. Cancer Res 2023; 83:34-48. [PMID: 36283023 PMCID: PMC9812886 DOI: 10.1158/0008-5472.can-22-2682] [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: 08/23/2022] [Revised: 10/18/2022] [Accepted: 10/20/2022] [Indexed: 02/03/2023]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive disease that disproportionately affects African American (AA) women. Limited targeted therapeutic options exist for patients with TNBC. Here, we employ spatial transcriptomics to interrogate tissue from a racially diverse TNBC cohort to comprehensively annotate the transcriptional states of spatially resolved cellular populations. A total of 38,706 spatial features from a cohort of 28 sections from 14 patients were analyzed. Intratumoral analysis of spatial features from individual sections revealed heterogeneous transcriptional substructures. However, integrated analysis of all samples resulted in nine transcriptionally distinct clusters that mapped across all individual sections. Furthermore, novel use of join count analysis demonstrated nonrandom directional spatial dependencies of the transcriptionally defined shared clusters, supporting a conserved spatio-transcriptional architecture in TNBC. These findings were substantiated in an independent validation cohort comprising 17,861 spatial features representing 15 samples from 8 patients. Stratification of samples by race revealed race-associated differences in hypoxic tumor content and regions of immune-rich infiltrate. Overall, this study combined spatial and functional molecular analyses to define the tumor architecture of TNBC, with potential implications in understanding TNBC disparities. SIGNIFICANCE Spatial transcriptomics profiling of a diverse cohort of triple-negative breast cancers and innovative informatics approaches reveal a conserved cellular architecture across cancers and identify proportional differences in tumor cell composition by race.
Collapse
Affiliation(s)
- Rania Bassiouni
- Department of Translational Genomics, Keck School of Medicine, University of Southern California; Los Angeles, CA
| | - Michael O. Idowu
- Department of Pathology, Virginia Commonwealth University; Richmond, VA
| | - Lee D. Gibbs
- Department of Translational Genomics, Keck School of Medicine, University of Southern California; Los Angeles, CA
| | - Valentina Robila
- Department of Pathology, Virginia Commonwealth University; Richmond, VA
| | | | - Michelle G. Webb
- Department of Translational Genomics, Keck School of Medicine, University of Southern California; Los Angeles, CA
| | - Jiarong Song
- Department of Translational Genomics, Keck School of Medicine, University of Southern California; Los Angeles, CA
| | - Ashley Noriega
- Department of Translational Genomics, Keck School of Medicine, University of Southern California; Los Angeles, CA
| | - David W. Craig
- Department of Translational Genomics, Keck School of Medicine, University of Southern California; Los Angeles, CA
- Translational and Clinical Sciences Program, Norris Comprehensive Cancer Center, University of Southern California; Los Angeles, CA
| | - John D. Carpten
- Department of Translational Genomics, Keck School of Medicine, University of Southern California; Los Angeles, CA
- Translational and Clinical Sciences Program, Norris Comprehensive Cancer Center, University of Southern California; Los Angeles, CA
| |
Collapse
|
21
|
López-Tejada A, Griñán-Lisón C, González-González A, Cara FE, Luque RJ, Rosa-Garrido C, Blaya-Cánovas JL, Navarro-Ocón A, Valenzuela-Torres M, Parra-López M, Calahorra J, Blancas I, Marchal JA, Granados-Principal S. TGFβ Governs the Pleiotropic Activity of NDRG1 in Triple-Negative Breast Cancer Progression. Int J Biol Sci 2023; 19:204-224. [PMID: 36594086 PMCID: PMC9760438 DOI: 10.7150/ijbs.78738] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/24/2022] [Indexed: 11/24/2022] Open
Abstract
In triple-negative breast cancer (TNBC), the pleiotropic NDRG1 (N-Myc downstream regulated gene 1) promotes progression and worse survival, yet contradictory results were documented, and the mechanisms remain unknown. Phosphorylation and localization could drive NDRG1 pleiotropy, nonetheless, their role in TNBC progression and clinical outcome was not investigated. We found enhanced p-NDRG1 (Thr346) by TGFβ1 and explored whether it drives NDRG1 pleiotropy and TNBC progression. In tissue microarrays of 81 TNBC patients, we identified that staining and localization of NDRG1 and p-NDRG1 (Thr346) are biomarkers and risk factors associated with shorter overall survival. We found that TGFβ1 leads NDRG1, downstream of GSK3β, and upstream of NF-κB, to differentially regulate migration, invasion, epithelial-mesenchymal transition, tumor initiation, and maintenance of different populations of cancer stem cells (CSCs), depending on the progression stage of tumor cells, and the combination of TGFβ and GSK3β inhibitors impaired CSCs. The present study revealed the striking importance to assess both total NDRG1 and p-NDRG1 (Thr346) positiveness and subcellular localization to evaluate patient prognosis and their stratification. NDRG1 pleiotropy is driven by TGFβ to differentially promote metastasis and/or maintenance of CSCs at different stages of tumor progression, which could be abrogated by the inhibition of TGFβ and GSK3β.
Collapse
Affiliation(s)
- Araceli López-Tejada
- Department of Biochemistry and Molecular Biology 2, School of Pharmacy, University of Granada, 18011 Granada, Spain.,GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada-University of Granada, Spain; Conocimiento s/n 18100, Granada. Spain
| | - Carmen Griñán-Lisón
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada-University of Granada, Spain; Conocimiento s/n 18100, Granada. Spain.,UGC de Oncología Médica, Hospital Universitario de Jaén, 23007 Jaén, Spain
| | - Adrián González-González
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain
| | - Francisca E. Cara
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain
| | - Rafael J. Luque
- UGC de Anatomía Patológica, Hospital Universitario de Jaén, Jaén, Spain
| | - Carmen Rosa-Garrido
- FIBAO, Hospital Universitario de Jaén, Servicio Andaluz de Salud, Jaén, Spain
| | - José L. Blaya-Cánovas
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada-University of Granada, Spain; Conocimiento s/n 18100, Granada. Spain.,UGC de Oncología Médica, Hospital Universitario de Jaén, 23007 Jaén, Spain
| | - Alba Navarro-Ocón
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada-University of Granada, Spain; Conocimiento s/n 18100, Granada. Spain
| | - María Valenzuela-Torres
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain
| | - Marisa Parra-López
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain
| | - Jesús Calahorra
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada-University of Granada, Spain; Conocimiento s/n 18100, Granada. Spain.,UGC de Oncología Médica, Hospital Universitario de Jaén, 23007 Jaén, Spain
| | - Isabel Blancas
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada-University of Granada, Spain; Conocimiento s/n 18100, Granada. Spain.,UGC de Oncología, Hospital Universitario “San Cecilio”, 18016 Granada, Spain
| | - Juan A. Marchal
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada-University of Granada, Spain; Conocimiento s/n 18100, Granada. Spain.,Department of Human Anatomy and Embryology, Biopathology and Regenerative Medicine Institute (IBIMER), University of Granada, 18011 Granada, Spain.,Excellence Research Unit “Modeling Nature” (MNat), University of Granada, Spain
| | - Sergio Granados-Principal
- Department of Biochemistry and Molecular Biology 2, School of Pharmacy, University of Granada, 18011 Granada, Spain.,GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada-University of Granada, Spain; Conocimiento s/n 18100, Granada. Spain.,✉ Corresponding author: E-mail: . Phone number: +34 651 55 79 21
| |
Collapse
|
22
|
NDRG1 in Cancer: A Suppressor, Promoter, or Both? Cancers (Basel) 2022; 14:cancers14235739. [PMID: 36497221 PMCID: PMC9737586 DOI: 10.3390/cancers14235739] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/21/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022] Open
Abstract
N-myc downregulated gene-1 (NDRG1) has been variably reported as a metastasis suppressor, a biomarker of poor outcome, and a facilitator of disease progression in a range of different cancers. NDRG1 is poorly understood in cancer due to its context-dependent and pleiotropic functions. Within breast cancer, NDRG1 is reported to be either a facilitator of, or an inhibitor of tumour progression and metastasis. The wide array of roles played by NDRG1 are dependent on post-translational modifications and subcellular localization, as well as the cellular context, for example, cancer type. We present an update on NDRG1, and its association with hallmarks of cancer such as hypoxia, its interaction with oncogenic proteins such as p53 as well its role in oncogenic and metastasis pathways in breast and other cancers. We further comment on its functional implications as a metastasis suppressor and promoter, its clinical relevance, and discuss its therapeutic targetability in different cancers.
Collapse
|
23
|
Zhao M, Pan B, He Y, Niu B, Gao X. Elucidating the pharmacological mechanism by which Si-Wu-Tang induces cellular senescence in breast cancer via multilevel data integration. Aging (Albany NY) 2022; 14:5812-5837. [PMID: 35859293 PMCID: PMC9365552 DOI: 10.18632/aging.204185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/27/2022] [Indexed: 01/10/2023]
Abstract
Traditional Chinese medicine (TCM) is a promising strategy for effectively treating cancer by inducing cellular senescence with minimal side effects. Si-Wu-Tang (SWT) is a TCM composed of four herbs that is commonly used in China for the treatment of gynecological diseases; SWT can prevent breast cancer (BC), but the molecular mechanism by which SWT induces cellular senescence and its clinical application value remain unknown. We identified 335 differentially expressed genes (DEGs) in SWT-treated MCF-7 cells through Gene Expression Omnibus (GEO) dataset analysis. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses revealed the enrichment of biological processes and key signaling pathways including cellular senescence, the cell cycle, the MAPK signaling pathway, and the p53 signaling pathway. Additionally, SWT induced BC cell senescence by upregulating the expression of 33 aging/senescence-induced genes (ASIGs). According to LASSO regression analysis, NDRG1, ERRFI1, SOCS1, IRS2, IGFBP4, and BIRC3 levels were associated with BC prognosis and were used to develop risk scores. ERRFI1, SOCS1, IRS2, IGFBP4, and BIRC3 were identified as protective factors (P < 0.05, HR < 1), while NDRG1 was identified as a risk factor (P < 0.05, HR > 1). Notably, patients with low risk scores had increased senescence-associated secretory phenotypes (SASPs) and immune cell infiltration. Overall, we systematically integrated biological databases and biocomputational methods to reveal the mechanisms by which SWT induces senescence in breast cancer and its clinical value.
Collapse
Affiliation(s)
- Minhong Zhao
- Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan 528000, PR China
| | - Botao Pan
- Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan 528000, PR China
| | - Yanjun He
- Department of Emergency, Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan 528000, PR China
| | - Bo Niu
- Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan 528000, PR China
| | - Xiuan Gao
- Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan 528000, PR China
| |
Collapse
|
24
|
Rehman AU, Khan P, Maurya SK, Siddiqui JA, Santamaria-Barria JA, Batra SK, Nasser MW. Liquid biopsies to occult brain metastasis. Mol Cancer 2022; 21:113. [PMID: 35538484 PMCID: PMC9088117 DOI: 10.1186/s12943-022-01577-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 04/19/2022] [Indexed: 02/08/2023] Open
Abstract
Brain metastasis (BrM) is a major problem associated with cancer-related mortality, and currently, no specific biomarkers are available in clinical settings for early detection. Liquid biopsy is widely accepted as a non-invasive method for diagnosing cancer and other diseases. We have reviewed the evidence that shows how the molecular alterations are involved in BrM, majorly from breast cancer (BC), lung cancer (LC), and melanoma, with an inception in how they can be employed for biomarker development. We discussed genetic and epigenetic changes that influence cancer cells to breach the blood-brain barrier (BBB) and help to establish metastatic lesions in the uniquely distinct brain microenvironment. Keeping abreast with the recent breakthroughs in the context of various biomolecules detections and identifications, the circulating tumor cells (CTC), cell-free nucleotides, non-coding RNAs, secretory proteins, and metabolites can be pursued in human body fluids such as blood, serum, cerebrospinal fluid (CSF), and urine to obtain potential candidates for biomarker development. The liquid biopsy-based biomarkers can overlay with current imaging techniques to amplify the signal viable for improving the early detection and treatments of occult BrM.
Collapse
Affiliation(s)
- Asad Ur Rehman
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68108, USA
| | - Parvez Khan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68108, USA
| | - Shailendra Kumar Maurya
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68108, USA
| | - Jawed A Siddiqui
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68108, USA.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68108, USA
| | | | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68108, USA.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68108, USA.,Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE-68198, USA
| | - Mohd Wasim Nasser
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68108, USA. .,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68108, USA.
| |
Collapse
|
25
|
You GR, Chang JT, Li HF, Cheng AJ. Multifaceted and Intricate Oncogenic Mechanisms of NDRG1 in Head and Neck Cancer Depend on Its C-Terminal 3R-Motif. Cells 2022; 11:cells11091581. [PMID: 35563887 PMCID: PMC9104279 DOI: 10.3390/cells11091581] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/04/2022] [Accepted: 05/05/2022] [Indexed: 12/12/2022] Open
Abstract
N-Myc downstream-regulated 1 (NDRG1) has inconsistent oncogenic functions in various cancers. We surveyed and characterized the role of NDRG1 in head and neck cancer (HNC). Cellular methods included spheroid cell formation, clonogenic survival, cell viability, and Matrigel invasion assays. Molecular techniques included transcriptomic profiling, RT-qPCR, immunoblotting, in vitro phosphorylation, immunofluorescent staining, and confocal microscopy. Prognostic significance was assessed by Kaplan–Meier analysis. NDRG1 participated in diverse oncogenic functions in HNC cells, mainly stress response and cell motility. Notably, NDRG1 contributed to spheroid cell growth, radio-chemoresistance, and upregulation of stemness-related markers (CD44 and Twist1). NDRG1 facilitated cell migration and invasion, and was associated with modulation of the extracellular matrix molecules (fibronectin, vimentin). Characterizing the 3R-motif in NDRG1 revealed its mechanism in the differential regulation of the phenotypes. The 3R-motif displayed minimal effect on cancer stemness but was crucial for cell motility. Phosphorylating the motif by GSK3b at serine residues led to its nuclear translocation to promote motility. Clinical analyses supported the oncogenic function of NDRG1, which was overexpressed in HNC and associated with poor prognosis. The data elucidate the multifaceted and intricate mechanisms of NDRG1 in HNC. NDRG1 may be a prognostic indicator or therapeutic target for refractory HNC.
Collapse
Affiliation(s)
- Guo-Rung You
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
| | - Joseph T. Chang
- Department of Radiation Oncology and Proton Therapy Center, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 33302, Taiwan;
- School of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Hsiao-Fan Li
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
| | - Ann-Joy Cheng
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
- Department of Radiation Oncology and Proton Therapy Center, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 33302, Taiwan;
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
- Correspondence: ; Tel.: +886-3-211-8800
| |
Collapse
|
26
|
de Nonneville A, Finetti P, Mamessier E, Bertucci F. RE: NDRG1 in Aggressive Breast Cancer Progression and Brain Metastasis. J Natl Cancer Inst 2022; 114:1046-1047. [PMID: 35148398 PMCID: PMC9275762 DOI: 10.1093/jnci/djac031] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 01/12/2022] [Accepted: 02/02/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Alexandre de Nonneville
- Laboratory of Predictive Oncology, Equipe labellisée Ligue Nationale Contre Le Cancer, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Marseille, France
| | - Pascal Finetti
- Laboratory of Predictive Oncology, Equipe labellisée Ligue Nationale Contre Le Cancer, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Marseille, France
| | - Emilie Mamessier
- Laboratory of Predictive Oncology, Equipe labellisée Ligue Nationale Contre Le Cancer, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Marseille, France
| | - François Bertucci
- Correspondence to: François Bertucci, MD, PhD, Department of Medical Oncology, Laboratory of Predictive Oncology, Institut Paoli-Calmettes, 232 Bd. Sainte-Marguerite, 13009 Marseille, France (e-mail: )
| |
Collapse
|