1
|
Zhang L, Yang J, Zhou Z, Ren Y, Chen B, Tang A, Zhang K, Li C, Zhou H, Fung KM, Xu C, Kang C, Battiste JD, Bronze MS, Houchen CW, Liu Z, Dunn IF, Cavenee WK, Li M. A zinc transporter drives glioblastoma progression via extracellular vesicles-reprogrammed microglial plasticity. Proc Natl Acad Sci U S A 2025; 122:e2427073122. [PMID: 40305049 PMCID: PMC12067291 DOI: 10.1073/pnas.2427073122] [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: 12/27/2024] [Accepted: 03/22/2025] [Indexed: 05/02/2025] Open
Abstract
Glioblastoma (GBM) is the most aggressive form of brain cancer, with limited therapeutic options. While microglia contribute to GBM progression, the mechanisms by which they foster a protumorigenic immune environment remain poorly understood. We identify the zinc transporter Zrt- And Irt-Like Protein 4 (ZIP4) as a pivotal regulator of the GBM immune landscape. In orthotopic mouse models, ZIP4 drives tumor growth and behavioral changes. Mechanistically, ZIP4 modulates microglial plasticity through tumor-derived extracellular vesicles carrying triggering receptor expressed on myeloid cells-1 (TREM1), a process regulated by the zinc-dependent transcription factor Zinc Finger E-box Binding Homeobox 1 in GBM cells. TREM1 enhances microglial plasticity through the spleen associated tyrosine kinase-Pyruvate dehydrogenase kinase-signal transducer and activator of transcription 3 (SYK-PDK-STAT3) signaling axis, ultimately promoting an immune environment favorable to tumor progression. ZIP4 depletion or TREM1 inhibition attenuates tumor growth and behavioral effects in vivo by disrupting the tumor-microglia interaction. These findings establish ZIP4 as a key modulator of the GBM immune landscape and suggest a promising therapeutic target to counteract microglia-mediated tumor progression.
Collapse
Affiliation(s)
- Liyang Zhang
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan410008, China
| | - Jingxuan Yang
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Zhijun Zhou
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Yu Ren
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Bo Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan410008, China
| | - Anliu Tang
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan410013, China
| | - Kailiang Zhang
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Chuntao Li
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan410008, China
| | - Hongshu Zhou
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan410008, China
| | - Kar-Ming Fung
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Chao Xu
- Department of Biostatistics and Epidemiology, College of Public Health, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Chunsheng Kang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Key Laboratory of Post-Trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin300052, China
| | - James D. Battiste
- Department of Neurosurgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Michael S. Bronze
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Courtney W. Houchen
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan410008, China
| | - Ian F. Dunn
- Department of Neurosurgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Webster K. Cavenee
- Department of Medicine, University of California at San Diego, San Diego, CA92093
| | - Min Li
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| |
Collapse
|
2
|
Buj R, Cole AR, Danielson J, Xu J, Hurd D, Kishore A, Kedziora KM, Chen J, Yang B, Barras D, Uboveja A, Amalric A, Apiz Saab JJ, Wickramasinghe J, Tangudu NK, Levasseur E, Wang H, Minasyan A, Dadey RE, Sharrow AC, Kunning S, Vendetti FP, Rivadeneira DB, Bakkenist CJ, Bruno TC, Delgoffe GM, Hempel N, Snyder NW, Bao R, Soloff AC, Kirk-Wood JM, Dangaj Laniti D, Kossenkov AV, Muir A, Das J, Davar D, Mesaros C, Aird KM. CDKN2A Low cancer cells outcompete macrophages for microenvironmental zinc to drive immunotherapy resistance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.08.637227. [PMID: 39975044 PMCID: PMC11839072 DOI: 10.1101/2025.02.08.637227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Approximately 50% of cancers exhibit decreased CDKN2A expression ( CDKN2A Low ), which is linked to immune checkpoint blockade (ICB) resistance. While CDKN2A is traditionally recognized as a tumor suppressor and cell cycle regulator, we have previously put forth a new paradigm demonstrating its role in intracellular metabolic reprogramming. Whether the metabolic derangement due to CDKN2A loss alters metabolites within the tumor microenvironment (TME) and how that affects the immune compartment and ICB response has never been investigated. Here we found that CDKN2A Low cancer cells reorganize zinc compartmentalization by upregulating the zinc importer SLC39A9 in the plasma membrane, leading to intracellular zinc accumulation in cancer cells and concurrent zinc depletion in the TME. This competition for zinc results in zinc-starved tumor-associated macrophages (TAMs), leading to reduced phagocytic activity. Increasing zinc in TAMs through multiple approaches, including a dietary intervention that increases availability of TME zinc, re-educates these TAMs to a pro-phagocytic phenotype. Remarkably, both knockdown of Slc39a9 in cancer cells or providing a high zinc diet sensitizes Cdkn2a Low tumors to ICB. TAMs, not T cells, are indispensable for ICB response. Clinically, TAMs from CDKN2A Low cancer patients have decreased zinc signatures, corresponding to reduced phagocytosis signatures. Moreover, patients with low circulating zinc levels have reduced time-to-event outcomes compared to those with higher zinc levels. Our work reveals a previously unrecognized mechanism through which CDKN2A Low cancer cells outcompete TAMs for zinc, directly disrupting their function and ICB efficacy.
Collapse
|
3
|
Zhang Y, Hu R, Su M, Hu J. Probing the substrate binding-induced conformational change of a ZIP metal transporter using a sandwich ELISA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.09.642161. [PMID: 40161815 PMCID: PMC11952358 DOI: 10.1101/2025.03.09.642161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Zrt-/Irt-like proteins (ZIPs), a family of divalent metal transporters, are crucial for maintaining the homeostasis of zinc, an essential trace element involved in numerous biological processes. While extensive research on the prototypical ZIP from Bordetella bronchiseptica (BbZIP) have suggested an elevator transport mechanism, the dynamic conformational changes during the transport cycle have not been thoroughly studied. In this work, we developed a sandwich ELISA using a custom anti-BbZIP monoclonal antibody to investigate the conformational change induced by the metal binding to the transport site. This was achieved by determining the accessibility of a cysteine residue introduced at a position exposed to the solvent only when the transporter adopts an outward-facing conformation. This assay allowed us to report the dissociation constants of BbZIP for Zn2+ and Cd2+ at low and sub-micromolar levels, respectively. Notably, the installation of a positive charge at the M2 site drastically reduced metal binding at the M1 site, consistent with an auxiliary role for the M2 site in metal transport. We also demonstrated that this assay can be used to rapidly screen variants for subsequent structural study. We anticipate that other transporters where substrate binding induces large conformational changes can also be studied using this method.
Collapse
Affiliation(s)
- Yao Zhang
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824
| | - Ryan Hu
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824
| | - Min Su
- Electron Microscopy Core, University of Missouri, MO 65211
- Department of Biochemistry, University of Missouri, MO 65211
| | - Jian Hu
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824
- Department of Chemistry, Michigan State University, East Lansing, MI 48824
| |
Collapse
|
4
|
Iseki M, Mizuma M, Shimura M, Kokumai T, Sato H, Kusaka A, Aoki S, Inoue K, Nakayama S, Douchi D, Miura T, Maeda S, Ishida M, Nakagawa K, Kamei T, Unno M. Preoperative Chemotherapy With Gemcitabine for Pancreatic Cancer Causes Zinc Deficiency. Pancreas 2025; 54:e75-e81. [PMID: 39102680 DOI: 10.1097/mpa.0000000000002396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
OBJECTIVES The aim of this study was to investigate how preoperative chemotherapy affected the serum zinc concentrations in patients with pancreatic cancer (PC). MATERIALS AND METHODS Two hundreds thirty-one patients with PC who underwent pancreatectomy at our department from 2013 to 2019 were enrolled in this study and measured for the serum zinc concentrations before pancreatectomy. Patient characteristics, course of treatment, and laboratory data were analyzed. RESULTS One hundred thirty-five patients underwent upfront pancreatectomy and 58 received preoperative gemcitabine + S1 (GEM + S1) and 29 received gemcitabine + nab-paclitaxel (GEM + nab-PTX). Comparing the serum zinc concentrations before and after preoperative treatment, it was found to decrease after treatment with statistical difference (79.3 μg/dL vs 68.7 μg/dL, P < 0.001). The result was consistent with the investigation for both the patients who received GEM + S1 and those who received GEM + nab-PTX ( P = 0.019, P < 0.001, respectively). CONCLUSIONS The preoperative chemotherapy consistently reduced the serum zinc concentrations in the PC patients, regardless of their regimen such as GEM + S1 and GEM + nab-PTX. Monitoring the serum zinc concentration and appropriate zinc supplementation may be essential for PC patients undergoing preoperative chemotherapy and pancreatectomy.
Collapse
Affiliation(s)
- Masahiro Iseki
- From the Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Woyciehowsky M, Larson P, Stephan AR, Dandridge SL, Idonije D, Berg KA, Lanthier A, Acuna SA, Stites SW, Gebhardt WJ, Holtzen SE, Rakshit A, Palmer AE. Systematic characterization of zinc in a series of breast cancer cell lines reveals significant changes in zinc homeostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.11.632547. [PMID: 39868107 PMCID: PMC11761790 DOI: 10.1101/2025.01.11.632547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
An optimal amount of labile zinc (Zn 2+ ) is essential for proliferation of human cells, where Zn 2+ levels that are too high or too low cause cell cycle exit. Tumors of the breast have been characterized by high levels of total Zn 2+ . Given the role of Zn 2+ in proliferation of human cells and elevation of zinc in breast cancer tumors, we examined the concentration of total and labile Zn 2+ across a panel of 5 breast cancer cell lines, compared to the normal MCF10A cell line. We found that three cell lines (MDA-MB-231, MDA-MB-157, and SK-Br-3) showed elevated labile Zn 2+ in the cytosol, while T-47D showed significantly lower Zn 2+ , and MCF7 showed no change compared to MCF10A cells. There was no change in total Zn 2+ across the cell lines, as measured by ICP-MS, but we did observe a difference in the cells ability to accumulate Zn 2+ when Zn 2+ in the media was elevated. Therefore, we examined how proliferation of each cell line was affected by increases and decreases in the media. We found striking differences, where three cancer cell lines (MDA-MB-231, MDA-MB-157, and MCF7) showed robust proliferation in high Zn 2+ at concentrations that killed MCF10A, T-47D, and SK-Br-3 cells. We also discovered that 4 of the 5 cancer cell lines demonstrate compromised proliferation and increased cell death in low Zn 2+ , suggesting these cells may be addicted to Zn 2+ . Overall, our study suggests significant differences in Zn 2+ homeostasis and regulation in different types of breast cancer cells, with consequences for both proliferation and cell viability.
Collapse
|
6
|
Wang X, Zhang M, Ma J, Tie Y, Wang S. Biochemical Markers of Zinc Nutrition. Biol Trace Elem Res 2024; 202:5328-5338. [PMID: 38319550 DOI: 10.1007/s12011-024-04091-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 01/29/2024] [Indexed: 02/07/2024]
Abstract
Zinc is an important trace element involved in the biochemical and physiological functions of the organism and is essential in the human body. It has been reported that 17.3% of people around the world are at risk of many diseases due to zinc deficiency, which has already affected people's healthy lives. Currently, mild zinc deficiency is difficult to diagnose early due to the lack of typical clinical manifestations, so finding zinc biomarkers is crucial for people's health. The present article reviews the main representative zinc biomarkers, such as body fluid zinc levels, zinc-dependent proteins, tissue zinc, and zinc-containing enzymes, to provide a reference for actively promoting the study of zinc nutritional status and early clinical diagnosis.
Collapse
Affiliation(s)
- Xinying Wang
- North China University of Science and Technology, Tangshan, Hebei Province, 063210, China
| | - Menghui Zhang
- North China University of Science and Technology, Tangshan, Hebei Province, 063210, China
| | - Jing Ma
- Hebei Key Laboratory of Reproductive Medicine, Hebei Reproductive Health Hospital, Shijiazhuang, Hebei Province, 050071, China
| | - Yanqing Tie
- Hebei General Hospital, Shijiazhuang, Hebei Province, 050051, China.
| | - Shusong Wang
- Hebei Key Laboratory of Reproductive Medicine, Hebei Reproductive Health Hospital, Shijiazhuang, Hebei Province, 050071, China.
| |
Collapse
|
7
|
Zhang Y, Jafari M, Zhang T, Sui D, Sagresti L, Merz KM, Hu J. Molecular insights into substrate translocation in an elevator-type metal transporter. Nat Commun 2024; 15:9665. [PMID: 39516201 PMCID: PMC11549095 DOI: 10.1038/s41467-024-54048-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
The Zrt/Irt-like protein (ZIP) metal transporters are key players in maintaining the homeostasis of a panel of essential microelements. The prototypical ZIP from Bordetella bronchiseptica (BbZIP) is an elevator transporter, but how the metal substrate moves along the transport pathway and how the transporter changes conformation to allow alternating access remain to be elucidated. Here, we combine structural, biochemical, and computational approaches to investigate the process of metal substrate translocation along with the global structural rearrangement. Our study reveals an upward hinge motion of the transport domain in a high-resolution crystal structure of a cross-linked variant, elucidates the mechanisms of metal release from the transport site into the cytoplasm and activity regulation by a cytoplasmic metal-binding loop, and unravels an unusual elevator mode in enhanced sampling simulations that distinguishes BbZIP from other elevator transporters. This work provides important insights into the metal transport mechanism of the ZIP family.
Collapse
Affiliation(s)
- Yao Zhang
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Majid Jafari
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Tuo Zhang
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, MI, USA
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Dexin Sui
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Luca Sagresti
- Scuola Normale Superiore, Pisa, Italy
- Istituto Nazionale di Fisica Nucleare (INFN) sezione di Pisa, Pisa, Italy
| | - Kenneth M Merz
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, MI, USA.
- Department of Chemistry, Michigan State University, East Lansing, MI, USA.
- Center for Computational Life Sciences, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, USA.
| | - Jian Hu
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, MI, USA.
- Department of Chemistry, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
8
|
Guo H, Wang S, Zhang H, Li J, Wang C, Liu Z, Chen J, Wang K, Wei X, Wei Q, Xu X. Research progress on the molecular structure, function, and application in tumor therapy of zinc transporter ZIP4. Int J Biol Sci 2024; 20:5910-5924. [PMID: 39664563 PMCID: PMC11628325 DOI: 10.7150/ijbs.102460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 10/22/2024] [Indexed: 12/13/2024] Open
Abstract
ZIP4, a pivotal member of the ZIP family, is the causative gene for the hereditary disorder AE (acrodermatitis enteropathica) in humans, and plays an essential role in regulating zinc ion balance within cells. While research on the molecular structure of ZIP4 continues, there remains a lack of full understanding regarding the stereo-structural conformation of ZIP4 molecules. Currently, there are two hypotheses concerning the transport of zinc ions into the cytoplasm by ZIP4, with some contradictions between experimental studies. Recent investigations have revealed that ZIP4 is involved in tumor growth, metastasis, drug tolerance, and various other processes. Most studies suggest that ZIP4 regulates the malignant biological behavior of tumors through zinc ions as a second messenger: however, latest research has identified that ZIP4 itself binds to Ephrin-B1 to regulate tumor metastasis. This review provides a comprehensive summary of the molecular structure of ZIP4 and its mechanism for transporting zinc ions while also exploring mutual regulation between zinc ions and ZIP4. Furthermore, it summarizes recent research progress on the role of ZIP4 in tumors and discusses its potential as a target for anticancer therapy based on an extensive analysis of research findings. These insights can guide future investigations into the role of ZIP4 in tumors.
Collapse
Affiliation(s)
- Haijun Guo
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Shaohua Wang
- Zhejiang University School of Medicine, Hangzhou, 310058, China
- Shaoxing city Keqiao District TCM hospital Medical Alliance General Hospital, Shaoxing, 312000, China
| | - Hui Zhang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medicine University, Hangzhou, 310053, China
| | - Jie Li
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Chao Wang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Zhikun Liu
- Department of Hepatobiliary and Pancreatic Surgery, People's Hospital Affiliated to Hangzhou Medical College (Zhejiang Provincial People's Hospital), Hangzhou, 310024, China
| | - Jun Chen
- Department of Hepatobiliary and Pancreatic Surgery, People's Hospital Affiliated to Hangzhou Medical College (Zhejiang Provincial People's Hospital), Hangzhou, 310024, China
| | - Kai Wang
- Department of Hepatobiliary and Pancreatic Surgery, People's Hospital Affiliated to Hangzhou Medical College (Zhejiang Provincial People's Hospital), Hangzhou, 310024, China
| | - Xuyong Wei
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Qiang Wei
- Department of Hepatobiliary and Pancreatic Surgery, People's Hospital Affiliated to Hangzhou Medical College (Zhejiang Provincial People's Hospital), Hangzhou, 310024, China
| | - Xiao Xu
- Zhejiang University School of Medicine, Hangzhou, 310058, China
- Department of Hepatobiliary and Pancreatic Surgery, People's Hospital Affiliated to Hangzhou Medical College (Zhejiang Provincial People's Hospital), Hangzhou, 310024, China
- Institute of Translational Medical, Zhejiang University, Hangzhou, 310006, China
| |
Collapse
|
9
|
Iwabuchi E, Miki Y, Xu J, Kanai A, Ishida T, Sasano H, Suzuki T. Zinc transporter ZnT5 is associated with epithelial mesenchymal transition via SMAD1 in breast cancer. Int J Exp Pathol 2024; 105:184-192. [PMID: 39138630 PMCID: PMC11574640 DOI: 10.1111/iep.12515] [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: 11/29/2023] [Revised: 07/16/2024] [Accepted: 07/27/2024] [Indexed: 08/15/2024] Open
Abstract
Zinc levels in breast cancer tissues have been reported to be higher than those in normal tissues. In addition, the expression levels of zinc transporters, including ZnT5 and ZnT6, are reportedly higher in breast cancer than in normal breast tissues. ZnT5 and ZnT6 also contribute to heterodimer formation and are involved in several biological functions. However, the functions of ZnT5 and ZnT6 heterodimers in breast cancer remain unknown. Therefore, we first investigated the immunolocalization of ZnT5 and ZnT6 in pathological breast cancer specimens and in MCF-7 and T-47D breast cancer cells. Next, we used small interfering RNA to assess cell viability and migration in ZnT5 knockdown MCF-7 and T-47D cells. Immunohistochemical analysis showed that the number of ZnT5-positive breast cancer cells was inversely correlated with the pathologic N factor status. ZnT5 knockdown had no effect on cell viability in the presence of 100 μM ZnCl2 in MCF-7 and T-47D cells. In a wound healing assay, 100 μM ZnCl2 treatment inhibited cell migration of MCF-7 and T-47D cells, whereas ZnT5 knockdown promoted cell migration, decreased E-cadherin expression and increased vimentin, slug and matrix metalloproteinase 9 expression. Antibody arrays showed that ZnT5 knockdown increased the expression of SMAD1, and that dorsomorphin treatment inhibited the promotion of migratory ability induced by ZnT5 knockdown. The results of this study revealed that both ZnT5 may be involved in less aggressive breast cancer subtypes, possibly through inhibition of cell migration.
Collapse
Affiliation(s)
- Erina Iwabuchi
- Department of Pathology and Histotechnology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yasuhiro Miki
- Department of Anatomic Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Junyao Xu
- The Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Zhejiang, Hangzhou, China
| | - Ayako Kanai
- Department of Breast Surgery, Hachinohe City Hospital, Aomori, Japan
| | - Takanori Ishida
- Department of Breast and Endocrine Surgical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hironobu Sasano
- Department of Anatomic Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takashi Suzuki
- Department of Pathology and Histotechnology, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Anatomic Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Pathology, Tohoku University Hospital, Sendai, Japan
| |
Collapse
|
10
|
Hu J, Jiang Y. Evolution, classification, and mechanisms of transport, activity regulation, and substrate specificity of ZIP metal transporters. Crit Rev Biochem Mol Biol 2024; 59:245-266. [PMID: 39431645 DOI: 10.1080/10409238.2024.2405476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/23/2024] [Accepted: 09/13/2024] [Indexed: 10/22/2024]
Abstract
The Zrt/Irt-like protein (ZIP) family consists of ubiquitously expressed divalent d-block metal transporters that play central roles in the uptake, secretion, excretion, and distribution of several essential and toxic metals in living organisms. The past few years has witnessed rapid progress in the molecular basis of these membrane transport proteins. In this critical review, we summarize the research progress at the molecular level of the ZIP family and discuss the future prospects. Furthermore, an evolutionary path for the unique ZIP fold and a new classification of the ZIP family are proposed based on the presented structural and sequence analyses.
Collapse
Affiliation(s)
- Jian Hu
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
- Department of Chemistry, Michigan State University, East Lansing, MI, USA
| | - Yuhan Jiang
- Department of Chemistry, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
11
|
Tang Y, Guo S, Yu N, Li H. ZIP4: a promising early diagnostic and therapeutic targets for pancreatic cancer. Am J Cancer Res 2024; 14:4652-4664. [PMID: 39417191 PMCID: PMC11477812 DOI: 10.62347/avym3477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 09/15/2024] [Indexed: 10/19/2024] Open
Abstract
Pancreatic cancer is an aggressive and metastatic tumor that lacks effective early detection and treatment methods. There is an urgent need to further understand its underlying molecular mechanisms and identify new biomarkers for early detection. Zinc, a critical trace element and catalytic cofactor, is tightly regulated within cells. ZIP4, a zinc transporter protein significantly overexpressed in human pancreatic cancer, appears to play a pivotal role in tumor development by modulating intracellular zinc concentration. This review highlights the role of ZIP4 in tumorigenesis, including its impact on pancreatic cancer growth, proliferation, migration, and drug resistance. ZIP4 exerts its effects by regulating zinc dependent transcriptional factors like CREB, STAT3, and ZEB1, resulting in upregulation of Cyclin D1, TP53INP1, ITGA3, CD44, ENT1 proteins, and miR-373. Moreover, ZIP4 mediates the miR373-PHLPP2-AKT signaling axis, which increases TGF-β expression. Coupled with CREB-activated macrophage catabolism-related genes SDC1 and DNM2, ZIP4 promotes cancer cachexia and supports amino acids to tumor cells under metabolic stress. Furthermore, ZIP4 facilitates bone resorption by osteoclasts via the RANKL-activated NF-κB pathway. A deeper understanding of these mechanisms may unveil potential targets for early diagnosis, prognosis assessment, and dietary recommendations for pancreatic cancer. These findings hold clinical significance not only for pancreatic cancer but also for other malignancies exhibiting heightened ZIP4 expression.
Collapse
Affiliation(s)
- Yunpeng Tang
- Department of Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai, PR China
| | - Sheng Guo
- Department of Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai, PR China
| | - Nianhui Yu
- Department of Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai, PR China
| | - Hui Li
- Department of Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai, PR China
| |
Collapse
|
12
|
Zhang Y, Jafari M, Zhang T, Sui D, Sagresti L, Merz KM, Hu J. Molecular insights into substrate translocation in an elevator-type metal transporter. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.18.613805. [PMID: 39345646 PMCID: PMC11429975 DOI: 10.1101/2024.09.18.613805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
The Zrt/Irt-like protein (ZIP) metal transporters are key players in maintaining the homeostasis of a panel of essential microelements. The prototypical ZIP from Bordetella bronchiseptica (BbZIP) is an elevator transporter, but how the metal substrate moves along the transport pathway and how the transporter changes conformation to allow alternating access remain to be elucidated. Here, we combined structural, biochemical, and computational approaches to investigate the process of metal substrate translocation along with the global structural rearrangement. Our study revealed an upward hinge motion of the transport domain in a high-resolution crystal structure of a cross-linked variant, elucidated the mechanisms of metal release from the transport site into the cytoplasm and activity regulation by a cytoplasmic metal-binding loop, and unraveled an unusual elevator mode in enhanced sampling simulations that distinguishes BbZIP from other elevator transporters. This work provides important insights into the metal transport mechanism of the ZIP family.
Collapse
Affiliation(s)
- Yao Zhang
- Department of Biochemistry & Molecular Biology, Michigan State University, MI 48824
| | - Majid Jafari
- Department of Biochemistry & Molecular Biology, Michigan State University, MI 48824
| | - Tuo Zhang
- Department of Biochemistry & Molecular Biology, Michigan State University, MI 48824
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Dexin Sui
- Department of Biochemistry & Molecular Biology, Michigan State University, MI 48824
| | - Luca Sagresti
- Scuola Normale Superiore, Piazza dei Cavalieri 7, I-56126 Pisa, Italy and CSGI
- Istituto Nazionale di Fisica Nucleare (INFN) sezione di Pisa, Largo Bruno Pontecorvo 3, 56127 Pisa, Italy
| | - Kenneth M. Merz
- Department of Biochemistry & Molecular Biology, Michigan State University, MI 48824
- Department of Chemistry, Michigan State University, MI 48824
| | - Jian Hu
- Department of Biochemistry & Molecular Biology, Michigan State University, MI 48824
- Department of Chemistry, Michigan State University, MI 48824
| |
Collapse
|
13
|
Jiang Y, MacRenaris K, O'Halloran TV, Hu J. Determination of metal ion transport rate of human ZIP4 using stable zinc isotopes. J Biol Chem 2024; 300:107661. [PMID: 39128710 PMCID: PMC11630640 DOI: 10.1016/j.jbc.2024.107661] [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: 01/06/2024] [Revised: 07/28/2024] [Accepted: 08/01/2024] [Indexed: 08/13/2024] Open
Abstract
The essential microelement zinc is absorbed in the small intestine mainly by the zinc transporter ZIP4, a representative member of the Zrt/Irt-like protein (ZIP) family. ZIP4 is reportedly upregulated in many cancers, making it a promising oncology drug target. To date, there have been no reports on the turnover number of ZIP4, which is a crucial missing piece of information needed to better understand the transport mechanism. In this work, we used a nonradioactive zinc isotope, 70Zn, and inductively coupled plasma mass spectrometry to study human ZIP4 (hZIP4) expressed in Human embryonic kidney 293 cells. Our data showed that 70Zn can replace the radioactive 65Zn as a tracer in kinetic evaluation of hZIP4 activity. This approach, combined with the quantification of the cell surface expression of hZIP4 using biotinylation or surface-bound antibody, allowed us to estimate the apparent turnover number of hZIP4 to be in the range of 0.08 to 0.2 s-1. The turnover numbers of the truncated hZIP4 variants are significantly smaller than that of the full-length hZIP4, confirming a crucial role for the extracellular domain in zinc transport. Using 64Zn and 70Zn, we measured zinc efflux during the cell-based transport assay and found that it has little effect on the zinc import analysis under these conditions. Finally, we demonstrated that use of laser ablation inductively coupled plasma-TOF-mass spectrometry on samples applied to a solid substrate significantly increased the throughput of the transport assay. We envision that the approach reported here can be applied to the studies of metal transporters beyond the ZIP family.
Collapse
Affiliation(s)
- Yuhan Jiang
- Department of Chemistry, Michigan State University, East Lansing, Michigan, USA
| | - Keith MacRenaris
- Department of Chemistry, Michigan State University, East Lansing, Michigan, USA; Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA; Elemental Health Institute, Michigan State University, East Lansing, Michigan, USA; Quantitative Bio Element Analysis and Mapping (QBEAM) Center, Michigan State University, East Lansing, Michigan, USA
| | - Thomas V O'Halloran
- Department of Chemistry, Michigan State University, East Lansing, Michigan, USA; Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA; Elemental Health Institute, Michigan State University, East Lansing, Michigan, USA; Quantitative Bio Element Analysis and Mapping (QBEAM) Center, Michigan State University, East Lansing, Michigan, USA.
| | - Jian Hu
- Department of Chemistry, Michigan State University, East Lansing, Michigan, USA; Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, USA.
| |
Collapse
|
14
|
Li Y, Liu L. UKLF/PCBP2 axis governs the colorectal cancer development by transcriptionally activating SLC39A4. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119755. [PMID: 38768927 DOI: 10.1016/j.bbamcr.2024.119755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 04/27/2024] [Accepted: 05/07/2024] [Indexed: 05/22/2024]
Abstract
Colorectal cancer (CRC) is one of the most prevalent malignant tumors with limited treatment options. Therefore, there is an urgent need to investigate new therapeutic targets against CRC. Ubiquitous Kruppel-like factor (UKLF) is involved in various cancer processes, but its effect and detailed molecular mechanism in CRC are not yet fully understood. Here, this study aimed to investigate the function and mechanism of UKLF in the development of CRC. The results showed that UKLF was highly expressed in CRC tissues from clinical patients and its high expression was related to poor prognosis. UKLF promoted cell proliferation, migration and invasion, and inhibited cell apoptosis. The promotion effect of UKLF on tumor growth was further confirmed in vivo. As far as the mechanism was concerned, poly (C) binding protein 2 (PCBP2) was verified to bind to the 3'-UTR of UKLF mRNA and enhance its mRNA stability. Moreover, UKLF modulated the expression of solute carrier family 39 member 4 (SLC39A4) at the transcriptional level. Taken together, these findings elucidated the regulatory mechanism of UKLF and uncovered the importance of the PCBP2/UKLF/SLC39A4 pathway. The targeting of UKLF may be a novel direction for molecular-targeted CRC therapy.
Collapse
Affiliation(s)
- Yunze Li
- Department of General Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang 110004, China
| | - Lina Liu
- Department of General Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang 110004, China.
| |
Collapse
|
15
|
Acevedo S, Segovia MF, de la Fuente-Ortega E. Emerging Perspectives in Zinc Transporter Research in Prostate Cancer: An Updated Review. Nutrients 2024; 16:2026. [PMID: 38999774 PMCID: PMC11243615 DOI: 10.3390/nu16132026] [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: 05/09/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/14/2024] Open
Abstract
Dysregulation of zinc and zinc transporters families has been associated with the genesis and progression of prostate cancer. The prostate epithelium utilizes two types of zinc transporters, the ZIP (Zrt-, Irt-related Protein) and the ZnTs (Zinc Transporter), to transport zinc from the blood plasma to the gland lumen. ZIP transporters uptake zinc from extracellular space and organelle lumen, while ZnT transporters release zinc outside the cells or to organelle lumen. In prostate cancer, a commonly observed low zinc concentration in prostate tissue has been correlated with downregulations of certain ZIPs (e.g., ZIP1, ZIP2, ZIP3, ZIP14) and upregulations of specific ZnTs (e.g., ZnT1, ZnT9, ZnT10). These alterations may enable cancer cells to adapt to toxic high zinc levels. While zinc supplementation has been suggested as a potential therapy for this type of cancer, studies have yielded inconsistent results because some trials have indicated that zinc supplementation could exacerbate cancer risk. The reason for this discrepancy remains unclear, but given the high molecular and genetic variability present in prostate tumors, it is plausible that some zinc transporters-comprising 14 ZIP and 10 ZnT members-could be dysregulated in others patterns that promote cancer. From this perspective, this review highlights novel dysregulation, such as ZIP-Up/ZnT-Down, observed in prostate cancer cell lines for ZIP4, ZIP8, ZnT2, ZnT4, ZnT5, etc. Additionally, an in silico analysis of an available microarray from mouse models of prostate cancer (Nkx3.1;Pten) predicts similar dysregulation pattern for ZIP4, ZIP8, and ZnT2, which appear in early stages of prostate cancer progression. Furthermore, similar dysregulation patterns are supported by an in silico analysis of RNA-seq data from human cancer tumors available in cBioPortal. We discuss how these dysregulations of zinc transporters could impact zinc supplementation trials, particularly focusing on how the ZIP-Up/ZnT-Down dysregulation through various mechanisms might promote prostate cancer progression.
Collapse
Affiliation(s)
- Samantha Acevedo
- Laboratorio Estrés Celular y Enfermedades Crónicas No Transmisibles, Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Larrondo 1281, Coquimbo 1781421, Chile
| | - María Fernanda Segovia
- Laboratorio Estrés Celular y Enfermedades Crónicas No Transmisibles, Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Larrondo 1281, Coquimbo 1781421, Chile
| | - Erwin de la Fuente-Ortega
- Laboratorio Estrés Celular y Enfermedades Crónicas No Transmisibles, Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Larrondo 1281, Coquimbo 1781421, Chile
- Centro de Investigación y Desarrollo Tecnológico en Algas y Otros Recursos Biológicos (CIDTA), Facultad de Ciencias del Mar, Universidad Católica del Norte, Coquimbo 1781421, Chile
- Núcleo de Investigación en Prevención y Tratamiento de Enfermedades Crónicas no Transmisibles (NiPTEC), Universidad Católica del Norte, Coquimbo 1781421, Chile
| |
Collapse
|
16
|
Kim JA, Lee JK, Lee SY. Serum trace elements during treatment in pancreatic cancer patients and their associations with cancer prognosis. Clin Nutr 2024; 43:1459-1472. [PMID: 38714150 DOI: 10.1016/j.clnu.2024.04.012] [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: 07/31/2023] [Revised: 02/27/2024] [Accepted: 04/09/2024] [Indexed: 05/09/2024]
Abstract
BACKGROUND & AIMS In this study, we assessed serum trace element concentrations in patients with pancreatic cancer and compared the results to those of healthy controls and patients with chronic pancreatitis. We evaluated the association between trace element concentrations during cancer treatment and the risk of cancer progression and mortality in pancreatic cancer patients. METHODS A retrospective cohort study was conducted at a tertiary center in Korea. Serum trace element concentrations of cobalt (Co), copper (Cu), selenium (Se), and zinc (Zn) were measured at diagnosis using an inductively coupled plasma-mass spectrometry in 124 patients with pancreatic cancer, 50 patients with chronic pancreatitis, and 120 healthy controls. Trace elements were measured after a median of 282.5 (95% confidence interval [CI], 224.0-326.5) days from treatment initiation to assess changes in trace element concentrations during treatment. RESULTS Serum Co concentrations were significantly higher in patients with chronic pancreatitis and pancreatic cancer compared to healthy controls, while serum Se concentrations were significantly lower. During treatment, serum concentrations of Cu, Se, and Zn significantly decreased in patients with pancreatic cancer. During the follow-up (median 152.5; 95% CI, 142.8-160.0 months), 85.5% of patients experienced progression or relapse, and 84.7% of patients died. Patients with decreased Se and Zn concentrations during treatment had a higher mortality (hazard ratio [HR], 2.10; 95% CI, 1.31-3.38; P = 0.0020 for Se; HR, 1.72; 95% CI, 1.06-2.79; P = 0.0269 for Zn) compared to those with unchanged or increased trace element concentrations during treatment. Patients with a greater reduction in Zn concentrations during treatment had a higher mortality than those with a smaller reduction (HR, 1.59; 95% CI, 1.01-2.52; P = 0.0483). Patients whose Zn status changed from normal to deficient during treatment had an increased mortality (HR, 1.76; 95% CI, 1.16-2.67, P = 0.0084). Patients with multiple (≥2) trace element deficiencies after treatment had poorer outcomes than those with no or single trace element deficiency. CONCLUSIONS This study revealed that decreases in Se and Zn concentrations during cancer treatment were associated with adverse outcomes in terms of cancer progression and mortality in patients with pancreatic cancer. Further prospective investigations are recommended.
Collapse
Affiliation(s)
- Jee Ah Kim
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, South Korea; Department of Laboratory Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, 29 Saemunan-ro, Jongno-gu, Seoul 03181, South Korea
| | - Jong Kyun Lee
- Division of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, South Korea.
| | - Soo-Youn Lee
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, South Korea.
| |
Collapse
|
17
|
Liu M, Ren Y, Zhou Z, Yang J, Shi X, Cai Y, Arreola AX, Luo W, Fung KM, Xu C, Nipp RD, Bronze MS, Zheng L, Li YP, Houchen CW, Zhang Y, Li M. The crosstalk between macrophages and cancer cells potentiates pancreatic cancer cachexia. Cancer Cell 2024; 42:885-903.e4. [PMID: 38608702 PMCID: PMC11162958 DOI: 10.1016/j.ccell.2024.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 12/18/2023] [Accepted: 03/15/2024] [Indexed: 04/14/2024]
Abstract
With limited treatment options, cachexia remains a major challenge for patients with cancer. Characterizing the interplay between tumor cells and the immune microenvironment may help identify potential therapeutic targets for cancer cachexia. Herein, we investigate the critical role of macrophages in potentiating pancreatic cancer induced muscle wasting via promoting TWEAK (TNF-like weak inducer of apoptosis) secretion from the tumor. Specifically, depletion of macrophages reverses muscle degradation induced by tumor cells. Macrophages induce non-autonomous secretion of TWEAK through CCL5/TRAF6/NF-κB pathway. TWEAK promotes muscle atrophy by activating MuRF1 initiated muscle remodeling. Notably, tumor cells recruit and reprogram macrophages via the CCL2/CCR2 axis and disrupting the interplay between macrophages and tumor cells attenuates muscle wasting. Collectively, this study identifies a feedforward loop between pancreatic cancer cells and macrophages, underlying the non-autonomous activation of TWEAK secretion from tumor cells thereby providing promising therapeutic targets for pancreatic cancer cachexia.
Collapse
Affiliation(s)
- Mingyang Liu
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Yu Ren
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Zhijun Zhou
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Jingxuan Yang
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Xiuhui Shi
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Yang Cai
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Alex X Arreola
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Wenyi Luo
- Department of Pathology, Yale School of Medicine, New Haven, CT 06519, USA
| | - Kar-Ming Fung
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Chao Xu
- Department of Biostatistics and Epidemiology, Hudson College of Public Health, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Ryan D Nipp
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Michael S Bronze
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Lei Zheng
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Yi-Ping Li
- Department of Integrative Biology & Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Courtney W Houchen
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Yuqing Zhang
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| | - Min Li
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
18
|
Chen B, Yu P, Chan WN, Xie F, Zhang Y, Liang L, Leung KT, Lo KW, Yu J, Tse GMK, Kang W, To KF. Cellular zinc metabolism and zinc signaling: from biological functions to diseases and therapeutic targets. Signal Transduct Target Ther 2024; 9:6. [PMID: 38169461 PMCID: PMC10761908 DOI: 10.1038/s41392-023-01679-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 09/15/2023] [Accepted: 10/10/2023] [Indexed: 01/05/2024] Open
Abstract
Zinc metabolism at the cellular level is critical for many biological processes in the body. A key observation is the disruption of cellular homeostasis, often coinciding with disease progression. As an essential factor in maintaining cellular equilibrium, cellular zinc has been increasingly spotlighted in the context of disease development. Extensive research suggests zinc's involvement in promoting malignancy and invasion in cancer cells, despite its low tissue concentration. This has led to a growing body of literature investigating zinc's cellular metabolism, particularly the functions of zinc transporters and storage mechanisms during cancer progression. Zinc transportation is under the control of two major transporter families: SLC30 (ZnT) for the excretion of zinc and SLC39 (ZIP) for the zinc intake. Additionally, the storage of this essential element is predominantly mediated by metallothioneins (MTs). This review consolidates knowledge on the critical functions of cellular zinc signaling and underscores potential molecular pathways linking zinc metabolism to disease progression, with a special focus on cancer. We also compile a summary of clinical trials involving zinc ions. Given the main localization of zinc transporters at the cell membrane, the potential for targeted therapies, including small molecules and monoclonal antibodies, offers promising avenues for future exploration.
Collapse
Affiliation(s)
- Bonan Chen
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Peiyao Yu
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Wai Nok Chan
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Fuda Xie
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Yigan Zhang
- Institute of Biomedical Research, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Li Liang
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Kam Tong Leung
- Department of Pediatrics, The Chinese University of Hong Kong, Hong Kong, China
| | - Kwok Wai Lo
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Jun Yu
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Gary M K Tse
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China.
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China.
| | - Ka Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
19
|
Forte G, Pisano A, Bocca B, Fenu G, Farace C, Etzi F, Perra T, Sabalic A, Porcu A, Madeddu R. Toxic Metal and Essential Element Concentrations in the Blood and Tissues of Pancreatic Ductal Adenocarcinoma Patients. TOXICS 2024; 12:32. [PMID: 38250988 PMCID: PMC10818929 DOI: 10.3390/toxics12010032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 01/23/2024]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive lethal neoplasm, and it has an average 5-year survival rate of less than 10%. Although the factors that influence PDAC development remain unclear, exposure to toxic metals or the imbalance in essential elements may have a role in PDAC-associated metabolic pathways. METHODS This study determined the concentrations of Cd, Cr, Cu, Fe, Mn, Ni, Pb, Se and Zn in whole blood, cancer and non-cancer tissues of patients affected by PDAC, and compared them with levels in healthy controls using inductively coupled plasma mass spectrometry. RESULTS Results of the whole blood showed significantly higher levels of Cr, Cu and Cu/Zn ratio in PDAC patients compared to the controls. In addition, the concentrations of Cu, Se, Fe and Zn significantly increased in cancer tissue compared to the healthy counterparts. CONCLUSIONS This study revealed evidence of altered metal levels in the blood and pancreatic tissues of PDAC patients with respect to healthy controls. These changes may contribute to multiple mechanisms involved in metal-induced carcinogenesis, including oxidative stress, DNA damage, genetic alteration, decreased antioxidant barriers and inflammatory responses. Thus, the analysis of metals can be used in the diagnosis and monitoring of PDAC neoplasms.
Collapse
Affiliation(s)
- Giovanni Forte
- Department of Environment and Health, Italian National Institute of Health, 00161 Rome, Italy;
| | - Andrea Pisano
- Department of Biomedical Science—Histology, University of Sassari, 07100 Sassari, Italy; (A.P.); (G.F.); (C.F.); (F.E.); (A.S.); (R.M.)
| | - Beatrice Bocca
- Department of Environment and Health, Italian National Institute of Health, 00161 Rome, Italy;
| | - Grazia Fenu
- Department of Biomedical Science—Histology, University of Sassari, 07100 Sassari, Italy; (A.P.); (G.F.); (C.F.); (F.E.); (A.S.); (R.M.)
| | - Cristiano Farace
- Department of Biomedical Science—Histology, University of Sassari, 07100 Sassari, Italy; (A.P.); (G.F.); (C.F.); (F.E.); (A.S.); (R.M.)
- National Institute of Biostructures and Biosystems, Interuniversity Consortium INBB, 00136 Rome, Italy
| | - Federica Etzi
- Department of Biomedical Science—Histology, University of Sassari, 07100 Sassari, Italy; (A.P.); (G.F.); (C.F.); (F.E.); (A.S.); (R.M.)
| | - Teresa Perra
- Department of Medicine, Surgery and Pharmacy, Unit of General Surgery, University of Sassari, 07100 Sassari, Italy; (T.P.); (A.P.)
| | - Angela Sabalic
- Department of Biomedical Science—Histology, University of Sassari, 07100 Sassari, Italy; (A.P.); (G.F.); (C.F.); (F.E.); (A.S.); (R.M.)
| | - Alberto Porcu
- Department of Medicine, Surgery and Pharmacy, Unit of General Surgery, University of Sassari, 07100 Sassari, Italy; (T.P.); (A.P.)
| | - Roberto Madeddu
- Department of Biomedical Science—Histology, University of Sassari, 07100 Sassari, Italy; (A.P.); (G.F.); (C.F.); (F.E.); (A.S.); (R.M.)
- National Institute of Biostructures and Biosystems, Interuniversity Consortium INBB, 00136 Rome, Italy
| |
Collapse
|
20
|
Zhang X, Hou Y, Huang Y, Chen W, Zhang H. Interplay between zinc and cell proliferation and implications for the growth of livestock. J Anim Physiol Anim Nutr (Berl) 2023; 107:1402-1418. [PMID: 37391879 DOI: 10.1111/jpn.13851] [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: 12/15/2022] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 07/02/2023]
Abstract
Zinc (Zn) plays a critical role in the growth of livestock, which depends on cell proliferation. In addition to modifying the growth associated with its effects on food intake, mitogenic hormones, signal transduction and gene transcription, Zn also regulates body weight gain through mediating cell proliferation. Zn deficiency in animals leads to growth inhibition, along with an arrest of cell cycle progression at G0/G1 and S phase due to depression in the expression of cyclin D/E and DNA synthesis. Therefore, in the present study, the interplay between Zn and cell proliferation and implications for the growth of livestock were reviewed, in which Zn regulates cell proliferation in several ways, especially cell cycle progression at the G0/G1 phase DNA synthesis and mitosis. During the cell cycle, the Zn transporters and major Zn binding proteins such as metallothioneins are altered with the requirements of cellular Zn level and nuclear translocation of Zn. In addition, calcium signaling, MAPK pathway and PI3K/Akt cascades are also involved in the process of Zn-interfering cell proliferation. The evidence collected over the last decade highlights the necessity of Zn for normal cell proliferation, which suggests Zn supplementation should be considered for the growth and health of poultry.
Collapse
Affiliation(s)
- Xiangli Zhang
- College of Animal Science and Technology, Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, Henan Agricultural University, Zhengzhou, China
| | - Yuhuang Hou
- Laboratory for Animal Nutrition and Animal Product Quality, Department of Animal Sciences and Aquatic Ecology, Ghent University, Ghent, Belgium
| | - Yanqun Huang
- College of Animal Science and Technology, Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, Henan Agricultural University, Zhengzhou, China
| | - Wen Chen
- College of Animal Science and Technology, Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, Henan Agricultural University, Zhengzhou, China
| | - Huaiyong Zhang
- College of Animal Science and Technology, Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, Henan Agricultural University, Zhengzhou, China
- Laboratory for Animal Nutrition and Animal Product Quality, Department of Animal Sciences and Aquatic Ecology, Ghent University, Ghent, Belgium
| |
Collapse
|
21
|
Lim B, Kim KS, Na K. pH-Responsive Zinc Ion Regulating Immunomodulatory Nanoparticles for Effective Cancer Immunotherapy. Biomacromolecules 2023; 24:4263-4273. [PMID: 37616157 DOI: 10.1021/acs.biomac.3c00557] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Herein, we introduce a novel approach involving the utilization of a human serum albumin-coated zeolite imidazolate framework-8 containing a photosensitizer (HPZ) that exhibits targeted recognition of the tumor microenvironment, enabling the rapid elevation of zinc ion concentrations while facilitating the controlled release of an encapsulated photosensitizer (PS). At a physiological pH of 7.4, HPZ demonstrates a size of approximately 170 nm, significantly decreasing to less than 10 nm under pH 6.5 acidic conditions. Acid-induced decomposition of HPZ triggers a rapid increase in zinc ion concentration, eliciting potent cytotoxic effects against colorectal, breast, and pancreatic cancers. Additionally, upon laser irradiation, the encapsulated PS within HPZ initiates the generation of reactive oxygen species, synergistically augmenting the cytotoxicity induced by zinc ions. Intravenous administration of HPZ in a CT26 tumor-bearing mouse model resulted in a notable expansion of CD3+CD4+ helper T cells and CD3+CD8+ cytotoxic T cells, accompanied by a reduction in the CD4+CD25+Foxp3+ regulatory T-cell population. These changes led to significant inhibition of tumor growth, highlighting the efficacy of HPZ in this experimental model. Importantly, HPZ exhibits favorable safety characteristics, displaying no toxicity toward vital organs and inducing no weight loss. Thus, HPZ holds immense promise as a standalone treatment or in combination with diverse anticancer immunotherapies, underscoring its potential in the field of anticancer immunotherapy.
Collapse
Affiliation(s)
- Byoungjun Lim
- Department of BioMedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Kyoung Sub Kim
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Kun Na
- Department of BioMedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| |
Collapse
|
22
|
Figiel M, Górka AK, Górecki A. Zinc Ions Modulate YY1 Activity: Relevance in Carcinogenesis. Cancers (Basel) 2023; 15:4338. [PMID: 37686614 PMCID: PMC10487186 DOI: 10.3390/cancers15174338] [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: 07/27/2023] [Revised: 08/17/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
YY1 is widely recognized as an intrinsically disordered transcription factor that plays a role in development of many cancers. In most cases, its overexpression is correlated with tumor progression and unfavorable patient outcomes. Our latest research focusing on the role of zinc ions in modulating YY1's interaction with DNA demonstrated that zinc enhances the protein's multimeric state and affinity to its operator. In light of these findings, changes in protein concentration appear to be just one element relevant to modulating YY1-dependent processes. Thus, alterations in zinc ion concentration can directly and specifically impact the regulation of gene expression by YY1, in line with reports indicating a correlation between zinc ion levels and advancement of certain tumors. This review concentrates on other potential consequences of YY1 interaction with zinc ions that may act by altering charge distribution, conformational state distribution, or oligomerization to influence its interactions with molecular partners that can disrupt gene expression patterns.
Collapse
Affiliation(s)
| | | | - Andrzej Górecki
- Faculty of Biochemistry, Biophysics and Biotechnology, Department of Physical Biochemistry, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.F.); (A.K.G.)
| |
Collapse
|
23
|
Marin-Muller C, Li D, Lü JM, Liang Z, Vega-Martínez O, Crawford SE, Estes MK, Fisher WE, Chen C, Yao Q. Nanoparticle-Mediated Therapy with miR-198 Sensitizes Pancreatic Cancer to Gemcitabine Treatment through Downregulation of VCP-Mediated Autophagy. Pharmaceutics 2023; 15:2038. [PMID: 37631252 PMCID: PMC10457905 DOI: 10.3390/pharmaceutics15082038] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/18/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains an extremely aggressive disease characterized by rapidly acquired multi-drug resistance, including to first-line chemotherapeutic agent gemcitabine. Autophagy is a process that is often exploited by cancer and is one of several intrinsic factors associated with resistance to gemcitabine. We have previously found that miR-198 acts as a tumor suppressor in PDAC through the targeting of factors including Valosin-containing protein (VCP). VCP has been reported to play an important role in autophagic flux. In this study, we investigated whether the repression of VCP through miR-198 administration disrupts the autophagy process and sensitizes PDAC cells to gemcitabine treatment in vitro. Moreover, we used LGA-PEI (LPNP) nanoparticles to effectively administer miR-198 to tumors in vivo, inducing tumor sensitization to gemcitabine and leading to a significant reduction in tumor burden and metastases and a concomitant downregulation of VCP expression and autophagy maturation. Our results indicate a potential therapeutic strategy for targeting gemcitabine resistant PDAC and establishes the use of LPNPs for effective therapeutic delivery of nucleic acids in vitro and in vivo.
Collapse
Affiliation(s)
- Christian Marin-Muller
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (C.M.-M.)
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- Speratum Biopharma, Inc., Dover, DE 19901, USA
| | - Dali Li
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (C.M.-M.)
| | - Jian-Ming Lü
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (C.M.-M.)
| | - Zhengdong Liang
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (C.M.-M.)
| | | | - Sue E. Crawford
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mary K. Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - William E. Fisher
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (C.M.-M.)
| | - Changyi Chen
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (C.M.-M.)
| | - Qizhi Yao
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (C.M.-M.)
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, TX 77030, USA
| |
Collapse
|
24
|
Liu Y, Bafaro EM, Dempski RE. Single-molecule quantification of the oligomeric state of ZIP transporters in mammalian cells with fluorescence correlation spectroscopy. Methods Enzymol 2023; 687:103-137. [PMID: 37666629 DOI: 10.1016/bs.mie.2023.04.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
The SLC39 family of transporters, otherwise known as ZIPs for Zrt and Irt-like Proteins, function to increase cytosolic levels of transition metals. ZIP transporters have been identified at all phylogenetic levels and are members of the SoLute Carrier (SLC) superfamily. There are fourteen ZIP transporters encoded in the human genome. ZIP transmembrane proteins are expressed in the plasma membrane or membranes of intracellular organelles and have unique expression profiles across cell types. While direct structural efforts including x-ray crystallography, NMR and ab initio approaches have been effective tools in elucidating the structure of ZIPs, direct elucidation of the oligomeric state of these proteins is essential in understanding how wild type ZIP proteins function and whether mutations alter the oligomeric state of ZIPs. Unfortunately, several tools to quantify oligomeric states of proteins require overexpression of proteins which can lead to artifacts in experimental results. In contrast, fluorescence correlation spectroscopy (FCS) is a single-molecule technique which can be used to quantify the oligomeric state of transmembrane proteins. FCS takes advantage of the observation that the molecular brightness of a cluster of fluorescent molecules is directly proportional to the number of fluorescent molecules within the protein complex. This chapter describes how to implement FCS, focused on ZIP transporters, to quantify the oligomeric state of transmembrane in vivo. Included within this chapter are procedures to design constructs for experiments, transfection of mammalian cells as well as data acquisition and analysis. Taken together, FCS is a powerful mechanism to investigate the oligomeric state of proteins embedded within membranes of cells.
Collapse
Affiliation(s)
- Yuting Liu
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, MA, United States
| | - Elizabeth M Bafaro
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, MA, United States
| | - Robert E Dempski
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, MA, United States.
| |
Collapse
|
25
|
Zhang T, Zhang Y, Sui D, Hu J. High-resolution structure of a mercury cross-linked ZIP metal transporter reveals delicate motions and metal relay for regulated zinc transport. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.20.537755. [PMID: 37131590 PMCID: PMC10153219 DOI: 10.1101/2023.04.20.537755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Zrt-/Irt-like protein (ZIP) divalent metal transporters play a central role in maintaining trace element homeostasis. The prototypical ZIP from Bordetella bronchiseptica (BbZIP) is an elevator-type transporter, but the dynamic motions and detailed transport mechanism remain to be elucidated. Here, we report a high-resolution crystal structure of a mercury-crosslinked BbZIP variant at 1.95 Å, revealing an upward rotation of the transport domain in the new inward-facing conformation and a water-filled metal release channel that is divided into two parallel pathways by the previously disordered cytoplasmic loop. Mutagenesis and transport assays indicated that the newly identified high-affinity metal binding site in the primary pathway acts as a "metal sink" to reduce the transport rate. The discovery of a hinge motion around an extracellular axis allowed us to propose a sequential hinge-elevator-hinge movement of the transport domain to achieve alternating access. These findings provide key insights into the transport mechanisms and activity regulation.
Collapse
Affiliation(s)
- Tuo Zhang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
- Department of Biochemistry & Molecular Biology, Michigan State University, MI 48824
| | - Yao Zhang
- Department of Biochemistry & Molecular Biology, Michigan State University, MI 48824
| | - Dexin Sui
- Department of Biochemistry & Molecular Biology, Michigan State University, MI 48824
| | - Jian Hu
- Department of Biochemistry & Molecular Biology, Michigan State University, MI 48824
- Department of Chemistry, Michigan State University, MI 48824
| |
Collapse
|
26
|
Emerging Mechanisms of Skeletal Muscle Homeostasis and Cachexia: The SUMO Perspective. Cells 2023; 12:cells12040644. [PMID: 36831310 PMCID: PMC9953977 DOI: 10.3390/cells12040644] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023] Open
Abstract
Mobility is an intrinsic feature of the animal kingdom that stimulates evolutionary processes and determines the biological success of animals. Skeletal muscle is the primary driver of voluntary movements. Besides, skeletal muscles have an immense impact on regulating glucose, amino acid, and lipid homeostasis. Muscle atrophy/wasting conditions are accompanied by a drastic effect on muscle function and disrupt steady-state muscle physiology. Cachexia is a complex multifactorial muscle wasting syndrome characterized by extreme loss of skeletal muscle mass, resulting in a dramatic decrease in life quality and reported mortality in more than 30% of patients with advanced cancers. The lack of directed treatments to prevent or relieve muscle loss indicates our inadequate knowledge of molecular mechanisms involved in muscle cell organization and the molecular etiology of cancer-induced cachexia (CIC). This review highlights the latest knowledge of regulatory mechanisms involved in maintaining muscle function and their deregulation in wasting syndromes, particularly in cachexia. Recently, protein posttranslational modification by the small ubiquitin-like modifier (SUMO) has emerged as a key regulatory mechanism of protein function with implications for different aspects of cell physiology and diseases. We also review an atypical association of SUMO-mediated pathways in this context and deliberate on potential treatment strategies to alleviate muscle atrophy.
Collapse
|
27
|
Li N, Zhou H, Holden VK, Deepak J, Dhilipkannah P, Todd NW, Stass SA, Jiang F. Streptococcus pneumoniae promotes lung cancer development and progression. iScience 2023; 26:105923. [PMID: 36685035 PMCID: PMC9852931 DOI: 10.1016/j.isci.2022.105923] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 11/12/2022] [Accepted: 12/30/2022] [Indexed: 01/06/2023] Open
Abstract
Streptococcus pneumoniae (SP) is associated with lung cancer, yet its role in the tumorigenesis remains uncertain. Herein we find that SP attaches to lung cancer cells via binding pneumococcal surface protein C (PspC) to platelet-activating factor receptor (PAFR). Interaction between PspC and PAFR stimulates cell proliferation and activates PI3K/AKT and nuclear factor kB (NF-kB) signaling pathways, which trigger a pro-inflammatory response. Lung cancer cells infected with SP form larger tumors in BALB/C mice compared to untreated cells. Mice treated with tobacco carcinogen and SP develop more lung tumors and had shorter survival period than mice treated with the carcinogen alone. Mutating PspC or PAFR abolishes tumor-promoting effects of SP. Overabundance of SP is associated with the survival. SP may play a driving role in lung tumorigenesis by activating PI3K/AKT and NF-kB pathways via binding PspC to PAFR and provide a microbial target for diagnosis and treatment of the disease.
Collapse
Affiliation(s)
- Ning Li
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Huifen Zhou
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Van K. Holden
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Janaki Deepak
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Pushpa Dhilipkannah
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Nevins W. Todd
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sanford A. Stass
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Feng Jiang
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
28
|
Zhang Y, Jiang Y, Gao K, Sui D, Yu P, Su M, Wei GW, Hu J. Structural insights into the elevator-type transport mechanism of a bacterial ZIP metal transporter. Nat Commun 2023; 14:385. [PMID: 36693843 PMCID: PMC9873690 DOI: 10.1038/s41467-023-36048-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 01/13/2023] [Indexed: 01/26/2023] Open
Abstract
The Zrt-/Irt-like protein (ZIP) family consists of ubiquitously expressed divalent metal transporters critically involved in maintaining systemic and cellular homeostasis of zinc, iron, and manganese. Here, we present a study on a prokaryotic ZIP from Bordetella bronchiseptica (BbZIP) by combining structural biology, evolutionary covariance, computational modeling, and a variety of biochemical assays to tackle the issue of the transport mechanism which has not been established for the ZIP family. The apo state structure in an inward-facing conformation revealed a disassembled transport site, altered inter-helical interactions, and importantly, a rigid body movement of a 4-transmembrane helix (TM) bundle relative to the other TMs. The computationally generated and biochemically validated outward-facing conformation model revealed a slide of the 4-TM bundle, which carries the transport site(s), by approximately 8 Å toward the extracellular side against the static TMs which mediate dimerization. These findings allow us to conclude that BbZIP is an elevator-type transporter.
Collapse
Affiliation(s)
- Yao Zhang
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Yuhan Jiang
- Department of Chemistry, Michigan State University, East Lansing, MI, USA
| | - Kaifu Gao
- Department of Mathematics, Michigan State University, East Lansing, MI, USA
| | - Dexin Sui
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Peixuan Yu
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Min Su
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Guo-Wei Wei
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
- Department of Mathematics, Michigan State University, East Lansing, MI, USA
| | - Jian Hu
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA.
- Department of Chemistry, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
29
|
Willekens J, Runnels LW. Impact of Zinc Transport Mechanisms on Embryonic and Brain Development. Nutrients 2022; 14:2526. [PMID: 35745255 PMCID: PMC9231024 DOI: 10.3390/nu14122526] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/09/2022] [Accepted: 06/09/2022] [Indexed: 12/04/2022] Open
Abstract
The trace element zinc (Zn) binds to over ten percent of proteins in eukaryotic cells. Zn flexible chemistry allows it to regulate the activity of hundreds of enzymes and influence scores of metabolic processes in cells throughout the body. Deficiency of Zn in humans has a profound effect on development and in adults later in life, particularly in the brain, where Zn deficiency is linked to several neurological disorders. In this review, we will summarize the importance of Zn during development through a description of the outcomes of both genetic and early dietary Zn deficiency, focusing on the pathological consequences on the whole body and brain. The epidemiology and the symptomology of Zn deficiency in humans will be described, including the most studied inherited Zn deficiency disease, Acrodermatitis enteropathica. In addition, we will give an overview of the different forms and animal models of Zn deficiency, as well as the 24 Zn transporters, distributed into two families: the ZIPs and the ZnTs, which control the balance of Zn throughout the body. Lastly, we will describe the TRPM7 ion channel, which was recently shown to contribute to intestinal Zn absorption and has its own significant impact on early embryonic development.
Collapse
Affiliation(s)
| | - Loren W. Runnels
- Department of Pharmacology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA;
| |
Collapse
|
30
|
Barman SK, Zaman MS, Veljanoski F, Malladi CS, Mahns DA, Wu MJ. Expression profiles of the genes associated with zinc homeostasis in normal and cancerous breast and prostate cells. Metallomics 2022; 14:6601457. [PMID: 35657662 DOI: 10.1093/mtomcs/mfac038] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/12/2022] [Indexed: 11/14/2022]
Abstract
Zn2+ dyshomeostasis is an intriguing phenomenon in breast and prostate cancers, with breast cancer cells exhibiting higher intracellular Zn2+ level compared to their corresponding normal epithelial cells, in contrast to the low Zn2+ level in prostate cancer cells. In order to gain molecular insights into the zinc homeostasis of breast and prostate cancer cells, this study profiled the expression of 28 genes, including 14 zinc importer genes (SLC39A1-14) which encode ZIP1-14 to transport Zn2+ into the cytoplasm, 10 zinc exporter genes (SLC30A1-10) which encode ZnT1-10 to transport Zn2+ out of the cytoplasm and 4 metallothionein genes (MT1B, MT1F, MT1X, MT2A) in breast (MCF10A, MCF-7, MDA-MB-231) and prostate (RWPE-1, PC3, DU145) cell lines in response to extracellular zinc exposures at a mild cytotoxic dosage and a benign dosage. The RNA samples were prepared at 0 min (T0), 30 min (T30) and 120 min (T120) in a time course with or without zinc exposure, which were used for profiling the baseline and dynamic gene expression. The up-regulation of MT genes was observed across the breast and prostate cancer cell lines. The expression landscape of SLC39A and SLC30A was revealed by the qRT-PCR data of this study, which sheds light on the divergence of intracellular Zn2+ levels for breast and prostate cancer cells. Taken together, the findings are valuable in unravelling the molecular intricacy of zinc homeostasis in breast and prostate cancer cells.
Collapse
Affiliation(s)
- Shital K Barman
- School of Science, Western Sydney University, Locked Bag 1797, Penrith, NSW 2751
| | - Mohammad S Zaman
- School of Science, Western Sydney University, Locked Bag 1797, Penrith, NSW 2751
| | - Filip Veljanoski
- School of Science, Western Sydney University, Locked Bag 1797, Penrith, NSW 2751
| | - Chandra S Malladi
- Proteomics and Lipidomics Lab, School of Medicine, Western Sydney University, Locked
| | - David A Mahns
- School of Medicine, Western Sydney University, Locked Bag 1797, Penrith, NSW 2751
| | - Ming J Wu
- School of Science, Western Sydney University, Locked Bag 1797, Penrith, NSW 2751
| |
Collapse
|
31
|
Shi X, Yang J, Liu M, Zhang Y, Zhou Z, Luo W, Fung KM, Xu C, Bronze MS, Houchen CW, Li M. Circular RNA ANAPC7 Inhibits Tumor Growth and Muscle Wasting via PHLPP2-AKT-TGF-β Signaling Axis in Pancreatic Cancer. Gastroenterology 2022; 162:2004-2017.e2. [PMID: 35176309 PMCID: PMC10428768 DOI: 10.1053/j.gastro.2022.02.017] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/26/2022] [Accepted: 02/09/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Pancreatic cancer has the highest prevalence of cancer-associated cachexia among all cancers. ZIP4 promotes pancreatic cancer progression by regulating oncogenic miR-373, and perturbation of circular RNAs (circRNAs) is associated with cancer aggressiveness. This study aimed to identify circRNAs involved in ZIP4/miR-373-driven cancer growth and cachexia and decipher the underlying mechanism. METHODS Differentially expressed circRNAs and potential targets of microRNA were identified through in silico analysis. The RNA interactions were determined by means of biotinylated microRNA pulldown, RNA immunoprecipitation, and luciferase reporter assays. The function of circRNA in ZIP4-miR-373 signaling axis were examined in human pancreatic cancer cells, 3-dimensional spheroids and organoids, mouse models, and clinical specimens. Mouse skeletal muscles were analyzed by means of histology. RESULTS We identified circANAPC7 as a sponge for miR-373, which inhibited tumor growth and muscle wasting in vitro and in vivo. Mechanistic studies showed that PHLPP2 is a downstream target of ZIP4/miR-373. CircANAPC7 functions through PHLPP2-mediated dephosphorylation of AKT, thus suppressing cancer cell proliferation by down-regulating cyclin D1 and inhibiting muscle wasting via decreasing the secretion of transforming growth factor-β through STAT5. We further demonstrated that PHLPP2 induced dephosphorylation of CREB, a zinc-dependent transcription factor activated by ZIP4, thereby forming a CREB-miR-373-PHLPP2 feed-forward loop to regulate tumor progression and cancer cachexia. CONCLUSION This study identified circANAPC7 as a novel tumor suppressor, which functions through the CREB-miR-373-PHLPP2 axis, leading to AKT dephosphorylation, and cyclin D1 and transforming growth factor-β down-regulation to suppress tumor growth and muscle wasting in pancreatic cancer.
Collapse
Affiliation(s)
- Xiuhui Shi
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Jingxuan Yang
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Mingyang Liu
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Yuqing Zhang
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Zhijun Zhou
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Wenyi Luo
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Kar-Ming Fung
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Chao Xu
- Department of Biostatistics and Epidemiology, Hudson College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Michael S Bronze
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Courtney W Houchen
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Min Li
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma.
| |
Collapse
|
32
|
Heterologous Expression of Full-Length and Truncated Human ZIP4 Zinc Transporter in Saccharomyces cerevisiae. Biomolecules 2022; 12:biom12050726. [PMID: 35625653 PMCID: PMC9138318 DOI: 10.3390/biom12050726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 02/01/2023] Open
Abstract
The human (h) transporter hZIP4 is the primary Zn2+ importer in the intestine. hZIP4 is also expressed in a variety of organs such as the pancreas and brain. Dysfunction of hZIP4 can result in the Zn2+ deficiency disease acrodermatitis enteropathica (AE). AE can disrupt digestive and immune system homeostasis. A limited number of hZIP4 expression strategies have hindered increasing knowledge about this essential transmembrane protein. Here, we report the heterologous expression of hZIP4 in Saccharomyces cerevisiae. Both a wild-type and a mutant S. cerevisiae strain, in which the endogenous Zn2+ transporters were deleted, were used to test the expression and localization of an hZIP4–GFP fusion protein. A full-length hZIP4–GFP and a truncated membrane-domain-only (mhZIP4–GFP) protein were observed to be present in the plasma membrane in yeast.
Collapse
|
33
|
Sophisticated expression responses of ZNT1 and MT in response to changes in the expression of ZIPs. Sci Rep 2022; 12:7334. [PMID: 35513474 PMCID: PMC9072671 DOI: 10.1038/s41598-022-10925-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 04/15/2022] [Indexed: 12/16/2022] Open
Abstract
The zinc homeostatic proteins Zn transporter 1 (ZNT1) and metallothionein (MT) function in dampening increases in cytosolic zinc concentrations. Conversely, the expression of ZNT1 and MT is expected to be suppressed during decreases in cytosolic zinc concentrations. Thus, ZNT1/MT homeostatic responses are considered to be essential for maintaining cellular zinc homeostasis because cellular zinc concentrations are readily altered by changes in the expression of several Zrt-/Irt-like proteins (ZIPs) under both physiological and pathological conditions. However, this notion remains to be tested experimentally. Here, we investigated the aforementioned homeostatic process by analyzing ZNT1 and MT protein expression in response to ZIP expression. Overexpression of cell-surface-localized ZIPs, such as ZIP4 and ZIP5, increased the cellular zinc content, which caused an increase in the expression of cell-surface ZNT1 and cytosolic MT in the absence of zinc supplementation in the culture medium. By contrast, elimination of the overexpressed ZIP4 and ZIP5 resulted in decreased expression of ZNT1 but not MT, which suggests that differential regulation of ZNT1 and MT expression at the protein level underlies the homeostatic responses necessary for zinc metabolism under certain conditions. Moreover, increased expression of apically localized ZIP4 facilitated basolateral ZNT1 expression in polarized cells, which indicates that such a coordinated expression mechanism is crucial for vectorial transcellular transport. Our results provide novel insights into the physiological maintenance of cellular zinc homeostasis in response to alterations in cytosolic zinc concentrations caused by changes in the expression of ZIPs.
Collapse
|
34
|
Upper Gastrointestinal Cancer and Liver Cirrhosis. Cancers (Basel) 2022; 14:cancers14092269. [PMID: 35565397 PMCID: PMC9105927 DOI: 10.3390/cancers14092269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/26/2022] [Accepted: 04/29/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary There is a higher incidence rate of upper gastrointestinal cancer in those with liver cirrhosis. The contributing factors include gastric ulcers, congestive gastropathy, zinc deficiency, alcohol drinking, tobacco use and gut microbiota. Most of the de novo malignancies that develop after liver transplantation for cirrhotic patients are upper gastrointestinal cancers. The surgical risk of upper gastrointestinal cancers in cirrhotic patients with advanced liver cirrhosis is higher. Abstract The extended scope of upper gastrointestinal cancer can include esophageal cancer, gastric cancer and pancreatic cancer. A higher incidence rate of gastric cancer and esophageal cancer in patients with liver cirrhosis has been reported. It is attributable to four possible causes which exist in cirrhotic patients, including a higher prevalence of gastric ulcers and congestive gastropathy, zinc deficiency, alcohol drinking and tobacco use and coexisting gut microbiota. Helicobacter pylori infection enhances the development of gastric cancer. In addition, Helicobacter pylori, Porphyromonas gingivalis and Aggregatibacter actinomycetemcomitans also contribute to the development of pancreatic cancer in cirrhotic patients. Cirrhotic patients (especially those with alcoholic liver cirrhosis) who undergo liver transplantation have a higher overall risk of developing de novo malignancies. Most de novo malignancies are upper gastrointestinal malignancies. The prognosis is usually poor. Considering the surgical risk of upper gastrointestinal cancer among those with liver cirrhosis, a radical gastrectomy with D1 or D2 lymph node dissection can be undertaken in Child class A patients. D1 lymph node dissection can be performed in Child class B patients. Endoscopic submucosal dissection for gastric cancer or esophageal cancer can be undertaken safely in selected cirrhotic patients. In Child class C patients, a radical gastrectomy is potentially fatal. Pancreatic radical surgery should be avoided in those with liver cirrhosis with Child class B or a MELD score over 15. The current review focuses on the recent reports on some factors in liver cirrhosis that contribute to the development of upper gastrointestinal cancer. Quitting alcohol drinking and tobacco use is important. How to decrease the risk of the development of gastrointestinal cancer in those with liver cirrhosis remains a challenging problem.
Collapse
|
35
|
Ha JH, Prela O, Carpizo DR, Loh SN. p53 and Zinc: A Malleable Relationship. Front Mol Biosci 2022; 9:895887. [PMID: 35495631 PMCID: PMC9043292 DOI: 10.3389/fmolb.2022.895887] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 03/30/2022] [Indexed: 12/14/2022] Open
Abstract
A large percentage of transcription factors require zinc to bind DNA. In this review, we discuss what makes p53 unique among zinc-dependent transcription factors. The conformation of p53 is unusually malleable: p53 binds zinc extremely tightly when folded, but is intrinsically unstable in the absence of zinc at 37°C. Whether the wild-type protein folds in the cell is largely determined by the concentration of available zinc. Consequently, zinc dysregulation in the cell as well as a large percentage of tumorigenic p53 mutations can cause p53 to lose zinc, misfold, and forfeit its tumor suppressing activity. We highlight p53’s noteworthy biophysical properties that give rise to its malleability and how proper zinc binding can be restored by synthetic metallochaperones to reactivate mutant p53. The activity and mechanism of metallochaperones are compared to those of other mutant p53-targeted drugs with an emphasis on those that have reached the clinical trial stage.
Collapse
Affiliation(s)
- Jeung-Hoi Ha
- Department of Biochemistry and Molecular Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Orjola Prela
- Division of Surgical Oncology, Department of Surgery, Wilmot Cancer Center, University of Rochester, Rochester, NY, United States
| | - Darren R Carpizo
- Division of Surgical Oncology, Department of Surgery, Wilmot Cancer Center, University of Rochester, Rochester, NY, United States
| | - Stewart N Loh
- Department of Biochemistry and Molecular Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| |
Collapse
|
36
|
Gou Y, Yang D, Tian T, Zhu X, Zhang R, Ren J, Tu D, Luo Y, Miao Y, Zhao H, Wang Y, Wei B. The Transcription of ZIP9 Is Associated With the Macrophage Polarization and the Pathogenesis of Hepatocellular Carcinoma. Front Immunol 2022; 13:725595. [PMID: 35371096 PMCID: PMC8967370 DOI: 10.3389/fimmu.2022.725595] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 02/22/2022] [Indexed: 12/14/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common digestive system cancers (DSCs) with a poor prognosis. Zinc‐regulated transporter (ZRT)/iron‐regulated transporter (IRT) like protein transporters (ZIPs) encode membrane transport proteins, which are responsible for the absorption of zinc and play important roles in the pathogenesis of various human cancers. Tumor-associated macrophages (TAMs) are important participants in the regulation of tumor microenvironment and the development of HCC. Individual role of each ZIP involved in hepatocarcinogenesis remains elusive. In this study, the transcription patterns of ZIPs in the DSCs were screened firstly through GEPIA2 database. Interestingly, the analysis of the DSCs data showed the distinct mRNA levels of ZIPs between DSCs tissues and healthy controls. Notably, the transcription levels of ZIP2, ZIP5, ZIP8, ZIP9 and ZIP14 were decreased significantly in the tissues of human liver cancer compared to paracarcinoma liver tissues. To further confirm the mRNA transcriptional changes of Zips in HCC, N-Nitrosodiethylamine (DEN) combined with carbon tetrachloride (CCl4) inducing mouse model of HCC were established. Consistently, the mRNA levels of Zip2, Zip9, and Zip14 in liver tissues of HCC induced mice were also decreased compared with the healthy controls. In addition, mouse peritoneal elucidated macrophages (PEMs)-derived M1/M2 macrophages in vitro, as well as human patients of HCC-derived TAMs, were used to examine the transcription levels of ZIPs. Our results showed that both Zip2 and Zip9 were up-regulated in M2-polarized macrophages. Zip2 transcript was also up-regulated M1-polarized macrophages, but Zip9 was slightly down-regulated. TAMs generated from human liver cancer tissues also displayed a decrease in ZIP9 transcription compared to paracarcinoma tissues. To further explore the role of Zip9 in M1/M2 polarization, the siRNA knockdown results revealed that Zip9, but not Zip2, could promote M2 macrophage polarization and impair M1 macrophage polarization. Mechanistically, Zip9 enhances phosphorylated STAT6 to promote M2 macrophage polarization but suppresses the phosphorylation of IκBα/β to inhibit M1 macrophage polarization. Together, our results indicate that ZIP9 may involve in macrophages polarity in HCC development and may be a potent new biomarker for the diagnosis of HCC.
Collapse
Affiliation(s)
- Yingying Gou
- School of Life Sciences, Shanghai University, Shanghai, China.,Shanghai Engineering Research Center of Organ Repair, Shanghai University, Shanghai, China
| | - Dan Yang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Science, Beijing, China
| | - Taikun Tian
- School of Life Sciences, Shanghai University, Shanghai, China.,Shanghai Engineering Research Center of Organ Repair, Shanghai University, Shanghai, China
| | - Xingguo Zhu
- School of Life Sciences, Shanghai University, Shanghai, China.,Shanghai Engineering Research Center of Organ Repair, Shanghai University, Shanghai, China
| | - Raorao Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Science, Beijing, China
| | - Jiaqi Ren
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Science, Beijing, China
| | - Dezhen Tu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Yi Luo
- Department of Head & Neck Surgery, Fudan University Shanghai Cancer Center/Cancer Institute, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuqing Miao
- Department of Respiration Medicine, Affiliated Nantong Hospital of Shanghai University, Nantong, China
| | - Huan Zhao
- Department of Respiration Medicine, Affiliated Nantong Hospital of Shanghai University, Nantong, China
| | - Yu Wang
- Department of Head & Neck Surgery, Fudan University Shanghai Cancer Center/Cancer Institute, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bin Wei
- School of Life Sciences, Shanghai University, Shanghai, China.,Shanghai Engineering Research Center of Organ Repair, Shanghai University, Shanghai, China.,Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
37
|
Asahina K. Induction of Cell Death in Pancreatic Tumors by Zinc and Its Fluorescence Chelator TSQ. Biol Trace Elem Res 2022; 200:1667-1676. [PMID: 34100198 DOI: 10.1007/s12011-021-02770-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 05/26/2021] [Indexed: 12/29/2022]
Abstract
Pancreatic ductal adenocarcinoma is a devastating cancer and is the fourth-leading cause of cancer death in the USA. Zinc is abundant in the pancreas, but its role in pancreatic cancer remains elusive. The aim of this study is to determine effects of zinc chelators in pancreatic cancer. Pdx1Cre and LSL-KrasG12D mice expressing an oncogenic mutation of KRAS develop pancreatic intraepithelial neoplasia in the pancreas. We found that EPCAM + tumors developed in the mouse pancreas store zinc that is detectable by fluorescence-activated cell sorting using N-(6-methoxy-8-quinolyl)-p-toluenesulfonamide (TSQ), a fluorescence chelator. EPCAM + TSQ + tumor cells isolated from the mouse pancreas formed organoids in matrigel. Upon treatment with N,N,N',N'-tetrakis(2-pyridinylmethyl)-1,2-ethanediamine (TPEN), a zinc chelator, the organoids degenerated and its negative effect was rescued by co-treatment with zinc, indicating that zinc is necessary for the growth and survival of tumor organoids. Different from TPEN, TSQ treatment did not affect the organoid growth and survival. Interestingly, co-treatment with TSQ and zinc resulted in strong emission of TSQ fluorescence in the organoid and its degeneration. The combination of zinc with TSQ, but not with TPEN, also induced cell death in PANC-1, a human pancreatic cancer cell line. These results suggest that a TSQ-zinc complex formed in pancreatic tumors induces cell death if zinc is overloaded.
Collapse
Affiliation(s)
- Kinji Asahina
- The Southern California Research Center for ALPD & Cirrhosis, Department of Pathology, Keck School of Medicine of the University of Southern California, 1333 San Pablo St., MMR 402, Los Angeles, CA, 90033, USA.
| |
Collapse
|
38
|
Ye C, Lian G, Wang T, Chen A, Chen W, Gong J, Luo L, Wang H, Xie L. The zinc transporter ZIP12 regulates monocrotaline-induced proliferation and migration of pulmonary arterial smooth muscle cells via the AKT/ERK signaling pathways. BMC Pulm Med 2022; 22:111. [PMID: 35346134 PMCID: PMC8962172 DOI: 10.1186/s12890-022-01905-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 03/17/2022] [Indexed: 01/05/2024] Open
Abstract
Background The zinc transporter ZIP12 is a membrane-spanning protein that transports zinc ions into the cytoplasm from the extracellular space. Recent studies demonstrated that upregulation of ZIP12 is involved in elevation of cytosolic free zinc and excessive proliferation of pulmonary arterial smooth muscle cells (PASMCs) induced by hypoxia. However, the expression of ZIP12 and its role in pulmonary arterial hypertension (PAH) induced by monocrotaline (MCT) in rats have not been evaluated previously. The aim of this study was to investigate the effect of ZIP12 on the proliferation and migration of PASMCs and its underlying mechanisms in MCT-induced PAH. Methods A PAH rat model was generated by intraperitoneal injection of 20 mg/kg MCT twice at one-week intervals. PASMCs were isolated from the pulmonary arteries of rats with MCT-induced PAH or control rats. The expression of ZIP12 and related molecules was detected in the lung tissues and cells. A ZIP12 knockdown lentivirus and an overexpressing lentivirus were constructed and transfected into PASMCs derived from PAH and control rats, respectively. EdU assays, wound healing assays and Western blotting were carried out to explore the function of ZIP12 in PASMCs. Results Increased ZIP12 expression was observed in PASMCs derived from MCT-induced PAH rats. The proliferation and migration of PASMCs from PAH rats were significantly increased compared with those from control rats. These results were corroborated by Western blot analysis of PCNA and cyclin D1. All these effects were significantly reversed by silencing ZIP12. Comparatively, ZIP12 overexpression resulted in the opposite effects as shown in PASMCs from control rats. Furthermore, selective inhibition of AKT phosphorylation by LY294002 abolished the effect of ZIP12 overexpression on enhancing cell proliferation and migration and partially suppressed the increase in ERK1/2 phosphorylation induced by ZIP12 overexpression. However, inhibition of ERK activity by U0126 resulted in partial reversal of this effect and did not influence an increase in AKT phosphorylation induced by ZIP12 overexpression. Conclusions ZIP12 is involved in MCT-induced pulmonary vascular remodeling and enhances the proliferation and migration of PASMCs. The mechanism of these effects was partially mediated by enhancing the AKT/ERK signaling pathways. Supplementary Information The online version contains supplementary material available at 10.1186/s12890-022-01905-3.
Collapse
Affiliation(s)
- Chaoyi Ye
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, Fujian, People's Republic of China.,Department of General Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China.,Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| | - Guili Lian
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| | - Tingjun Wang
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, Fujian, People's Republic of China.,Department of General Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China.,Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| | - Ai Chen
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| | - Weixiao Chen
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| | - Jin Gong
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, Fujian, People's Republic of China.,Department of General Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China.,Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| | - Li Luo
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, Fujian, People's Republic of China.,Department of General Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China.,Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| | - Huajun Wang
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| | - Liangdi Xie
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, Fujian, People's Republic of China. .,Department of General Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China. .,Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China.
| |
Collapse
|
39
|
|
40
|
Edamatsu H. Zinc ions negatively regulate proapoptotic signaling in cells expressing oncogenic mutant Ras. Biometals 2022; 35:349-362. [PMID: 35212861 DOI: 10.1007/s10534-022-00376-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 02/16/2022] [Indexed: 12/13/2022]
Abstract
Mutational activation of the Ras family of proto-oncogenes promotes cell survival and proliferation. Studies using cells cultured in vitro have shown that ectopic expression of constitutively active Ras suppresses apoptosis induced by serum deprivation. However, in some cellular contexts, constitutively active Ras exerts the opposite effects, including apoptosis of serum-starved embryonic fibroblasts. Such observations first came over two decades ago, but the molecular mechanisms by which mutant Ras increases the susceptibility of cells to serum deprivation leading to apoptosis are still not fully understood. To revisit this issue, I investigate the effects of serum depletion and mutant Ras expression on intracellular signaling and transcriptome of cells carrying an inducible allele of constitutively active mutant Hras (HrasG12V). I identify zinc ions (Zn2+) as a serum factor that suppresses proapoptotic signaling in cells expressing HrasG12V. Mechanistically, HrasG12V expression along with Zn2+ deficiency activates c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK), which are required for caspase-3 activation involved in the induction of cell death. Transcriptome analyses suggest that HrasG12V induces the unfolded protein response (UPR). Further analyses of intracellular signaling biomolecules related to the UPR indicate that HrasG12V activates inositol-requiring protein 1 (IRE1), which synergizes with Zn2+ deficiency to activate JNK and p38 MAPK signaling. These results provide insights into a role of Zn2+ that counteracts proapoptotic signaling activated by mutationally activated Ras.
Collapse
Affiliation(s)
- Hironori Edamatsu
- Department of Biology, Juntendo University School of Medicine, 1-1 Hirakagakuendai, Inzai, Chiba, 270-1695, Japan.
| |
Collapse
|
41
|
Wang M, Li Y, Wang M, Liu K, Hoover AR, Li M, Towner RA, Mukherjee P, Zhou F, Qu J, Chen WR. Synergistic interventional photothermal therapy and immunotherapy using an iron oxide nanoplatform for the treatment of pancreatic cancer. Acta Biomater 2022; 138:453-462. [PMID: 34757232 PMCID: PMC10960566 DOI: 10.1016/j.actbio.2021.10.048] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/13/2021] [Accepted: 10/26/2021] [Indexed: 12/12/2022]
Abstract
Pancreatic cancer (PC) is the most lethal malignancy due to its high metastatic ability and poor drug permeability. Here, a synergized interventional photothermal-immunotherapy strategy was developed with imaging guidance and temperature monitoring by magnetic resonance imaging (MRI) technique, for the local treatment of metastatic PC. A tumor microenvironment (TME)-responsive nanoplatform was fabricated via coating of DSPE-PEG and indocyanine green (ICG) onto imiquimod (IMQ) loaded amorphous iron oxide nanoparticles (IONs). This unique nanoplatform, IMQ@IONs/ICG, served as a contrast agent for MRI, a drug delivery vehicle for IMQ and ICG, and a catalyst for TME modulation. The biodegradable IMQ@IONs/ICG was also non-toxic, and improved the penetration of the loaded drugs in PC to maximize thermal ablation of the tumor and minimize damage to the surrounding healthy tissue. For the treatment of aggressive, metastatic Panc02-H7 pancreatic tumors in mice, ION-assisted MRI was employed to guide the administration of interventional photothermal therapy (IPTT) and monitor the temperature distribution in target tumor and surrounding tissue during treatment. The local IPTT treatment induced in situ immunogenic cell death (ICD), and, in combination with released IMQ, triggered a strong antitumor immunity, leading to decreased metastases and increased CD8+ in spleen and tumors. With precise local treatment and monitoring, treated primary tumors were completely eradicated, mesentery metastases were dramatically reduced, and the survival time was significantly prolonged, without damage to normal tissue and systemic autoimmunity. Overall, this synergistic strategy represents a promising approach to treat PC with significant potential for clinical applications. STATEMENT OF SIGNIFICANCE: Pancreatic cancer (PC) is one of the most lethal malignancies because it is non-permeable to drugs and highly metastatic. In this study, we designed a tumor microenvironment-responsive amorphous iron oxide nanoplatform (ION) to co-deliver photothermal agent (ICG) and toll-like-receptor-7 agonist (IMQ). This biodegradable nanoplatform IMQ@IONs/ICG improved the penetration of the loaded drugs in pancreatic tumor. With MR imaging guidance and temperature monitoring, the precise interventional photothermal therapy on mouse Panc02-H7 orthotopic tumors releases tumor antigens to initiate tumor-special immune responses, amplified by the released IMQ. Our results demonstrate that IMQ@IONs/ICG overcomes the obstacle of drug delivery to pancreatic tumors, and when combined with photothermal therapy, induces a systemic antitumor immunity to control metastatic tumors.
Collapse
Affiliation(s)
- Meng Wang
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Yong Li
- Interventional Therapy Department, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Miao Wang
- School of Biomedical Engineering, Hainan University, Haikou 570228, China
| | - Kaili Liu
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA
| | - Ashley R Hoover
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA
| | - Min Li
- Department of Medicine, Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Rheal A Towner
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Priyabrata Mukherjee
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Feifan Zhou
- School of Biomedical Engineering, Hainan University, Haikou 570228, China.
| | - Junle Qu
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China.
| | - Wei R Chen
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA.
| |
Collapse
|
42
|
Qu YY, Guo RY, Luo ML, Zhou Q. Pan-Cancer Analysis of the Solute Carrier Family 39 Genes in Relation to Oncogenic, Immune Infiltrating, and Therapeutic Targets. Front Genet 2021; 12:757582. [PMID: 34925450 PMCID: PMC8675640 DOI: 10.3389/fgene.2021.757582] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 11/12/2021] [Indexed: 12/28/2022] Open
Abstract
Background: Emerging pieces of evidence demonstrated that the solute carrier family 39 (SLC39A) members are critical for the oncogenic and immune infiltrating targets in multiple types of tumors. However, the precise relationship between the SLC39A family genes and clinical prognosis as well as the pan-cancer tumor cell infiltration has not been fully elucidated. Methods: In this study, the pan-cancer expression profile, genetic mutation, prognostic effect, functional enrichment, immune infiltrating, and potential therapeutic targets of the SLC39A family members were investigated by analyzing multiple public databases such as the Oncomine, TIMER, GEPIA, cBioPortal, KM-plotter, PrognoScan, GeneMANIA, STRING, DAVID, TIMER 2.0, and CellMiner databases. Results: The expression levels of most SLC39 family genes in the tumor tissues were found to be significantly upregulated compared to the normal group. In mutation analysis, the mutation frequencies of SLC39A4 and SLC39A1 were found to be higher among all the members (6 and 4%, respectively). Moreover, the overall mutation frequency of the SLC39A family genes ranged from 0.8 to 6% pan-cancer. Also, the function of the SLC39A highly related genes was found to be enriched in functions such as zinc II ion transport across the membrane, steroid hormone biosynthesis, and chemical carcinogenesis. In immune infiltration analysis, the expression level of the SLC39A family genes was found to be notably related to the immune infiltration levels of six types of immune cells in specific types of tumors. In addition, the SLC39A family genes were significantly related to the sensitivity or resistance of 63 antitumor drugs in a variety of tumor cell lines. Conclusion: These results indicate that the SLC39 family genes are significant for determining cancer progression, immune infiltration, and drug sensitivity in multiple cancers. This study, therefore, provides novel insights into the pan-cancer potential targets of the SLC39 family genes.
Collapse
Affiliation(s)
- Yi-Yuan Qu
- Department of Gynecology and Obstetrics, the People's Hospital of China Three Gorges University/The First People's Hospital of Yichang, Yichang, China
| | - Rong-Yan Guo
- Emergency Services Department, HanYang Hospital Affiliated of Wuhan University of Science and Technology, Wuhan, China
| | - Meng-Ling Luo
- Department of Gynecology and Obstetrics, the People's Hospital of China Three Gorges University/The First People's Hospital of Yichang, Yichang, China
| | - Quan Zhou
- Department of Gynecology and Obstetrics, the People's Hospital of China Three Gorges University/The First People's Hospital of Yichang, Yichang, China
| |
Collapse
|
43
|
Csikós A, Kozma B, Pór Á, Kovács I, Lampé R, Miklós I, Takacs P. Zinc Transporter 9 (SLC30A9) Expression Is Decreased in the Vaginal Tissues of Menopausal Women. Biol Trace Elem Res 2021; 199:4011-4019. [PMID: 33409913 DOI: 10.1007/s12011-020-02525-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/29/2020] [Indexed: 12/11/2022]
Abstract
Our aim was to compare zinc transporter (ZnT/SLC30A, and ZIP/SLC39A) expression between pre- and postmenopausal women in human vaginal tissues. Zinc transporter families are responsible for the maintenance of intracellular zinc concentrations. Zinc has significant effects on the extracellular matrix composition. Vaginal wall biopsies were obtained from seven premenopausal and seven postmenopausal women. mRNA expression of twenty-four zinc transporters was determined by quantitative real-time PCR. Zinc transporter expression at the protein level was assessed by immunohistochemistry. Student's t test and Mann-Whitney U test were used to compare data. ZnT4 and ZnT9 mRNA expression were significantly lower in postmenopausal women compared with premenopausal women (mean ± SD mRNA expression in relative units, 96.43 ± 140.61 vs. 410.59 ± 304.34, p = 0.03 and 0.62 ± 0.39 vs. 1.13 ± 0.31, p = 0.02). In addition, ZIP2, ZIP3, and ZIP6 mRNA expressions were significantly lower in postmenopausal women compared with premenopausal women (mean ± SD mRNA expression in relative units, 1.11 ± 0.61 vs. 2.29 ± 1.20, p = 0.04; 2.32 ± 1.90 vs. 15.82 ± 12.97, p = 0.02 and 1.10 ± 0.80 vs. 5.73 ± 4.72, p = 0.03). ZnT9 protein expression in the stratum spinosum was significantly lower in postmenopausal women (p = 0.012). Zinc transporters were expressed differentially in the vaginal tissues. ZnT9 expression was significantly lower in postmenopausal women compared with premenopausal women.
Collapse
Affiliation(s)
- Anett Csikós
- Molecular Biology Group, FemPharma, LLC, Vígkedvű Mihály utca 21. II/5, Debrecen, 4024, Hungary.
| | - Bence Kozma
- Department of Obstetrics and Gynecology, University of Debrecen, Debrecen, Hungary
| | - Ágnes Pór
- Department of Pathology, Gyula Kenézy County Hospital, University of Debrecen, Debrecen, Hungary
| | - Ilona Kovács
- Department of Pathology, Gyula Kenézy County Hospital, University of Debrecen, Debrecen, Hungary
| | - Rudolf Lampé
- Department of Obstetrics and Gynecology, University of Debrecen, Debrecen, Hungary
| | - Ida Miklós
- Department of Genetics and Applied Microbiology, University of Debrecen, Debrecen, Hungary
| | - Peter Takacs
- Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, VA, USA
| |
Collapse
|
44
|
Abstract
Claudins are adhesion molecules located at the tight junctions between epithelial cells. A series of studies have now reported aberrant expression of claudin proteins in the context of neoplastic transformation, suggesting its role in tumorigenesis. However, the precise mechanisms are still not well understood. Studies on expression alterations of claudins have revealed a range of outcomes that reflect the complexity of claudins in terms of spatial localization, tumor type and stage of disease. The diverse and dynamic expression patterns of claudins in cancer are tightly controlled by a wide range of regulatory mechanisms, which are commonly modulated by oncogenic signaling pathways. The present review summarizes the recent knowledge describing the dysregulation of claudin expression in cancer and discusses the intrinsic and extrinsic determinants of the context-specific expression patterns of claudins.
Collapse
Affiliation(s)
- Jian Li
- Department of General Surgery, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, Sichuan 621000, P.R. China
| |
Collapse
|
45
|
Vogel-González M, Musa-Afaneh D, Rivera Gil P, Vicente R. Zinc Favors Triple-Negative Breast Cancer's Microenvironment Modulation and Cell Plasticity. Int J Mol Sci 2021; 22:ijms22179188. [PMID: 34502091 PMCID: PMC8431059 DOI: 10.3390/ijms22179188] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/19/2021] [Accepted: 08/21/2021] [Indexed: 01/19/2023] Open
Abstract
Triple-negative breast cancer (TNBC) tends to metastasize to the brain, a step that worsens the patient’s prognosis. The specific hallmarks that determine successful metastasis are motility and invasion, microenvironment modulation, plasticity, and colonization. Zinc, an essential trace element, has been shown to be involved in all of these processes. In this work, we focus our attention on the potential role of zinc during TNBC metastasis. We used MDA-MB-BrM2 (BrM2) cells, a brain metastasis model derived from the parental TNBC cell line MDA-MB-231. Our studies show that BrM2 cells had double the zinc content of MDA-MB-231 cells. Moreover, exploring different metastatic hallmarks, we found that the zinc concentration is especially important in the microenvironment modulation of brain metastatic cells, enhancing the expression of SerpinB2. Furthermore, we show that zinc promotes the tumorigenic capacity of breast cancer stem cells. In addition, by causing a disturbance in MDA-MB-231 zinc homeostasis by overexpressing the Zip4 transporter, we were able to increase tumorigenicity. Nevertheless, this strategy did not completely recapitulate the BrM2 metastatic phenotype. Altogether, our work suggests that zinc plays an important role in the transformative steps that tumoral cells take to acquire tumorigenic potential and niche specificity.
Collapse
Affiliation(s)
- Marina Vogel-González
- Laboratory of Molecular Physiology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (M.V.-G.); (D.M.-A.)
| | - Dunia Musa-Afaneh
- Laboratory of Molecular Physiology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (M.V.-G.); (D.M.-A.)
| | - Pilar Rivera Gil
- Integrative Biomedical Materials and Nanomedicine Lab, Department of Experimental and Health Sciences, Pompeu Fabra University, 08003 Barcelona, Spain;
| | - Rubén Vicente
- Laboratory of Molecular Physiology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (M.V.-G.); (D.M.-A.)
- Correspondence: ; Tel.: +34-933-160-854
| |
Collapse
|
46
|
Jiang Y, Zhan H, Zhang Y, Yang J, Liu M, Xu C, Fan X, Zhang J, Zhou Z, Shi X, Ramesh R, Li M. ZIP4 promotes non-small cell lung cancer metastasis by activating snail-N-cadherin signaling axis. Cancer Lett 2021; 521:71-81. [PMID: 34450198 DOI: 10.1016/j.canlet.2021.08.025] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/16/2021] [Accepted: 08/22/2021] [Indexed: 02/07/2023]
Abstract
Non-small cell lung cancer (NSCLC) is one of the most critical health problems worldwide, with high incidence and poor survival rate. A zinc importer ZIP4 has been implicated in the process of tumor growth and metastasis of many cancers. However, its exact role and the underlying mechanism in NSCLC remains to be elucidated. In the present study, we found that human ZIP4 was substantially overexpressed in NSCLC tissues and was correlated with poor overall survival (OS) and progression-free survival (PFS). Overexpression of ZIP4 promoted cell migration, invasion and metastasis both in vitro and in a mouse lung metastasis model. Silencing of ZIP4 attenuated migration, invasion and metastasis. Mechanistically, overexpression of ZIP4 increased the expression of Snail, Slug and N-cadherin while genetic inactivation of ZIP4 downregulated the expression of above-mentioned genes. Further analysis showed that transcriptional factor Snail which modulates N-cadherin was involved in the process of ZIP4-mediated NSCLC migration and invasion. We also demonstrated that ZIP4 positively correlates with the levels of Snail, Slug and N-cadherin in mice lung metastasis tumors. Together, these results suggest that ZIP4 acts as an important regulator of Snail-N-cadherin signaling axis in promoting NSCLC progression and may serve as a novel predictive marker and therapeutic target in NSCLC.
Collapse
Affiliation(s)
- Yuanyuan Jiang
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital, Shandong University, Jinan, Shandong, China; Department of Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Surgery, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Hanxiang Zhan
- Department of Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Surgery, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Yuqing Zhang
- Department of Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Surgery, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jingxuan Yang
- Department of Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Surgery, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Mingyang Liu
- Department of Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Surgery, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Chao Xu
- Department of Biostatistics and Epidemiology, Hudson College of Public Health, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Xiao Fan
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junxia Zhang
- Department of Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Surgery, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhijun Zhou
- Department of Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Surgery, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Xiuhui Shi
- Department of Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Surgery, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Rajagopal Ramesh
- Department of Pathology, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Min Li
- Department of Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Surgery, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
47
|
Liu Z, Hu Z, Cai X, Liu S. SLC39A5 promotes lung adenocarcinoma cell proliferation by activating PI3K/AKT signaling. Pathol Res Pract 2021; 224:153541. [PMID: 34252710 DOI: 10.1016/j.prp.2021.153541] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 07/01/2021] [Accepted: 07/01/2021] [Indexed: 12/13/2022]
Abstract
Lung cancer is the most common cancer and the primary cause of cancer-related deaths worldwide. Solute carrier family 39 member 5 (SLC39A5) regulates cellular zinc homeostasis and plays a vital role in several human cancers. However, the clinical significance and biological function of SLC39A5 in lung adenocarcinoma (LUAD) remain unclear. Hence, we sought to elucidate the role of SLC39A5 in LUAD pathophysiology in this study. The expression and clinical significance of SLC39A5 were evaluated using The Cancer Genome Atlas, the Gene Expression Omnibus, and tissue microarray data. We used the Cell Counting Kit-8, flow cytometry, western blotting, and quantitative reverse transcriptase-polymerase chain reaction analyses to determine the function of SLC39A5 in vitro. We also used a mouse xenograft model to evaluate the function of SLC39A5 in vivo. Our results indicate that SLC39A5 was upregulated in LUAD tissues compared with that in adjacent non-tumor lung tissues. SLC39A5 overexpression correlated with poor survival in patients with LUAD. SLC39A5 promoted LUAD cell proliferation by accelerating the G1-to-S phase transition and inhibiting apoptosis. SLC39A5 knockdown inhibited the tumorigenesis of LUAD cells in a nude mouse model of xenograft tumors. SLC39A5 promoted LUAD cell proliferation by activating the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling. SLC39A5 played an oncogenic role in LUAD by activating the PI3K/AKT signaling. Hence, SLC39A5 may serve as a novel prognostic biomarker and potential therapeutic target for LUAD.
Collapse
Affiliation(s)
- Zhaohui Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Department of Respiratory Medicine, Chenzhou No.1 People's Hospital, Chenzhou 423000, China
| | - Zheng Hu
- Translational Medicine Institute, the First People's Hospital of Chenzhou Affiliated to University of South China, Chenzhou 432000, China
| | - Xingdong Cai
- Department of Respiratory Medicine, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Shengming Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China.
| |
Collapse
|
48
|
Fan Q, Li L, Wang TL, Emerson RE, Xu Y. A Novel ZIP4-HDAC4-VEGFA Axis in High-Grade Serous Ovarian Cancer. Cancers (Basel) 2021; 13:cancers13153821. [PMID: 34359722 PMCID: PMC8345154 DOI: 10.3390/cancers13153821] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 07/25/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Despite tremendous research efforts, epithelial ovarian cancer (EOC) remains one of the most difficult cancers to detect early and treat successfully for >5-year survival. We have recently shown that ZIP4, a zinc transporter, is a novel cancer stem cell (CSC) marker and a therapeutic target for EOC. The current work focuses on developing new strategies to target ZIP4 and inhibit its CSC activities in EOC. We found that cells expressing high levels of ZIP4 were supersensitive to a group of inhibitors called HDACis. One of the major targets of these inhibitors is a protein called HDAC4. We revealed the new molecular bases for the ZIP4-HDAC4 axis and tested the efficacies of targeting this axis in the lab and in mouse models. Our study provides a new mechanistic-based targeting strategy for EOC. Abstract We have recently identified ZIP4 as a novel cancer stem cell (CSC) marker in high-grade serous ovarian cancer (HGSOC). While it converts drug-resistance to cisplatin (CDDP), we unexpectedly found that ZIP4 induced sensitization of HGSOC cells to histone deacetylase inhibitors (HDACis). Mechanistically, ZIP4 selectively upregulated HDAC IIa HDACs, with little or no effect on HDACs in other classes. HDAC4 knockdown (KD) and LMK-235 inhibited spheroid formation in vitro and tumorigenesis in vivo, with hypoxia inducible factor-1 alpha (HIF1α) and endothelial growth factor A (VEGFA) as functional downstream mediators of HDAC4. Moreover, we found that ZIP4, HDAC4, and HIF1α were involved in regulating secreted VEGFA in HGSOC cells. Furthermore, we tested our hypothesis that co-targeting CSC via the ZIP4-HDAC4 axis and non-CSC using CDDP is necessary and highly effective by comparing the effects of ZIP4-knockout/KD, HDAC4-KD, and HDACis, in the presence or absence of CDDP on tumorigenesis in mouse models. Our results showed that the co-targeting strategy was highly effective. Finally, data from human HGSOC tissues showed that ZIP4 and HDAC4 were upregulated in a subset of recurrent tumors, justifying the clinical relevance of the study. In summary, our study provides a new mechanistic-based targeting strategy for HGSOC.
Collapse
Affiliation(s)
- Qipeng Fan
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, 950 W. Walnut St. R2-E380, Indianapolis, IN 46202, USA;
| | - Lihong Li
- Department of Gynecology and Obstetrics, Johns Hopkins Medical Institutions, 600 North Wolfe St., Baltimore, MD 21287, USA;
| | - Tian-Li Wang
- Department of Gynecology, Oncology, and Pathology, Johns Hopkins Medical Institutions, 1550 Orleans Street, Baltimore, MD 21231, USA;
| | - Robert E. Emerson
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indiana University Health Pathology Laboratory, 350 W. 11th Street, Room 4010, Indianapolis, IN 46202, USA;
| | - Yan Xu
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, 950 W. Walnut St. R2-E380, Indianapolis, IN 46202, USA;
- Correspondence: ; Tel.: +1-(317)-274-3972
| |
Collapse
|
49
|
Li J. Context-Dependent Roles of Claudins in Tumorigenesis. Front Oncol 2021; 11:676781. [PMID: 34354941 PMCID: PMC8329526 DOI: 10.3389/fonc.2021.676781] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 07/05/2021] [Indexed: 12/16/2022] Open
Abstract
The barrier and fence functions of the claudin protein family are fundamental to tissue integrity and human health. Increasing evidence has linked claudins to signal transduction and tumorigenesis. The expression of claudins is frequently dysregulated in the context of neoplastic transformation. Studies have uncovered that claudins engage in nearly all aspects of tumor biology and steps of tumor development, suggesting their promise as targets for treatment or biomarkers for diagnosis and prognosis. However, claudins can be either tumor promoters or tumor suppressors depending on the context, which emphasizes the importance of taking various factors, including organ type, environmental context and genetic confounders, into account when studying the biological functions and targeting of claudins in cancer. This review discusses the complicated roles and intrinsic and extrinsic determinants of the context-specific effects of claudins in cancer.
Collapse
Affiliation(s)
- Jian Li
- Department of General Surgery, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, China
| |
Collapse
|
50
|
He X, Ma Y, Xie H, Rao G, Yang Z, Zhang J, Feng Z. Biomimetic Nanostructure Platform for Cancer Diagnosis Based on Tumor Biomarkers. Front Bioeng Biotechnol 2021; 9:687664. [PMID: 34336803 PMCID: PMC8320534 DOI: 10.3389/fbioe.2021.687664] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/07/2021] [Indexed: 12/11/2022] Open
Abstract
Biomarker discovery and its clinical use have attracted considerable attention since early cancer diagnosis can significantly decrease mortality. Cancer biomarkers include a wide range of biomolecules, such as nucleic acids, proteins, metabolites, sugars, and cytogenetic substances present in human biofluids. Except for free-circulating biomarkers, tumor-extracellular vesicles (tEVs) and circulating tumor cells (CTCs) can serve as biomarkers for the diagnosis and prognosis of various cancers. Considering the potential of tumor biomarkers in clinical settings, several bioinspired detection systems based on nanotechnologies are in the spotlight for detection. However, tremendous challenges remain in detection because of massive contamination, unstable signal-to-noise ratios due to heterogeneity, nonspecific bindings, or a lack of efficient amplification. To date, many approaches are under development to improve the sensitivity and specificity of tumor biomarker isolation and detection. Particularly, the exploration of natural materials in biological frames has encouraged researchers to develop new bioinspired and biomimetic nanostructures, which can mimic the natural processes to facilitate biomarker capture and detection in clinical settings. These platforms have substantial influence in biomedical applications, owing to their capture ability, significant contrast increase, high sensitivity, and specificity. In this review, we first describe the potential of tumor biomarkers in a liquid biopsy and then provide an overview of the progress of biomimetic nanostructure platforms to isolate and detect tumor biomarkers, including in vitro and in vivo studies. Capture efficiency, scale, amplification, sensitivity, and specificity are the criteria that will be further discussed for evaluating the capability of platforms. Bioinspired and biomimetic systems appear to have a bright future to settle obstacles encountered in tumor biomarker detection, thus enhancing effective cancer diagnosis.
Collapse
Affiliation(s)
- Xiping He
- Department of Rehabilitation Medicine, The Affiliated Wenling Hospital of Wenzhou Medical University, Wenling, China
| | - Yifan Ma
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, United States
| | - Haotian Xie
- Department of Mathematics, The Ohio State University, Columbus, OH, United States
| | - Gaofeng Rao
- Department of Rehabilitation Medicine, The Affiliated Wenling Hospital of Wenzhou Medical University, Wenling, China
| | - Zhaogang Yang
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Jingjing Zhang
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, United States
| | - Zhong Feng
- Department of Neurology, The Affiliated Wenling Hospital of Wenzhou Medical University, Wenling, China
| |
Collapse
|