1
|
Cillo M, Buonomo V, Vainshtein A, Grumati P. Autophagy, ER-phagy and ER Dynamics During Cell Differentiation. J Mol Biol 2025:169151. [PMID: 40222412 DOI: 10.1016/j.jmb.2025.169151] [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: 11/30/2024] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 04/15/2025]
Abstract
The endoplasmic reticulum (ER) is a multifunctional organelle essential for protein and lipid synthesis, ion transport and inter-organelle communication. It comprises a highly dynamic network of membranes that continuously reshape to support a wide range of cellular processes. During cellular differentiation, extensive remodelling of both ER architecture and its proteome is required to accommodate alterations in cell morphology and function. Autophagy, and ER-phagy in particular, plays a pivotal role in reshaping the ER, enabling cells to meet their evolving needs and adapt to developmental cues. Despite the ER's critical role in cellular differentiation, the mechanisms responsible for regulating its dynamics are not fully understood. Emerging evidence suggests that transcriptional and post-translational regulation play a role in fine-tuning ER-phagy and the unfolded protein response (UPR). This review explores the molecular basis of autophagy and ER-phagy, highlighting their role in ER remodelling during cellular differentiation. A deeper understanding of these processes could open new avenues for targeted therapeutic approaches in conditions where ER remodelling is impaired.
Collapse
Affiliation(s)
- Michele Cillo
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy; Department of Clinical Medicine and Surgery, Federico II University, 80131 Naples, Italy
| | - Viviana Buonomo
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy; Department of Clinical Medicine and Surgery, Federico II University, 80131 Naples, Italy
| | | | - Paolo Grumati
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy; Department of Clinical Medicine and Surgery, Federico II University, 80131 Naples, Italy.
| |
Collapse
|
2
|
Ju S, Singh MK, Han S, Ranbhise J, Ha J, Choe W, Yoon KS, Yeo SG, Kim SS, Kang I. Oxidative Stress and Cancer Therapy: Controlling Cancer Cells Using Reactive Oxygen Species. Int J Mol Sci 2024; 25:12387. [PMID: 39596452 PMCID: PMC11595237 DOI: 10.3390/ijms252212387] [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: 09/26/2024] [Revised: 10/31/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
Cancer is a multifaceted disease influenced by various mechanisms, including the generation of reactive oxygen species (ROS), which have a paradoxical role in both promoting cancer progression and serving as targets for therapeutic interventions. At low concentrations, ROS serve as signaling agents that enhance cancer cell proliferation, migration, and resistance to drugs. However, at elevated levels, ROS induce oxidative stress, causing damage to biomolecules and leading to cell death. Cancer cells have developed mechanisms to manage ROS levels, including activating pathways such as NRF2, NF-κB, and PI3K/Akt. This review explores the relationship between ROS and cancer, focusing on cell death mechanisms like apoptosis, ferroptosis, and autophagy, highlighting the potential therapeutic strategies that exploit ROS to target cancer cells.
Collapse
Affiliation(s)
- Songhyun Ju
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Manish Kumar Singh
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sunhee Han
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jyotsna Ranbhise
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Joohun Ha
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Wonchae Choe
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Kyung-Sik Yoon
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seung Geun Yeo
- Department of Otorhinolaryngology—Head and Neck Surgery, College of Medicine, Kyung Hee University Medical Center, Kyung Hee University, Seoul 02453, Republic of Korea;
| | - Sung Soo Kim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Insug Kang
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
3
|
Knupp J, Pletan ML, Arvan P, Tsai B. Autophagy of the ER: the secretome finds the lysosome. FEBS J 2023; 290:5656-5673. [PMID: 37920925 PMCID: PMC11044768 DOI: 10.1111/febs.16986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/20/2023] [Accepted: 10/23/2023] [Indexed: 11/04/2023]
Abstract
Lysosomal degradation of the endoplasmic reticulum (ER) and its components through the autophagy pathway has emerged as a major regulator of ER proteostasis. Commonly referred to as ER-phagy and ER-to-lysosome-associated degradation (ERLAD), how the ER is targeted to the lysosome has been recently clarified by a growing number of studies. Here, we summarize the discoveries of the molecular components required for lysosomal degradation of the ER and their proposed mechanisms of action. Additionally, we discuss how cells employ these machineries to create the different routes of ER-lysosome-associated degradation. Further, we review the role of ER-phagy in viral infection pathways, as well as the implication of ER-phagy in human disease. In sum, we provide a comprehensive overview of the current field of ER-phagy.
Collapse
Affiliation(s)
- Jeffrey Knupp
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
- Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Madison L Pletan
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
- Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Peter Arvan
- Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Billy Tsai
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
- Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
4
|
Kanamori A, Hinaga S, Hirata Y, Amaya F, Oh-Hashi K. Molecular characterization of wild-type and HSAN2B-linked FAM134B. Mol Biol Rep 2023:10.1007/s11033-023-08517-y. [PMID: 37273064 DOI: 10.1007/s11033-023-08517-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 05/11/2023] [Indexed: 06/06/2023]
Abstract
BACKGROUND Family with sequence similarity 134, member B (FAM134B), also known as Reticulophagy regulator 1 (RETREG1), is an ER-phagy receptor involved in ER homeostasis. Congenital mutations in the FAM134B gene have been reported to be associated with hereditary sensory and autonomic neuropathy type 2B (HSAN2B); however, the molecular differences between wild-type and HSAN2B-linked FAM134B are not fully understood. METHODS AND RESULTS We prepared several human FAM134B constructs, such as the HSAN2B-linked mutant, and compared their features with those of wild-type FAM134B by transfecting these constructs into FAM134B-deficient Neuro2a cells. Although intrinsic FAM134B protein expression in wild-type Neuro2a cells was affected by the supply of amino acids in the culture medium, the expression of each HSAN2B-linked mutant FAM134B protein was hardly affected by serum and amino acid deprivation. On the other hand, the intracellular localization of GFP-tagged HSAN2B-linked mutants, except for P7Gfs133X, overlapped well with ER-localized SP-RFPKDEL and did not differ from that of GFP-tagged wild-type FAM134B. However, analysis of protein‒protein interactions using the NanoBiT reporter assay revealed the difference between wild-type and C-terminal truncated mutant FAM134B. Furthermore, this NanoBiT assay demonstrated that both wild-type and G216R FAM134B interacted with LC3/GABARAPL1 to the same extent, but the FAM134B construct with mutations near the LC3-interacting region (LIR) did not. Similar to the NanoBiT assay, the C-terminal-truncated FAM134B showed lower ER-phagy activities, as assessed by the cotransfection of GFP-tagged reporters. CONCLUSIONS We showed that wild-type and HSAN2B-linked FAM134B have different molecular characteristics by transfecting cells with various types of constructs. Thus, this study provides new insights into the molecular mechanisms underlying HSAN2B as well as the regulation of ER-phagy.
Collapse
Affiliation(s)
- Akane Kanamori
- Graduate School of Natural Science and Technology, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Shohei Hinaga
- Graduate School of Natural Science and Technology, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Yoko Hirata
- Graduate School of Natural Science and Technology, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Fumimasa Amaya
- Department of Pain Management and Palliative Care Medicine, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamikyo-Ku, Kyoto, 602-0841, Japan
| | - Kentaro Oh-Hashi
- Graduate School of Natural Science and Technology, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan.
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan.
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan.
- Center for One Medicine Innovative Translational Research (COMIT), Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan.
| |
Collapse
|
5
|
Chen B, Hu X, Chen M, Chen Y, Yan L, Zeng G, Wang C, Liu L, Yang C, Song W. Identification of sensory dysfunction and nervous structure changes in Fam134b knockout mice. Neurol Res 2023; 45:41-48. [PMID: 36302074 DOI: 10.1080/01616412.2022.2117947] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
OBJECTIVES Mutation in human FAM134B gene has been implicated in hereditary sensory and autonomic neuropathy type IIB. We aimed to knock out Fam134b in mice and explored its phenotypes to determine whether the genetic impairments and behavioral changes can mirror manifestations noted in humans. METHODS We used CRISPR/Cas9 technology to knockout the Fam134b gene in the C57BL/6 J mouse. After confirming the knockout was successful by Sanger sequencing and Western blot, sensory function was measured using the hot plate test and the 50% paw withdrawal threshold test. In addition, standard microscopy and transmission electron microscopy were performed to observe the structural changes of the dorsal root ganglion sensory neuron and the sciatic nerve. RESULTS DNA sequencing and Western blot analysis confirmed the mutation in the Fam134b mutation gene and the loss of expression of its products. Fam134b knockout mice exhibited heat pain insensitivity and mechanical hyperalgesia. Interestingly, limb damage was found in some homozygotes. Demyelination in the sciatic nerve was common. Golgi bodies were turgid in dorsal root ganglion neuron. CONCLUSIONS These findings indicate that peripheral neuropathy is common in Fam134b KO mice. We believe this novel animal model is likely to have significant future potential as a reliable model for the evaluation of peripheral neuropathy and its complications.
Collapse
Affiliation(s)
- Binghao Chen
- The Department of Orthopaedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xingyun Hu
- The department of endocrinology and Metabolism, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Meiling Chen
- The department of endocrinology and Metabolism, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuying Chen
- The department of endocrinology and Metabolism, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Li Yan
- The department of endocrinology and Metabolism, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Gang Zeng
- The Department of Orthopaedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Chuan Wang
- The department of endocrinology and Metabolism, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Lixuan Liu
- The department of endocrinology and Metabolism, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Chuan Yang
- The department of endocrinology and Metabolism, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Weidong Song
- The Department of Orthopaedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
6
|
Canonical and Noncanonical ER Stress-Mediated Autophagy Is a Bite the Bullet in View of Cancer Therapy. Cells 2022; 11:cells11233773. [PMID: 36497032 PMCID: PMC9738281 DOI: 10.3390/cells11233773] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/20/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
Cancer cells adapt multiple mechanisms to counter intense stress on their way to growth. Tumor microenvironment stress leads to canonical and noncanonical endoplasmic stress (ER) responses, which mediate autophagy and are engaged during proteotoxic challenges to clear unfolded or misfolded proteins and damaged organelles to mitigate stress. In these conditions, autophagy functions as a cytoprotective mechanism in which malignant tumor cells reuse degraded materials to generate energy under adverse growing conditions. However, cellular protection by autophagy is thought to be complicated, contentious, and context-dependent; the stress response to autophagy is suggested to support tumorigenesis and drug resistance, which must be adequately addressed. This review describes significant findings that suggest accelerated autophagy in cancer, a novel obstacle for anticancer therapy, and discusses the UPR components that have been suggested to be untreatable. Thus, addressing the UPR or noncanonical ER stress components is the most effective approach to suppressing cytoprotective autophagy for better and more effective cancer treatment.
Collapse
|
7
|
Wang S, Yang Y, Li L, Ma P, Jiang Y, Ge M, Yu Y, Huang H, Fang Y, Jiang N, Miao H, Guo H, Yan L, Ren Y, Sun L, Zha Y, Li N. Identification of Tumor Antigens and Immune Subtypes of Malignant Mesothelioma for mRNA Vaccine Development. Vaccines (Basel) 2022; 10:1168. [PMID: 35893817 PMCID: PMC9331978 DOI: 10.3390/vaccines10081168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/07/2022] [Accepted: 07/13/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND mRNA-based cancer vaccines have been considered a promising anticancer therapeutic approach against various cancers, yet their efficacy for malignant mesothelioma (MESO) is still not clear. The present study is designed to identify MESO antigens that have the potential for mRNA vaccine development, and to determine the immune subtypes for the selection of suitable patients. METHODS A total of 87 MESO datasets were used for the retrieval of RNA sequencing and clinical data from The Cancer Genome Atlas (TCGA) databases. The possible antigens were identified by a survival and a genome analysis. The samples were divided into two immune subtypes by the application of a consensus clustering algorithm. The functional annotation was also carried out by using the DAVID program. Furthermore, the characterization of each immune subtype related to the immune microenvironment was integrated by an immunogenomic analysis. A protein-protein interaction network was established to categorize the hub genes. RESULTS The five tumor antigens were identified in MESO. FAM134B, ALDH3A2, SAV1, and RORC were correlated with superior prognoses and the infiltration of antigen-presenting cells (APCs), while FN1 was associated with poor survival and the infiltration of APCs. Two immune subtypes were identified; TM2 exhibited significantly improved survival and was more likely to benefit from vaccination compared with TM1. TM1 was associated with a relatively quiet microenvironment, high tumor mutation burden, and enriched DNA damage repair pathways. The immune checkpoints and immunogenic cell death modulators were also differentially expressed between two subtypes. Finally, FN1 was identified to be the hub gene. CONCLUSIONS FAM134B, ALDH3A2, SAV1, RORC, and FN1 are considered as possible and effective mRNA anti-MESO antigens for the development of an mRNA vaccine, and TM2 patients are the most suitable for vaccination.
Collapse
Affiliation(s)
- Shuhang Wang
- Clinical Cancer Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (S.W.); (P.M.); (Y.J.); (Y.Y.); (H.H.); (Y.F.); (N.J.); (H.M.)
| | - Yuqi Yang
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People’s Hospital, Guiyang 550002, China;
| | - Lu Li
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing 210018, China; (L.L.); (M.G.); (H.G.); (L.Y.); (Y.R.)
| | - Peiwen Ma
- Clinical Cancer Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (S.W.); (P.M.); (Y.J.); (Y.Y.); (H.H.); (Y.F.); (N.J.); (H.M.)
| | - Yale Jiang
- Clinical Cancer Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (S.W.); (P.M.); (Y.J.); (Y.Y.); (H.H.); (Y.F.); (N.J.); (H.M.)
| | - Minghui Ge
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing 210018, China; (L.L.); (M.G.); (H.G.); (L.Y.); (Y.R.)
| | - Yue Yu
- Clinical Cancer Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (S.W.); (P.M.); (Y.J.); (Y.Y.); (H.H.); (Y.F.); (N.J.); (H.M.)
| | - Huiyao Huang
- Clinical Cancer Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (S.W.); (P.M.); (Y.J.); (Y.Y.); (H.H.); (Y.F.); (N.J.); (H.M.)
| | - Yuan Fang
- Clinical Cancer Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (S.W.); (P.M.); (Y.J.); (Y.Y.); (H.H.); (Y.F.); (N.J.); (H.M.)
| | - Ning Jiang
- Clinical Cancer Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (S.W.); (P.M.); (Y.J.); (Y.Y.); (H.H.); (Y.F.); (N.J.); (H.M.)
| | - Huilei Miao
- Clinical Cancer Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (S.W.); (P.M.); (Y.J.); (Y.Y.); (H.H.); (Y.F.); (N.J.); (H.M.)
| | - Hao Guo
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing 210018, China; (L.L.); (M.G.); (H.G.); (L.Y.); (Y.R.)
| | - Linlin Yan
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing 210018, China; (L.L.); (M.G.); (H.G.); (L.Y.); (Y.R.)
| | - Yong Ren
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing 210018, China; (L.L.); (M.G.); (H.G.); (L.Y.); (Y.R.)
| | - Lichao Sun
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yan Zha
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People’s Hospital, Guiyang 550002, China;
| | - Ning Li
- Clinical Cancer Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (S.W.); (P.M.); (Y.J.); (Y.Y.); (H.H.); (Y.F.); (N.J.); (H.M.)
| |
Collapse
|
8
|
Tan ES, Knepper TC, Wang X, Permuth JB, Wang L, Fleming JB, Xie H. Copy Number Alterations as Novel Biomarkers and Therapeutic Targets in Colorectal Cancer. Cancers (Basel) 2022; 14:2223. [PMID: 35565354 PMCID: PMC9101426 DOI: 10.3390/cancers14092223] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 04/21/2022] [Accepted: 04/24/2022] [Indexed: 12/10/2022] Open
Abstract
In colorectal cancer, somatic mutations have played an important role as prognostic and predictive biomarkers, with some also functioning as therapeutic targets. Another genetic aberration that has shown significance in colorectal cancer is copy number alterations (CNAs). CNAs occur when a change to the DNA structure propagates gain/amplification or loss/deletion in sections of DNA, which can often lead to changes in protein expression. Multiple techniques have been developed to detect CNAs, including comparative genomic hybridization with microarray, low pass whole genome sequencing, and digital droplet PCR. In this review, we summarize key findings in the literature regarding the role of CNAs in the pathogenesis of colorectal cancer, from adenoma to carcinoma to distant metastasis, and discuss the roles of CNAs as prognostic and predictive biomarkers in colorectal cancer.
Collapse
Affiliation(s)
- Elaine S. Tan
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive Tampa, Tampa, FL 33612, USA; (E.S.T.); (J.B.P.); (J.B.F.)
| | - Todd C. Knepper
- Department of Individualized Cancer Management, H. Lee Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive Tampa, Tampa, FL 33612, USA;
| | - Xuefeng Wang
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive Tampa, Tampa, FL 33612, USA;
| | - Jennifer B. Permuth
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive Tampa, Tampa, FL 33612, USA; (E.S.T.); (J.B.P.); (J.B.F.)
| | - Liang Wang
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, 12901 USF Magnolia Drive Tampa, Tampa, FL 33612, USA;
| | - Jason B. Fleming
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive Tampa, Tampa, FL 33612, USA; (E.S.T.); (J.B.P.); (J.B.F.)
| | - Hao Xie
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive Tampa, Tampa, FL 33612, USA; (E.S.T.); (J.B.P.); (J.B.F.)
| |
Collapse
|
9
|
ER-phagy in the Occurrence and Development of Cancer. Biomedicines 2022; 10:biomedicines10030707. [PMID: 35327508 PMCID: PMC8945671 DOI: 10.3390/biomedicines10030707] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/11/2022] [Accepted: 03/16/2022] [Indexed: 02/04/2023] Open
Abstract
As an organelle, the endoplasmic reticulum (ER) is closely related to protein synthesis and modification. When physiological or pathological stimuli induce disorders of ER function, misfolded proteins trigger ER-phagy, which is beneficial for restoring cell homeostasis or promoting cell apoptosis. As a double-edged sword, ER-phagy actively participates in various stages of development and progression in tumor cells, regulating tumorigenesis and maintaining tumor cell homeostasis. Through the unfolded protein response (UPR), the B cell lymphoma 2 (BCL-2) protein family, the Caspase signaling pathway, and others, ER-phagy plays an initiating role in tumor occurrence, migration, stemness, and proliferation. At the same time, many vital proteins strongly associated with ER-phagy, such as family with sequence similarity 134 member B (FAM134B), translocation protein SEC62 (SEC62), and C/EBP-homologous protein (CHOP), can produce a marked effect in many complex environments, which ultimately lead to entirely different tumor fates. Our article comprehensively focused on introducing the relationship and interaction between ER-phagy and cancers, as well as their molecular mechanism and regulatory pathways. Via these analyses, we tried to clarify the possibility of ER-phagy as a potential target for cancer therapy and provide ideas for further research.
Collapse
|
10
|
Reggiori F, Molinari M. ER-phagy: mechanisms, regulation and diseases connected to the lysosomal clearance of the endoplasmic reticulum. Physiol Rev 2022; 102:1393-1448. [PMID: 35188422 PMCID: PMC9126229 DOI: 10.1152/physrev.00038.2021] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
ER-phagy (reticulo-phagy) defines the degradation of portions of the endoplasmic reticulum (ER) within lysosomes or vacuoles. It is part of the self-digestion (i.e., auto-phagic) programs recycling cytoplasmic material and organelles, which rapidly mobilize metabolites in cells confronted with nutrient shortage. Moreover, selective clearance of ER subdomains participates to the control of ER size and activity during ER stress, the re-establishment of ER homeostasis after ER stress resolution and the removal of ER parts, in which aberrant and potentially cytotoxic material has been segregated. ER-phagy relies on the individual and/or concerted activation of the ER-phagy receptors, ER peripheral or integral membrane proteins that share the presence of LC3/Atg8-binding motifs in their cytosolic domains. ER-phagy involves the physical separation of portions of the ER from the bulk ER network, and their delivery to the endolysosomal/vacuolar catabolic district. This last step is accomplished by a variety of mechanisms including macro-ER-phagy (in which ER fragments are sequestered by double-membrane autophagosomes that eventually fuse with lysosomes/vacuoles), micro-ER-phagy (in which ER fragments are directly engulfed by endosomes/lysosomes/vacuoles), or direct fusion of ER-derived vesicles with lysosomes/vacuoles. ER-phagy is dysfunctional in specific human diseases and its regulators are subverted by pathogens, highlighting its crucial role for cell and organism life.
Collapse
Affiliation(s)
- Fulvio Reggiori
- Department of Biomedical Sciences of Cells & Systems, grid.4830.fUniversity of Groningen, Netherlands
| | - Maurizio Molinari
- Protein Folding and Quality Control, grid.7722.0Institute for Research in Biomedicine, Bellinzona, Switzerland
| |
Collapse
|
11
|
Zhu L, Wang X, Wang Y. Roles of FAM134B in diseases from the perspectives of organelle membrane morphogenesis and cellular homeostasis. J Cell Physiol 2021; 236:7242-7255. [PMID: 33843059 DOI: 10.1002/jcp.30377] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 03/09/2021] [Accepted: 03/15/2021] [Indexed: 11/07/2022]
Abstract
Family with sequence similarity 134 member B (FAM134B)/RETREG1/JK1 is a novel gene with recently reported roles in various diseases. Understanding the function and mechanism of action of FAM134B is necessary to develop disease therapies. Notably, emerging data are clarifying the molecular mechanisms of FAM134B function in organelle membrane morphogenesis and the regulation of signaling pathways, such as the Wnt and AKT signaling pathways. In addition, transcription factors, RNA N6 -methyladenosine-mediated epigenetic regulation, microRNA, and small molecules are involved in the regulation of FAM134B expression. This review comprehensively considers recent studies on the role of FAM134B and its potential mechanisms in neurodegenerative diseases, obesity, viral diseases, cancer, and other diseases. The functions of FAM134B in maintaining cell homeostasis by regulating Golgi morphology, endoplasmic reticulum autophagy, and mitophagy are also highlighted, which may be the underlying mechanism of FAM134B gene mutation-induced diseases. Moreover, the molecular mechanisms of the FAM134B function during numerous biological processes are discussed. This review provides novel insights into the functions and mechanisms of FAM134B in various diseases, which will inform the development of effective drugs to treat diseases.
Collapse
Affiliation(s)
- Luoyi Zhu
- Key Laboratory of Molecular Nutrition, Ministry of Education, Zhejiang University, Hangzhou, Zhejiang, China.,Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, Zhejiang University, Hangzhou, Zhejiang, China.,Key Laboratory of Animal Nutrition and Feed Science of Zhejiang Province, Institute of Feed Science, College of Animal Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xinxia Wang
- Key Laboratory of Molecular Nutrition, Ministry of Education, Zhejiang University, Hangzhou, Zhejiang, China.,Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, Zhejiang University, Hangzhou, Zhejiang, China.,Key Laboratory of Animal Nutrition and Feed Science of Zhejiang Province, Institute of Feed Science, College of Animal Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yizhen Wang
- Key Laboratory of Molecular Nutrition, Ministry of Education, Zhejiang University, Hangzhou, Zhejiang, China.,Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, Zhejiang University, Hangzhou, Zhejiang, China.,Key Laboratory of Animal Nutrition and Feed Science of Zhejiang Province, Institute of Feed Science, College of Animal Science, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
12
|
Miller DR, Thorburn A. Autophagy and organelle homeostasis in cancer. Dev Cell 2021; 56:906-918. [PMID: 33689692 PMCID: PMC8026727 DOI: 10.1016/j.devcel.2021.02.010] [Citation(s) in RCA: 138] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/11/2021] [Accepted: 02/09/2021] [Indexed: 12/16/2022]
Abstract
Beginning with the earliest studies of autophagy in cancer, there have been indications that autophagy can both promote and inhibit cancer growth and progression; autophagy regulation of organelle homeostasis is similarly complicated. In this review we discuss pro- and antitumor effects of organelle-targeted autophagy and how this contributes to several hallmarks of cancer, such as evading cell death, genomic instability, and altered metabolism. Typically, the removal of damaged or dysfunctional organelles prevents tumor development but can also aid in proliferation or drug resistance in established tumors. By better understanding how organelle-specific autophagy takes place and can be manipulated, it may be possible to go beyond the brute-force approach of trying to manipulate all autophagy in order to improve therapeutic targeting of this process in cancer.
Collapse
Affiliation(s)
- Dannah R Miller
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Andrew Thorburn
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| |
Collapse
|
13
|
Liang C, Mu Y, Tian H, Wang D, Zhang S, Wang H, Liu Y, Di C. MicroRNA-140 silencing represses the incidence of Alzheimer's disease. Neurosci Lett 2021; 758:135674. [PMID: 33529652 DOI: 10.1016/j.neulet.2021.135674] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 12/28/2020] [Accepted: 01/20/2021] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative condition leading to severe disability from progressive impairments in cognitive functions including memory and learning. Non-coding microRNAs (miRNAs or miRs) have been linked to the pathogenesis of AD. The present study aimed to investigate the clinical significance and biological function of miR-140 in AD. First, we examined the expression of miR-140 and PINK1 in brain tissues of the established AD model rats and neurons cultured with Aβ-derived diffusible ligands (AβDDLs). We identified an interaction between miR-140 and PINK1, and measured spatial learning and memory abilities of the model rats using the Morris water maze (MWM) test. After ectopic expression and depletion experiments in neurons and AD rats, we measured the levels of reactive oxygen species (ROS), and mitochondrial membrane potential (MMP), along with mTOR expression and phosphorylation, and autophagy-related factors. Results showed up-regulation of miR-140 and down-regulation of PINK1 in AD model rats and neurons. PINK1 was verified to be a direct target of miR-140, and silencing of miR-140 suppressed mitochondrial dysfunction, and enhanced autophagy in AD model rats and neurons, as supported by decreased levels of mTOR expression and phosphorylation, β-amyloid p-Tau (Ser396), p-Tau (Thr231), Tau and ROS, and increased MMP levels and expression of Beclin 1 expression and LC3-II/LC3-I. Collectively, functional suppression of miR-140 enhanced autophagy and prevented mitochondrial dysfunction by upregulating PINK1, ultimately suggesting a novel therapeutic target for AD.
Collapse
Affiliation(s)
- Chunming Liang
- The First Department of Neurology, The Second Affiliated Hospital of Qiqihar Medical University, Qiqihar, 161000, PR China.
| | - Yuyuan Mu
- The First Department of Neurology, The Second Affiliated Hospital of Qiqihar Medical University, Qiqihar, 161000, PR China
| | - Hua Tian
- Qiqihar Medical University, Qiqihar, 161006, PR China
| | - Dan Wang
- Department of Neurology, The First Hospital of Qiqihar (Qiqihar Hospital Affiliated to Southern Medical University), Qiqihar, 161000, PR China
| | - Shicun Zhang
- The Fourth Department of Neurology, The Second Affiliated Hospital of Qiqihar Medical University, Qiqihar, 161000, PR China
| | - Hongjiao Wang
- The First Department of Neurology, The Second Affiliated Hospital of Qiqihar Medical University, Qiqihar, 161000, PR China
| | - Yang Liu
- The First Department of Neurology, The Second Affiliated Hospital of Qiqihar Medical University, Qiqihar, 161000, PR China
| | - Cihan Di
- The First Department of Neurology, The Second Affiliated Hospital of Qiqihar Medical University, Qiqihar, 161000, PR China
| |
Collapse
|
14
|
Loi M, Marazza A, Molinari M. Endoplasmic Reticulum (ER) and ER-Phagy. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2021; 59:99-114. [PMID: 34050863 DOI: 10.1007/978-3-030-67696-4_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The endoplasmic reticulum (ER) is a biosynthetic organelle in eukaryotic cells. Its capacity to produce proteins, lipids and oligosaccharides responds to physiologic and pathologic demand. The transcriptional and translational unfolded protein response (UPR) programs increase ER size and activity. In contrast, ER-phagy programs in all their flavors select ER subdomains for lysosomal clearance. These programs are activated by nutrient deprivation, accumulation of excess ER (recov-ER-phagy), production of misfolded proteins that cannot be degraded by ER-associated degradation and that are removed from cells by the so-called ER-to-lysosome-associated degradation (ERLAD). Selection of ER subdomains to be cleared from cells relies on ER-phagy receptors, a class of membrane-bound proteins displaying cytosolic domains that engage the cytosolic ubiquitin-like protein LC3. Mechanistically, ER clearance proceeds via macro-ER-phagy, micro-ER-phagy and LC3-regulated vesicular delivery.
Collapse
Affiliation(s)
- Marisa Loi
- Università della Svizzera italiana, Lugano, Switzerland
- Institute for Research in Biomedicine, Bellinzona, Switzerland
| | - Alessandro Marazza
- Università della Svizzera italiana, Lugano, Switzerland
- Institute for Research in Biomedicine, Bellinzona, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Maurizio Molinari
- Università della Svizzera italiana, Lugano, Switzerland.
- Institute for Research in Biomedicine, Bellinzona, Switzerland.
- Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
| |
Collapse
|
15
|
Keles U, Iscan E, Yilmaz HE, Karakülah G, Suner A, Bal E, Tasdemir N, Cavga AD, Ekin U, Mutlu Z, Kahyaoglu S, Serdar MA, Atabey N, Ozturk M. Differential expression of full-length and NH 2 terminally truncated FAM134B isoforms in normal physiology and cancer. Am J Physiol Gastrointest Liver Physiol 2020; 319:G733-G747. [PMID: 33052704 PMCID: PMC7864244 DOI: 10.1152/ajpgi.00094.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Selective autophagy of the endoplasmic reticulum (ER), namely ER-phagy, is mediated by ER-localized receptors, which are recognized and sequestered by GABARAP/LC3B-decorated phagophores and transferred to lysosomes for degradation. Being one such receptor, FAM134B plays critical roles in cellular processes such as protein quality control and neuronal survival. FAM134B has also been associated with different cancers, although its exact role remains elusive. We report here that the FAM134B gene encodes not one but at least two different protein isoforms: the full-length and the NH2 terminally truncated forms. Their relative expression shows extreme variation, both within normal tissues and among cancer types. Expression of full-length FAM134B is restricted to the brain, testis, spleen, and prostate. In contrast, NH2 terminally truncated FAM134B is dominant in the heart, skeletal muscle, kidney, pancreas, and liver. We compared wild-type and knockout mice to study the role of the Fam134b gene in starvation. NH2 terminally truncated FAM134B-2 was induced in the liver, skeletal muscle, and heart but not in the pancreas and stomach following starvation. Upon starvation, Fam134b-/- mice differed from wild-type mice by less weight loss and less hyperaminoacidemic and hypocalcemic response but increased levels of serum albumin, total serum proteins, and α-amylase. Interestingly, either NH2 terminally truncated FAM134B or both isoforms were downregulated in liver, lung, and colon cancers. In contrast, upregulation was observed in stomach and chromophobe kidney cancers.NEW & NOTEWORTHY We reported tissues expressing FAM134B-2 such as the kidney, muscle, heart, and pancreas, some of which exhibit stimulated expression upon nutrient starvation. We also demonstrated the effect of Fam134b deletion during ad libitum and starvation conditions. Resistance to weight loss and hypocalcemia, accompanied by an increase in serum albumin and α-amylase levels, indicate critical roles of Fam134b in physiology. Furthermore, the differential expression of FAM134B isoforms was shown to be significantly dysregulated in human cancers.
Collapse
Affiliation(s)
- Umur Keles
- 1Izmir Biomedicine and Genome Center, Izmir, Turkey,2Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Evin Iscan
- 1Izmir Biomedicine and Genome Center, Izmir, Turkey,2Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Huriye Erbak Yilmaz
- 2Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Gökhan Karakülah
- 1Izmir Biomedicine and Genome Center, Izmir, Turkey,2Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Aslı Suner
- 3Department of Biostatistics and Medical Informatics, Ege University, Izmir, Turkey
| | - Erhan Bal
- 1Izmir Biomedicine and Genome Center, Izmir, Turkey
| | - Nilgun Tasdemir
- 4Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Ayse Derya Cavga
- 4Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Umut Ekin
- 1Izmir Biomedicine and Genome Center, Izmir, Turkey,2Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Zeynep Mutlu
- 1Izmir Biomedicine and Genome Center, Izmir, Turkey
| | | | | | - Nese Atabey
- 1Izmir Biomedicine and Genome Center, Izmir, Turkey
| | | |
Collapse
|
16
|
Mo J, Chen J, Zhang B. Critical roles of FAM134B in ER-phagy and diseases. Cell Death Dis 2020; 11:983. [PMID: 33199694 PMCID: PMC7670425 DOI: 10.1038/s41419-020-03195-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 10/05/2020] [Accepted: 10/08/2020] [Indexed: 12/14/2022]
Abstract
FAM134B (also called JK-1, RETREG1), a member of the family with sequence similarity 134, was originally discovered as an oncogene in esophageal squamous cell carcinoma. However, its most famous function is that of an ER-phagy-regulating receptor. Over the decades, the powerful biological functions of FAM134B were gradually revealed. Overwhelming evidence indicates that its dysfunction is related to pathophysiological processes such as neuropathy, viral replication, inflammation, and cancer. This review describes the biological functions of FAM134B, focusing on its role in ER-phagy. In addition, we summarize the diseases in which it is involved and review the underlying mechanisms.
Collapse
Affiliation(s)
- Jie Mo
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases; Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Clinical Medicine Research Center for Hepatic Surgery of Hubei Province; Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, Hubei, 430030, P.R. China
| | - Jin Chen
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases; Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Clinical Medicine Research Center for Hepatic Surgery of Hubei Province; Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, Hubei, 430030, P.R. China
| | - Bixiang Zhang
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases; Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Clinical Medicine Research Center for Hepatic Surgery of Hubei Province; Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, Hubei, 430030, P.R. China.
| |
Collapse
|
17
|
Lee KTW, Islam F, Vider J, Martin J, Chruścik A, Lu CT, Gopalan V, Lam AKY. Overexpression of family with sequence similarity 134, member B (FAM134B) in colon cancers and its tumor suppressive properties in vitro. Cancer Biol Ther 2020; 21:954-962. [PMID: 32857678 PMCID: PMC7583494 DOI: 10.1080/15384047.2020.1810535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 07/29/2020] [Accepted: 08/08/2020] [Indexed: 01/11/2023] Open
Abstract
This study aims to investigate the overexpression-induced properties of tumor suppressor FAM134B (family with sequence similarity 134, member B) in colon cancer, examine the potential gene regulators of FAM134B expression and its impact on mitochondrial function. FAM134B was overexpressed in colon cancer and non-neoplastic colonic epithelial cells. Various cell-based assays including apoptosis, cell cycle, cell proliferation, clonogenic, extracellular flux and wound healing assays were performed. Western blot analysis was used to confirm and identify potential interacting partners of FAM134B in vitro. Immunohistochemistry and qPCR were employed to determine the expressions of MIF and FAM134B, respectively, on 63 patients with colorectal carcinoma. Results showed that FAM134B is involved in the cell cycle and mitochondrial function of colon cancer. Overexpression of FAM134B was coupled with increased expression levels of APC, p53, and MIF. Increased expression of both APC and p53 further validates the potential role of tumor suppressor FAM134B in regulating cancer progression through the WNT/ß-catenin signaling pathway. In approximately 70% of the patients with colorectal cancer, FAM134B downregulation was correlated with MIF protein overexpression while the remaining 30% showed concurrent expression of FAM134B and MIF (P = .045). High expression of MIF coupled with low expression of FAM134B is associated with microsatellite instability status in colorectal carcinomas (P = .049). FAM134B may exert its tumor suppressive function through affecting cell cycle, mitochondrial function via potentially interacting with MIF and p53.
Collapse
Affiliation(s)
- Katherine Ting-Wei Lee
- Cancer Molecular Pathology, School of Medicine, Griffith University, Gold Coast, Australia
| | - Farhadul Islam
- Cancer Molecular Pathology, School of Medicine, Griffith University, Gold Coast, Australia
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, Bangladesh
| | - Jelena Vider
- School of Medical Science, Griffith University, Gold Coast, Australia
| | - Jeremy Martin
- Cancer Molecular Pathology, School of Medicine, Griffith University, Gold Coast, Australia
| | - Anna Chruścik
- Cancer Molecular Pathology, School of Medicine, Griffith University, Gold Coast, Australia
| | - Cu-Tai Lu
- Department of Surgery, Gold Coast University Hospital, Gold Coast, Australia
| | - Vinod Gopalan
- Cancer Molecular Pathology, School of Medicine, Griffith University, Gold Coast, Australia
| | - Alfred King-yin Lam
- Cancer Molecular Pathology, School of Medicine, Griffith University, Gold Coast, Australia
| |
Collapse
|
18
|
Song Q, Zhao F, Yao J, Dai H, Hu L, Yu S. Protective effect of microRNA-134-3p on multiple sclerosis through inhibiting PRSS57 and promotion of CD34 + cell proliferation in rats. J Cell Biochem 2020; 121:4347-4363. [PMID: 32619071 DOI: 10.1002/jcb.29643] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 12/19/2019] [Indexed: 12/23/2022]
Abstract
MicroRNAs (miRs) have been extensively studied for their involvement in multiple sclerosis (MS). We investigated the involvement of miR-134-3p on MS. The MS rat model was established, and positive expression of interleukin-17 (IL-17) was detected using the immunohistochemical method while the expression of miR-134-3p and serine protease 57 (PRSS57) was determined by means of reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and Western blot analysis. Second, the miR-134-3p overexpression or short hairpin RNA against PRSS57 was introduced into the CD34+ cells to investigate the levels of proliferation and apoptosis-related genes by RT-qPCR and Western blot analysis. In addition, analysis of the targeting relations of miR-134-3p and PRSS57 was conducted using online software and dual-luciferase reporter gene assay. Furthermore, neuronal functions, inflammatory response, proliferation, and apoptosis of CD34+ cells were assayed by flow cytometry, enzyme-linked immunosorbent assay, and methyl thiazolyl tetrazolium. IL-17 and PRSS57 expression increased while miR-134-3p expression decreased in the spinal cord from MS rats. miR-134-3p could target PRSS57. miR-134-3p overexpression or PRSS57 silencing enhanced mitochondrial activity of neurons, mitochondrial membrane potential content, CD34+ cell proliferation, while decreasing Cyt C content, inflammatory response, and cell apoptosis. Collectively, overexpression of miR-134-3p promotes CD34+ cell proliferation via inhibition of PRSS57 in MS, which may serve as a promising target for MS intervention.
Collapse
Affiliation(s)
- Qihan Song
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Department of Neurology, The No.2 Hospital of Baoding, Baoding, China
| | - Fengli Zhao
- Department of Neurology, The No.2 Hospital of Baoding, Baoding, China
| | - Jingfan Yao
- Department of Neurology, Beijing Tiantan Hospital of Capital Medical University, Beijing, China
| | - Hailin Dai
- Department of Neurology, The No.2 Hospital of Baoding, Baoding, China
| | - Lei Hu
- Department of Neurology, The No.2 Hospital of Baoding, Baoding, China
| | - Shun Yu
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China
| |
Collapse
|
19
|
HOTAIR drives autophagy in midbrain dopaminergic neurons in the substantia nigra compacta in a mouse model of Parkinson's disease by elevating NPTX2 via miR-221-3p binding. Aging (Albany NY) 2020; 12:7660-7678. [PMID: 32396526 PMCID: PMC7244061 DOI: 10.18632/aging.103028] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 02/05/2020] [Indexed: 01/17/2023]
Abstract
Parkinson’s disease (PD) is a neurodegenerative disorder characterized by progressive cell loss, largely confined to mesencephalic dopamine neurons of the substantia nigra. This study investigated the functional relevance of the HOX transcript antisense intergenic RNA (HOTAIR)/microRNA-221-3 (miR-221-3p)/neuronal pentraxin II (NPTX2) axis in the process of dopaminergic neuron autophagy using PD mouse models. The PD mouse models were established by intraperitoneal injection of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyrindine (MPTP), while PD cell model was constructed by pretreatment with 1-methyl-4-phenylpyridinium (MPP+). The expression of HOTAIR was then examined using RT-qPCR. In addition, the interactions between HOTAIR, miR-221-3p, and NPTX2 were detected through RIP and dual-luciferase reporter gene assays. CCK-8 assay was performed to measure cell viability, and the expression of autophagy-related genes was determined using Western blot analysis. HOTAIR was found to be significantly expressed in the substantia nigra compact tissues and MN9D cells following PD modeling. HOTAIR could bind to miR-221-3p and elevate the NPTX2 expression, which resulted in diminished cell viability and enhanced autophagy of dopaminergic neurons both in vitro and in vivo. In summary, down-regulation of HOTAIR could potentially inhibit the autophagy of dopaminergic neurons in the substantia nigra compacta in a mouse model of PD, thus saving the demise of dopaminergic neurons.
Collapse
|
20
|
Quinchia J, Echeverri D, Cruz-Pacheco AF, Maldonado ME, Orozco J. Electrochemical Biosensors for Determination of Colorectal Tumor Biomarkers. MICROMACHINES 2020; 11:E411. [PMID: 32295170 PMCID: PMC7231317 DOI: 10.3390/mi11040411] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/13/2020] [Accepted: 03/17/2020] [Indexed: 12/15/2022]
Abstract
The accurate determination of specific tumor markers associated with cancer with non-invasive or minimally invasive procedures is the most promising approach to improve the long-term survival of cancer patients and fight against the high incidence and mortality of this disease. Quantification of biomarkers at different stages of the disease can lead to an appropriate and instantaneous therapeutic action. In this context, the determination of biomarkers by electrochemical biosensors is at the forefront of cancer diagnosis research because of their unique features such as their versatility, fast response, accurate quantification, and amenability for multiplexing and miniaturization. In this review, after briefly discussing the relevant aspects and current challenges in the determination of colorectal tumor markers, it will critically summarize the development of electrochemical biosensors to date to this aim, highlighting the enormous potential of these devices to be incorporated into the clinical practice. Finally, it will focus on the remaining challenges and opportunities to bring electrochemical biosensors to the point-of-care testing.
Collapse
Affiliation(s)
- Jennifer Quinchia
- Max Planck Tandem Group in Nanobioengineering, University of Antioquia, Complejo Ruta N, Calle 67 No. 52-20, Medellín 050010, Colombia; (J.Q.); (D.E.); (A.F.C.-P.)
| | - Danilo Echeverri
- Max Planck Tandem Group in Nanobioengineering, University of Antioquia, Complejo Ruta N, Calle 67 No. 52-20, Medellín 050010, Colombia; (J.Q.); (D.E.); (A.F.C.-P.)
| | - Andrés Felipe Cruz-Pacheco
- Max Planck Tandem Group in Nanobioengineering, University of Antioquia, Complejo Ruta N, Calle 67 No. 52-20, Medellín 050010, Colombia; (J.Q.); (D.E.); (A.F.C.-P.)
| | - María Elena Maldonado
- Grupo Impacto de los Componentes Alimentarios en la Salud, School of Dietetics and Human Nutrition, University of Antioquia, A.A. 1226, Medellín 050010, Colombia;
| | - Jahir Orozco
- Max Planck Tandem Group in Nanobioengineering, University of Antioquia, Complejo Ruta N, Calle 67 No. 52-20, Medellín 050010, Colombia; (J.Q.); (D.E.); (A.F.C.-P.)
| |
Collapse
|
21
|
Abstract
We developed a new label-free biosensor technique for the detection of messenger ribonucleic acid (mRNA) that can be used in the prognosis and diagnosis of certain diseases. We observed a dependence of the negative dielectrophoresis (DEP) force applied to polystyrene microspheres (PMs) in conjugation with different types of mRNA and the frequency of the electric field produced by interdigitated microelectrodes. Since the frequency dependence of the negative DEP force is an effective transduction mechanism for the detection of mRNA, this sensing technology has the potential to be used in the diagnosis and identification of gene expression that is associated with various human disease.
Collapse
|
22
|
Li F, Dai L, Niu J. GPX2 silencing relieves epithelial-mesenchymal transition, invasion, and metastasis in pancreatic cancer by downregulating Wnt pathway. J Cell Physiol 2019; 235:7780-7790. [PMID: 31774184 DOI: 10.1002/jcp.29391] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 09/27/2019] [Indexed: 12/13/2022]
Abstract
Glutathione peroxidase 2 (GPX2) participates in many cancers including pancreatic cancer (PC), and overexpression of GPX2 promotes tumor growth. Herein, we identified the role of GPX2 in epithelial-mesenchymal transformation (EMT), invasion, and metastasis in PC. Bioinformatics prediction was applied to select PC-related genes. The regulatory function of GPX2 in PC was explored by treatment with short hairpin RNA against GPX2 or LiCl (activator of wingless-type MMTV integration site [Wnt] pathway) in PC cells. GPX2 level in PC tissues, the levels of GPX2, β-catenin, Vimentin, Snail, epithelial-cadherin (E-cadherin), matrix metalloproteinase 2 (MMP2), MMP9, and Wnt2 in cells were determined. Subsequently, cell proliferation, invasion, and metastasis were assayed. Bioinformatics analysis revealed that GPX2 was involved in PC development mediated by the Wnt pathway. GPX2 was highly expressed in PC tissues. GPX2 silencing downregulated levels of β-catenin, Vimentin, Snail, MMP2, MMP9, and Wnt2 but upregulated levels of E-cadherin. It was confirmed that GPX2 silencing suppressed PC cell proliferation, metastasis, and invasion. Furthermore, the trend of EMT and invasion and metastasis of PC induced by the LiCl-activated Wnt pathway was reversed when the GPX2 was silenced. GPX2 silencing could inhibit the Wnt pathway, subsequently suppress PC development.
Collapse
Affiliation(s)
- Fuzhou Li
- Department of Imaging, Linyi People's Hospital, Linyi, China
| | - Lan Dai
- Department of Gynaecology and Obstetrics, Chinese Medicine Hospital of Linyi City, Linyi, China
| | - Jixiang Niu
- Department of General Surgery, Linyi People's Hospital, Linyi, China
| |
Collapse
|
23
|
Chen Q, Zhang Y, Zhu H, Yuan X, Luo X, Wu X, Chen S, Chen Y, Xu J, Issa HA, Zheng Z, Hu J, Yang T. Bone marrow mesenchymal stem cells alleviate the daunorubicin-induced subacute myocardial injury in rats through inhibiting infiltration of T lymphocytes and antigen-presenting cells. Biomed Pharmacother 2019; 121:109157. [PMID: 31731195 DOI: 10.1016/j.biopha.2019.109157] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 06/19/2019] [Accepted: 06/19/2019] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Bone marrow mesenchymal stem cells (BMSCs) have been extensively investigated from a perspective on cardiac regeneration therapy. The current study aimed to investigate the protective effect conferred by BMSCs in subacute myocardial injury, and to identify an appropriate BMSC reinfusion time. METHODS BMSCs were isolated from human bone marrow blood. Daunorubicin (DNR)-induced subacute myocardial models were subsequently established. The rats with DNR-induced subacute myocardial injury were injected with dexrazoxane (DZR) and/or BMSCs at varying time points, after which cardiac function was evaluated by assessing left ventricular ejection fraction (LVEF) and fraction shortening (FS). The myocardial structural changes were analyzed, after which the levels of CD3 and human leukocyte antigen DR (HLA-DR) were examined to further validate the mechanism by which BMSCs could influence subacute myocardial injury. RESULTS BMSCs combined with DZR treatment enhanced the cardiac function of rats with DNR-induced myocardial injury, as reflected by increased LVEF and FS. DNR-induced myocardial injuries were mitigated via the application of BMSCs combined with treatment of DZR, accompanied by diminished infiltration or vacuolization. Moreover, BMSCs were observed to alleviate infiltration of T lymphocyte and antigen-presenting cells, as evidenced by reduced expression of CD3 and HLA-DR. CONCLUSION Taken together, this study demonstrates that BMSCs could protect against DNR-induced myocardial injury, especially in the first three days of DNR administration. BMSCs combined with DZR exert a better therapeutic effect, but there are individual differences.
Collapse
Affiliation(s)
- Qiuru Chen
- Department of Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou 350000, PR China
| | - Yuxin Zhang
- Department of Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou 350000, PR China
| | - Haojie Zhu
- Department of Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou 350000, PR China
| | - Xiaohong Yuan
- Department of Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou 350000, PR China
| | - Xiaofeng Luo
- Department of Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou 350000, PR China
| | - Xueqiong Wu
- Department of Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou 350000, PR China
| | - Shaozhen Chen
- Department of Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou 350000, PR China
| | - Yongquan Chen
- Department of Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou 350000, PR China
| | - Jingjing Xu
- Department of Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou 350000, PR China
| | - Hajji Ally Issa
- Department of Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou 350000, PR China
| | - Zhihong Zheng
- Department of Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou 350000, PR China
| | - Jianda Hu
- Department of Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou 350000, PR China.
| | - Ting Yang
- Department of Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou 350000, PR China.
| |
Collapse
|
24
|
ER-phagy and human diseases. Cell Death Differ 2019; 27:833-842. [PMID: 31659280 DOI: 10.1038/s41418-019-0444-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 10/04/2019] [Accepted: 10/11/2019] [Indexed: 12/27/2022] Open
Abstract
Autophagy regulates the degradation of unnecessary or dysfunctional cellular components. This catabolic process requires the formation of a double-membrane vesicle, the autophagosome, that engulfs the cytosolic material and delivers it to the lysosome. Substrate specificity is achieved by autophagy receptors, which are characterized by the presence of at least one LC3-interaction region (LIR) or GABARAP-interaction motif (GIM). Only recently, several receptors that mediate the specific degradation of endoplasmic reticulum (ER) components via autophagy have been identified (the process known as ER-phagy or reticulophagy). Here, we give an update on the current knowledge about the role of ER-phagy receptors in health and disease.
Collapse
|
25
|
Wang L, Zhang X, Liu Y, Xu S. Long noncoding RNA FBXL19-AS1 induces tumor growth and metastasis by sponging miR-203a-3p in lung adenocarcinoma. J Cell Physiol 2019; 235:3612-3625. [PMID: 31566718 DOI: 10.1002/jcp.29251] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 09/03/2019] [Indexed: 12/12/2022]
Abstract
The pivotal roles of long noncoding RNAs have been reported in various cancers. Recently, FBXL19-AS1 was proposed to be involved in tumor progression. However, its role in lung adenocarcinoma (LUAD) remains elusive. In this study, we observed that FBXL19-AS1 was significantly upregulated in LUAD tissues and high FBXL19-AS1 expression in LUAD was associated with a poor prognosis. Nevertheless, miR-203-3p showed the opposite effect. Moreover, cell viability and apoptosis analysis revealed that FBXL19-AS1 knockdown could arrest LUAD cells in G0/G1 phase and inhibit cell proliferation, migration and invasion in vitro and inhibited LUAD tumor progress in vivo. Mechanistically, we identified FBXL19-AS1 could act as a miR-203a-3p sponge using dual-luciferase reporter assay. In addition, we demonstrated that downregulation of miR-203a-3p reversed growth inhibition of LUAD cells caused by FBXL19-AS1 knockdown. Finally, FBXL19-AS1/miR-203a-3p axis was found to associate with baculoviral IAP repeat-containing protein 5.1-A-like (survivin), distal-less homeobox 5, E2F transcription factor 1, and zinc finger E-box binding homeobox 2 to regulate metastasis in LUAD cells. This study reveals a significance and mechanism of FBXL19-AS1 in LUAD proliferation and metastasis and offers a potential prognostic marker and a therapeutic target for patients with LUAD.
Collapse
Affiliation(s)
- Liming Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xin Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yang Liu
- Department of Pharmacy, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Shun Xu
- Department of Thoracic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
26
|
Kim MS, Kim D, Kim JR. Stage-Dependent Gene Expression Profiling in Colorectal Cancer. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2019; 16:1685-1692. [PMID: 29994071 DOI: 10.1109/tcbb.2018.2814043] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Temporal gene expression profiles have been widely considered to uncover the mechanism of cancer development and progression. Gene expression patterns, however, have been analyzed for limited stages with small samples, without proper data pre-processing, in many cases. With those approaches, it is difficult to unveil the mechanism of cancer development over time. In this study, we analyzed gene expression profiles of two independent colorectal cancer sample datasets, each of which contained 556 and 566 samples, respectively. To find specific gene expression changes according to cancer stage, we applied the linear mixed-effect regression model (LMER) that controls other clinical variables. Based on this methodology, we found two types of gene expression patterns: continuously increasing and decreasing genes as cancer develops. We found that continuously increasing genes are related to the nervous and developmental system, whereas the others are related to the cell cycle and metabolic processes. We further analyzed connected sub-networks related to the two types of genes. From these results, we suggest that the gene expression profile analysis can be used to understand underlying the mechanisms of cancer development such as cancer growth and metastasis. Furthermore, our approach can provide a good guideline for advancing our understanding of cancer developmental processes.
Collapse
|
27
|
Islam F, Gopalan V, Law S, Tang JCO, Lam AKY. FAM134B promotes esophageal squamous cell carcinoma in vitro and its correlations with clinicopathologic features. Hum Pathol 2019; 87:1-10. [PMID: 30794892 DOI: 10.1016/j.humpath.2018.11.033] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 11/07/2018] [Accepted: 11/12/2018] [Indexed: 01/06/2023]
Abstract
Family with sequence similarity 134, member B (FAM134B) is an autophagy regulator of endoplasmic reticulum first discovered to be involved in the pathogenesis of esophageal squamous cell carcinoma (ESCC). The present study examined the functional behavior of FAM134B in cancer cells and the association of FAM134B expression with clinicopathologic factors in patients with ESCC. Expression at both the mRNA and protein levels was investigated using real-time polymerase chain reaction and immunohistochemistry. The results were correlated with the clinical and pathological features of the patients. In addition, in vitro functional assays were used to investigate the roles of FAM134B in ESCC cells in response to gene silencing with shRNA lentiviral particles. Overexpression of FAM134B mRNA and protein was present in 31.2% (n = 29/93) and 36.6% (n = 41/112), respectively, in tumors, whereas downregulation occurred in 39.8% (n = 37/93) and 63.4% (n = 71/112), respectively. Expression of FAM134B protein in ESCC correlated with histologic grade (P = .002) and pathologic stage (P = .012). In vitro suppression of FAM134B in ESCC induced significant reductions of cell proliferation and colony formation (P < .05). In addition, suppression of FAM134B caused reduction of wound healing, migration, and invasion capacities of ESCC. To conclude, FAM134B could play crucial roles in the initiation and progression of ESCC, and FAM134B protein expression has potential predictive value. Therefore, development of strategies targeting FAM134B could have therapeutic value in the management of patients with ESCC.
Collapse
Affiliation(s)
- Farhadul Islam
- Department of Cancer Molecular Pathology, School of Medicine, Griffith University, Gold Coast, Queensland, Australia; Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi-6205, Bangladesh
| | - Vinod Gopalan
- Department of Cancer Molecular Pathology, School of Medicine, Griffith University, Gold Coast, Queensland, Australia
| | - Simon Law
- Department of Surgery, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| | - Johnny Cheuk-On Tang
- State Key Laboratory of Chirosciences, Lo Ka Chung Centre for Natural Anti-cancer Drug Development, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong.
| | - Alfred King-Yin Lam
- Department of Cancer Molecular Pathology, School of Medicine, Griffith University, Gold Coast, Queensland, Australia.
| |
Collapse
|
28
|
Zhang Z, Chen J, Huang W, Ning D, Liu Q, Wang C, Zhang L, Ren L, Chu L, Liang H, Fan H, Zhang B, Chen X. FAM134B induces tumorigenesis and epithelial-to-mesenchymal transition via Akt signaling in hepatocellular carcinoma. Mol Oncol 2019; 13:792-810. [PMID: 30556279 PMCID: PMC6441892 DOI: 10.1002/1878-0261.12429] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 11/02/2018] [Accepted: 11/13/2018] [Indexed: 01/03/2023] Open
Abstract
Fam134b (JK-1, RETREG1) was first identified as an oncogene in esophageal squamous cell carcinoma. However, the roles of FAM134B during tumorigenesis of hepatocellular carcinoma (HCC) and in epithelial-to-mesenchymal transition (EMT) were previously unclear. In this study, we investigated the function of FAM134B in HCC and the related tumorigenesis mechanisms, as well as how FAM134B induces EMT. We detected the expression of FAM134B in a normal hepatic cell line, HCC cell lines, fresh specimens, and a HCC tissue microarray. A retrospective study of 122 paired HCC tissue microarrays was used to analyze the correlation between FAM134B and clinical features. Gain- and loss-of-function experiments, rescue experiments, Akt pathway activator/inhibitors, nude mice xenograft models, and nude mice lung metastasis models were used to determine the underlying mechanisms of FAM134B in inducing tumorigenesis and EMT in vitro and in vivo. The expression level of FAM134B was highly elevated in HCC, as compared with that in normal liver tissues and normal hepatic cells. Overexpression of FAM134B was significantly associated with tumor size (P = 0.025), pathological vascular invasion (P = 0.026), differentiation grade (P = 0.023), cancer recurrence (P = 0.044), and portal vein tumor thrombus (P = 0.036) in HCC. Patients with high expression of FAM134B had shorter overall survival and disease-free survival than patients with non-high expression of FAM134B. Furthermore, knockdown of FAM134B with shRNAs inhibited cell growth and motility, as well as tumor formation and metastasis in nude mice, all of which were promoted by overexpression of FAM134B. Our study demonstrated that Fam134b is an oncogene that plays a crucial role in HCC via the Akt signaling pathway with subsequent glycogen synthase kinase-3β phosphorylation, accumulation of β-catenin, and stabilization of Snail, which promotes tumorigenesis, EMT, and tumor metastasis in HCC.
Collapse
Affiliation(s)
- Zhao‐qi Zhang
- Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Organ TransplantationMinistry of Education and Ministry of Public HealthWuhanChina
- Clinical Medicine Research Center of Hepatic Surgery in Hubei ProvinceWuhanChina
| | - Jin Chen
- Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Organ TransplantationMinistry of Education and Ministry of Public HealthWuhanChina
- Clinical Medicine Research Center of Hepatic Surgery in Hubei ProvinceWuhanChina
| | - Wan‐qiu Huang
- Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Organ TransplantationMinistry of Education and Ministry of Public HealthWuhanChina
- Clinical Medicine Research Center of Hepatic Surgery in Hubei ProvinceWuhanChina
| | - Deng Ning
- Department of Biliary and Pancreatic SurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Qiu‐meng Liu
- Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Organ TransplantationMinistry of Education and Ministry of Public HealthWuhanChina
- Clinical Medicine Research Center of Hepatic Surgery in Hubei ProvinceWuhanChina
| | - Chao Wang
- Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Organ TransplantationMinistry of Education and Ministry of Public HealthWuhanChina
- Clinical Medicine Research Center of Hepatic Surgery in Hubei ProvinceWuhanChina
| | - Long Zhang
- Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Organ TransplantationMinistry of Education and Ministry of Public HealthWuhanChina
- Clinical Medicine Research Center of Hepatic Surgery in Hubei ProvinceWuhanChina
| | - Li Ren
- Department of Hepatopancreatobiliary SurgeryAffiliated Hospital of Qinghai UniversityXiningChina
| | - Liang Chu
- Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Organ TransplantationMinistry of Education and Ministry of Public HealthWuhanChina
- Clinical Medicine Research Center of Hepatic Surgery in Hubei ProvinceWuhanChina
| | - Hui‐fang Liang
- Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Organ TransplantationMinistry of Education and Ministry of Public HealthWuhanChina
- Clinical Medicine Research Center of Hepatic Surgery in Hubei ProvinceWuhanChina
| | - Hai‐ning Fan
- Department of Hepatopancreatobiliary SurgeryAffiliated Hospital of Qinghai UniversityXiningChina
| | - Bi‐xiang Zhang
- Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Organ TransplantationMinistry of Education and Ministry of Public HealthWuhanChina
- Clinical Medicine Research Center of Hepatic Surgery in Hubei ProvinceWuhanChina
| | - Xiao‐ping Chen
- Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Organ TransplantationMinistry of Education and Ministry of Public HealthWuhanChina
- Clinical Medicine Research Center of Hepatic Surgery in Hubei ProvinceWuhanChina
| |
Collapse
|
29
|
Hu X, Liu Y, Ai P, He S, Liu L, Chen C, Tan Y, Wang T. MicroRNA-186 promotes cell proliferation and inhibits cell apoptosis in cutaneous squamous cell carcinoma by targeting RETREG1. Exp Ther Med 2019; 17:1930-1938. [PMID: 30867688 DOI: 10.3892/etm.2019.7154] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 09/28/2018] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs (miRs), a class of small non-coding RNAs, have been demonstrated to be involved in the development and progression of human malignancies, including cutaneous squamous cell carcinoma (CSCC). miR-186 serves a suppressive role in certain common types of human cancer; however, its exact function in CSCC has not been reported previously. In the present study, the expression of miR-186 was significantly increased in CSCC tissues compared with adjacent non-tumour tissues. Overexpression of miR-186 significantly promoted CSCC cell proliferation while inhibiting cell apoptosis. Reticulophagy regulator 1 (RETREG1), a gene that is significantly downregulated in CSCC tissues and cell lines, was identified as a novel target of miR-186. In addition, the expression of RETREG1 was inversely correlated with miR-186 expression in CSCC tissues. Furthermore, the expression of RETREG1 was negatively regulated by miR-186 in CSCC cells, and restoration of RETREG1 attenuated the effects of miR-186 on CSCC cells. Taken together, the results of the current study suggest that miR-186 serves an oncogenic role in CSCC and may be used as a potential therapeutic target for the treatment of this disease.
Collapse
Affiliation(s)
- Xinde Hu
- Clinical Laboratory, Second People's Hospital of Shaoyang City, Shaoyang, Hunan 422000, P.R. China
| | - Yifeng Liu
- Department of Dermatology, Second People's Hospital of Shaoyang City, Shaoyang, Hunan 422000, P.R. China
| | - Ping Ai
- Department of General Surgery, Second People's Hospital of Shaoyang City, Shaoyang, Hunan 422000, P.R. China
| | - Shuguang He
- Clinical Laboratory, First Affiliated Hospital of Hunan University of Chinese Medicine, Zhuzhou, Hunan 412000, P.R. China
| | - Lingzhi Liu
- Clinical Laboratory, First Affiliated Hospital of Hunan University of Chinese Medicine, Zhuzhou, Hunan 412000, P.R. China
| | - Chaoying Chen
- Clinical Laboratory, First Affiliated Hospital of Hunan University of Chinese Medicine, Zhuzhou, Hunan 412000, P.R. China
| | - Yuansheng Tan
- Hanpu Science and Teaching Park, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Ting Wang
- Hanpu Science and Teaching Park, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| |
Collapse
|
30
|
Islam F, Chaousis S, Wahab R, Gopalan V, Lam AK. Protein interactions of FAM134B with EB1 and APC/beta‐catenin in vitro in colon carcinoma. Mol Carcinog 2018; 57:1480-1491. [DOI: 10.1002/mc.22871] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 06/25/2018] [Accepted: 06/28/2018] [Indexed: 12/13/2022]
Abstract
FAM134B is an autophagy regulator of endoplasmic reticulum and acts as a cancer suppressor in colon cancer. However, the molecular signaling pathways by which FAM134B interacts within colon carcinogenesis is still unknown. Herein, this study aims to determine the interacting partners of FAM134B for the first time in colon cancer and to explore the precise location of FAM134B in cancer signalling pathways. Liquid chromatography coupled with tandem mass spectrometry (LC‐MS/MS) followed by anti‐FAM134B co‐immune precipitation of FAM134B interacting complex was used to identify the potential interactors of FAM134B in colon cancer cells. Western blot and confocal microscopic analysis were used to validate the physical interactions of FAM134B with the interactors. Lentiviral shRNA mediated silencing of FAM134B was used to examine the modulation of FAM134B interactors in cells. We have identified 29 novel binding partners, including CAP1, RPS28, FTH1, KDELR2, MAP4, EB1, PSMD6, PPIB/CYPB etc. Subsequent immunoassays confirmed the direct physical interactions of FAM134B with CAP1, EB1, CYPB, and KDELR2 in colon cancer cells. Exogenous suppression of FAM134B has led to significant upregulation of EB1 as well as reduction of KDELR2 expression. It was noted that overexpression of EB1 promotes WNT/β‐catenin signaling pathways via inactivating tumor suppressor APC followed by activating β‐catenin in colorectal carcinogenesis. This study has first time reported the gene signaling networks with which FAM134B interacts and noted that FAM134B is involved in the regulation of WNT/β‐catenin pathway by EB1‐mediated modulating of APC in colon cancer cells.
Collapse
Affiliation(s)
- Farhadul Islam
- Cancer Molecular Pathology School of Medicine Menzies Health Institute Queensland Griffith University Gold Coast Queensland Australia
- Department of Biochemistry and Molecular Biology University of Rajshahi Rajshahi Bangladesh
| | - Stephanie Chaousis
- Australian Rivers Institute and School of Environment Griffith University Gold Coast Queensland Australia
| | - Riajul Wahab
- Cancer Molecular Pathology School of Medicine Menzies Health Institute Queensland Griffith University Gold Coast Queensland Australia
| | - Vinod Gopalan
- Cancer Molecular Pathology School of Medicine Menzies Health Institute Queensland Griffith University Gold Coast Queensland Australia
- School of Medical Science Menzies Health Institute Queensland Griffith University Gold Coast Queensland Australia
| | - Alfred K.‐Y. Lam
- Cancer Molecular Pathology School of Medicine Menzies Health Institute Queensland Griffith University Gold Coast Queensland Australia
| |
Collapse
|
31
|
Islam F, Gopalan V, Pillai S, Lu CT, Kasem K, Lam AKY. Promoter hypermethylation inactivate tumor suppressor FAM134B and is associated with poor prognosis in colorectal cancer. Genes Chromosomes Cancer 2018; 57:240-251. [PMID: 29318692 DOI: 10.1002/gcc.22525] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 01/05/2018] [Accepted: 01/07/2018] [Indexed: 01/04/2023] Open
Abstract
The present study aims to examine promoter methylation status of FAM134B in a large cohort of patients with colorectal adenocarcinomas. The clinical significances and correlations of FAM134B promoter methylation with its expression are also analysed. Methylation-specific high-resolution melt-curve analysis followed by sequencing was used to identify FAM134B promoter methylation in colorectal adenomas (N = 32), colorectal adenocarcinomas (N = 164), matched adjacent non-neoplastic colorectal mucosae (N = 83) and colon cancer cell lines (N = 4). FAM134B expression was studied by real-time quantitative polymerase chain reaction, immunohistochemistry, and Western blots. FAM134B promoter methylation was more frequent in adenocarcinomas (52%; 85/164) when compared to that of adenomas (28%; 9/32) and non-neoplastic mucosae (35%; 29/83). Cancer cells exhibited higher methylation when compared to non-neoplastic cells. FAM134B promoter methylation was inversely correlated with low FAM134B copy number and mRNA/protein expressions, whereas in-vitro demethylation has restored FAM134B expression in colon cancer cells. FAM134B promoter methylation was associated with high histological grade (P = .025), presence of peri-neural infiltration (P = .012), lymphovascular invasion (P = .021), lymph node metastasis (P = .0001), distant metastasis (P = .0001) and advanced pathological stages (P = .0001). In addition, FAM134B promoter methylation correlated with cancer recurrence and poor survival rates of patients with colorectal adenocarcinomas. To conclude, FAM134B promoter methylation plays a key role in regulating FAM134B expression in vitro and in vivo, which in turn contributes to the prediction of the biological aggressiveness of colorectal adenocarcinomas. Furthermore, FAM134B methylation might act as a marker in predicting clinical prognosis in patients with colorectal adenocarcinomas.
Collapse
Affiliation(s)
- Farhadul Islam
- Cancer Molecular Pathology, School of Medicine and Menzies Health Institute Queensland, Griffith University, Queensland, Australia
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Vinod Gopalan
- Cancer Molecular Pathology, School of Medicine and Menzies Health Institute Queensland, Griffith University, Queensland, Australia
| | - Suja Pillai
- Cancer Molecular Pathology, School of Medicine and Menzies Health Institute Queensland, Griffith University, Queensland, Australia
- School of Biomedical Sciences, University of Queensland, Queensland, Australia
| | - Cu-Tai Lu
- Department of Surgery, Gold Coast Hospital, Gold Coast, Queensland, Australia
| | - Kais Kasem
- Cancer Molecular Pathology, School of Medicine and Menzies Health Institute Queensland, Griffith University, Queensland, Australia
| | - Alfred King-Yin Lam
- Cancer Molecular Pathology, School of Medicine and Menzies Health Institute Queensland, Griffith University, Queensland, Australia
| |
Collapse
|
32
|
Islam F, Gopalan V, Lam AKY. RETREG1(FAM134B): A new player in human diseases: 15 years after the discovery in cancer. J Cell Physiol 2018; 233:4479-4489. [DOI: 10.1002/jcp.26384] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 12/04/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Farhadul Islam
- Cancer Molecular Pathology; School of Medicine and Griffith Health Institute; Griffith University; Gold Coast Queensland Australia
- Department of Biochemistry and Molecular Biology; University of Rajshahi; Rajshahi Bangladesh
| | - Vinod Gopalan
- Cancer Molecular Pathology; School of Medicine and Griffith Health Institute; Griffith University; Gold Coast Queensland Australia
| | - Alfred King-yin Lam
- Cancer Molecular Pathology; School of Medicine and Griffith Health Institute; Griffith University; Gold Coast Queensland Australia
| |
Collapse
|
33
|
Islam F, Gopalan V, Vider J, Wahab R, Ebrahimi F, Lu CT, Kasem K, Lam AKY. MicroRNA-186-5p overexpression modulates colon cancer growth by repressing the expression of the FAM134B tumour inhibitor. Exp Cell Res 2017; 357:260-270. [PMID: 28549913 DOI: 10.1016/j.yexcr.2017.05.021] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 05/21/2017] [Accepted: 05/23/2017] [Indexed: 02/07/2023]
Abstract
OBJECTIVES The role and underlying mechanism of miR-186-5p in colorectal cancer remain unknown. The present study aims to examine the various cellular effects of miR-186-5p in the carcinogenesis of colorectal cancer. Also, the interacting targets and association of clinicopathological factors with miR-186-5p expression in patients with colorectal cancer were analysed. METHODS The miR-186-5p expression levels in colorectal cancer tissues (n=126) and colon cancer cell lines (n=3) were analysed by real-time PCR. Matched non-neoplastic colorectal tissues and a non-neoplastic colonic epithelial cell line were used as controls. Various in vitro assays such as cell proliferation, wound healing and colony formation assays were performed to examine the miR-186-5p specific cellular effects. Western blots and immunohistochemistry analysis were performed to examine the modulation of FAM134B, PARP9 and KLF7 proteins expression. RESULTS Significant high expression of miR-186-5p was noted in cancer tissues (p< 0.001) and cell lines (p<0.05) when compared to control tissues and cells. The majority of the patients with colorectal cancer (88/126) had shown overexpression of miR-186-5p. This miR-186-5p overexpression was predominantly noted with in cancer with distant metastasis (p=0.001), lymphovascular permeation (p=0.037), microsatellite instability (MSI) stable (p=0.015), in distal colorectum (p=0.043) and with associated adenomas (p=0.047). Overexpression of miR-186-5p resulted in increased cell proliferation, colony formation, wound healing capacities and induced alteration of cell cycle kinetics in colon cancer cells. On the other hand, inhibition of endogenous miR-186-5p reduced the cancer growth properties. miR-186-5p overexpression reduced FAM134B expression significantly in the cancer cells (p<0.01). Also, FAM134B and miR-186-5p expressions are inversely correlated in colorectal cancer tissues and cells. CONCLUSION The miR-186-5p expression promotes colorectal cancer pathogenesis by regulating tumour suppressor FAM134B. Reduced cancer cells growth followed by inhibition of miR-186-5p highlights the potential of miR-186-5p inhibitor as a novel strategy for targeting colorectal cancer initiation and progression.
Collapse
Affiliation(s)
- Farhadul Islam
- Cancer Molecular Pathology, School of Medicine and Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia; Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Vinod Gopalan
- Cancer Molecular Pathology, School of Medicine and Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - Jelena Vider
- School of Medical Science and Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - Riajul Wahab
- Cancer Molecular Pathology, School of Medicine and Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - Faeza Ebrahimi
- Cancer Molecular Pathology, School of Medicine and Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - Cu-Tai Lu
- Department of Surgery, Gold Coast Hospital, Gold Coast, Queensland, Australia
| | - Kais Kasem
- Cancer Molecular Pathology, School of Medicine and Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - Alfred K Y Lam
- Cancer Molecular Pathology, School of Medicine and Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia.
| |
Collapse
|
34
|
Haque MH, Gopalan V, Islam MN, Masud MK, Bhattacharjee R, Hossain MSA, Nguyen NT, Lam AK, Shiddiky MJA. Quantification of gene-specific DNA methylation in oesophageal cancer via electrochemistry. Anal Chim Acta 2017; 976:84-93. [PMID: 28576321 DOI: 10.1016/j.aca.2017.04.034] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 04/10/2017] [Accepted: 04/10/2017] [Indexed: 02/07/2023]
Abstract
Development of simple and inexpensive method for the analysis of gene-specific DNA methylation is important for the diagnosis and prognosis of patients with cancer. Herein, we report a relatively simple and inexpensive electrochemical method for the sensitive and selective detection of gene-specific DNA methylation in oesophageal cancer. The underlying principle of the method relies on the affinity interaction between DNA bases and unmodified gold electrode. Since the affinity trend of DNA bases towards the gold surface follows as adenine (A) > cytosine (C) > guanine (G)> thymine (T), a relatively larger amount of bisulfite-treated adenine-enriched unmethylated DNA adsorbs on the screen-printed gold electrodes (SPE-Au) in comparison to the guanine-enriched methylated sample. The methylation levels were (i.e., different level of surface attached DNA molecules due to the base dependent differential adsorption pattern) quantified by measuring saturated amount of charge-compensating [Ru(NH3)6]3+ molecules in the surface-attached DNAs by chronocoulometry as redox charge of the [Ru(NH3)6]3+ molecules quantitatively reflects the amount of the adsorbed DNA confined at the electrode surface. The assay could successfully distinguish methylated and unmethylated DNA sequences at single CpG resolution and as low as 10% differences in DNA methylation. In addition, the assay showed fairly good reproducibility (% RSD= <5%) with better sensitivity and specificity by analysing various levels of methylation in two cell lines and eight fresh tissues samples from patients with oesophageal squamous cell carcinoma. Finally, the method was validated with methylation specific-high resolution melting curve analysis and Sanger sequencing methods.
Collapse
Affiliation(s)
- Md Hakimul Haque
- Cancer Molecular Pathology Laboratory in Menzies Health Institute Queensland, Griffith University and School of Medicine, Gold Coast, QLD 4222, Australia; School of Natural Sciences, Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
| | - Vinod Gopalan
- Cancer Molecular Pathology Laboratory in Menzies Health Institute Queensland, Griffith University and School of Medicine, Gold Coast, QLD 4222, Australia.
| | - Md Nazmul Islam
- School of Natural Sciences, Griffith University, Nathan Campus, Nathan, QLD 4111, Australia; Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
| | - Mostafa Kamal Masud
- School of Natural Sciences, Griffith University, Nathan Campus, Nathan, QLD 4111, Australia; Institute for Superconducting and Electronic Materials, Australian Institute for Innovative Materials, University of Wollongong, NSW 2519, Australia
| | - Ripon Bhattacharjee
- School of Natural Sciences, Griffith University, Nathan Campus, Nathan, QLD 4111, Australia; Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
| | - Md Shahriar Al Hossain
- Institute for Superconducting and Electronic Materials, Australian Institute for Innovative Materials, University of Wollongong, NSW 2519, Australia
| | - Nam-Trung Nguyen
- Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
| | - Alfred K Lam
- Cancer Molecular Pathology Laboratory in Menzies Health Institute Queensland, Griffith University and School of Medicine, Gold Coast, QLD 4222, Australia.
| | - Muhammad J A Shiddiky
- School of Natural Sciences, Griffith University, Nathan Campus, Nathan, QLD 4111, Australia; Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia.
| |
Collapse
|
35
|
Islam MN, Gopalan V, Haque MH, Masud MK, Hossain MSA, Yamauchi Y, Nguyen NT, Lam AKY, Shiddiky MJA. A PCR-free electrochemical method for messenger RNA detection in cancer tissue samples. Biosens Bioelectron 2017; 98:227-233. [PMID: 28688308 DOI: 10.1016/j.bios.2017.06.051] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 06/19/2017] [Accepted: 06/25/2017] [Indexed: 11/17/2022]
Abstract
Despite having reliable and excellent diagnostic performances, the currently available messenger RNA (mRNA) detection methods mostly use enzymatic amplification steps of the target mRNA which is generally affected by the sample manipulations, amplification bias and longer assay time. This paper reports an amplification-free electrochemical approach for the sensitive and selective detection of mRNA using a screen-printed gold electrode (SPE-Au). The target mRNA is selectively isolated by magnetic separation and adsorbed directly onto an unmodified SPE-Au. The surface-attached mRNA is then measured by differential pulse voltammetry (DPV) in the presence of [Fe(CN)6]4-/3- redox system. This method circumvents the PCR amplification steps as well as simplifies the assay construction by avoiding multiple steps involved in conventional biosensing approaches of using recognition and transduction layers. Our method has demonstrated good sensitivity (LOD = 1.0pM) and reproducibility (% RSD = <5%, for n = 3) for detecting FAM134B mRNA in two cancer cell lines and a small cohort of clinical samples (number of samples = 26) collected from patients with oesophageal cancer. The analytical performance of our method is validated with a standard qRT-PCR analysis. We believe that our PCR-free approach holds a great promise for the analysis of tumor-specific mRNA in clinical samples.
Collapse
Affiliation(s)
- Md Nazmul Islam
- School of Natural Sciences, Griffith University, Nathan Campus, QLD 4111, Australia; Queensland Micro-, and Nanotechnology Centre, Griffith University, Nathan Campus, QLD 4111, Australia
| | - Vinod Gopalan
- Cancer Molecular Pathology Laboratory in School of Medicine, Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, QLD 4222, Australia
| | - Md Hakimul Haque
- Cancer Molecular Pathology Laboratory in School of Medicine, Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, QLD 4222, Australia
| | - Mostafa Kamal Masud
- Queensland Micro-, and Nanotechnology Centre, Griffith University, Nathan Campus, QLD 4111, Australia; Australian Institute for Innovative Materials (AIIM), University of Wollongong, Squires Way, Innovation Campus, North Wollongong, NSW 2519, Australia
| | - Md Shahriar Al Hossain
- Australian Institute for Innovative Materials (AIIM), University of Wollongong, Squires Way, Innovation Campus, North Wollongong, NSW 2519, Australia
| | - Yusuke Yamauchi
- Australian Institute for Innovative Materials (AIIM), University of Wollongong, Squires Way, Innovation Campus, North Wollongong, NSW 2519, Australia; International Center for Materials Nanoarchitectonics (MANA), National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| | - Nam-Trung Nguyen
- Queensland Micro-, and Nanotechnology Centre, Griffith University, Nathan Campus, QLD 4111, Australia
| | - Alfred King-Yin Lam
- Cancer Molecular Pathology Laboratory in School of Medicine, Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, QLD 4222, Australia
| | - Muhammad J A Shiddiky
- School of Natural Sciences, Griffith University, Nathan Campus, QLD 4111, Australia; Queensland Micro-, and Nanotechnology Centre, Griffith University, Nathan Campus, QLD 4111, Australia.
| |
Collapse
|
36
|
Islam F, Haque MH, Yadav S, Islam MN, Gopalan V, Nguyen NT, Lam AK, Shiddiky MJA. An electrochemical method for sensitive and rapid detection of FAM134B protein in colon cancer samples. Sci Rep 2017; 7:133. [PMID: 28273937 PMCID: PMC5428029 DOI: 10.1038/s41598-017-00206-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 02/14/2017] [Indexed: 12/29/2022] Open
Abstract
Despite the excellent diagnostic applications of the current conventional immunoassay methods such as ELISA, immunostaining and Western blot for FAM134B detection, they are laborious, expensive and required a long turnaround time. Here, we report an electrochemical approach for rapid, sensitive, and specific detection of FAM134B protein in biological (colon cancer cell extracts) and clinical (serum) samples. The approach utilises a differential pulse voltammetry (DPV) in the presence of the [Fe(CN)6]3-/4- redox system to quantify the FAM134B protein in a two-step strategy that involves (i) initial attachment of FAM134B antibody on the surface of extravidin-modified screen-printed carbon electrode, and (ii) subsequent detection of FAM134B protein present in the biological/clinical samples. The assay system was able to detect FAM134B protein at a concentration down to 10 pg μL-1 in phosphate buffered saline (pH 7.4) with a good inter-assay reproducibility (% RSD = <8.64, n = 3). We found excellent sensitivity and specificity for the analysis of FAM134B protein in a panel of colon cancer cell lines and serum samples. Finally, the assay was further validated with ELISA method. We believe that our assay could potentially lead a low-cost alternative to conventional immunological assays for target antigens analysis in point-of-care applications.
Collapse
Affiliation(s)
- Farhadul Islam
- Cancer Molecular Pathology Laboratory in School of Medicine, Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Australia
| | - Md Hakimul Haque
- Cancer Molecular Pathology Laboratory in School of Medicine, Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Australia
- School of Natural Sciences, Griffith University, Nathan Campus, QLD 4111, Australia
| | - Sharda Yadav
- School of Natural Sciences, Griffith University, Nathan Campus, QLD 4111, Australia
- Queensland Micro and Nanotechnology Centre, Griffith University, Nathan Campus, QLD 4111, Australia
| | - Md Nazmul Islam
- School of Natural Sciences, Griffith University, Nathan Campus, QLD 4111, Australia
- Queensland Micro and Nanotechnology Centre, Griffith University, Nathan Campus, QLD 4111, Australia
| | - Vinod Gopalan
- Cancer Molecular Pathology Laboratory in School of Medicine, Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Australia.
| | - Nam-Trung Nguyen
- Queensland Micro and Nanotechnology Centre, Griffith University, Nathan Campus, QLD 4111, Australia
| | - Alfred K Lam
- Cancer Molecular Pathology Laboratory in School of Medicine, Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Australia.
| | - Muhammad J A Shiddiky
- School of Natural Sciences, Griffith University, Nathan Campus, QLD 4111, Australia.
- Queensland Micro and Nanotechnology Centre, Griffith University, Nathan Campus, QLD 4111, Australia.
| |
Collapse
|
37
|
Islam F, Gopalan V, Wahab R, Lee KTW, Haque MH, Mamoori A, Lu CT, Smith RA, Lam AKY. Novel FAM134B mutations and their clinicopathological significance in colorectal cancer. Hum Genet 2017; 136:321-337. [DOI: 10.1007/s00439-017-1760-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 01/21/2017] [Indexed: 12/13/2022]
|
38
|
Haque MH, Islam MN, Islam F, Gopalan V, Nguyen NT, Lam AK, Shiddiky MJA. Electrochemical Detection of FAM134B Mutations in Oesophageal Cancer Based on DNA-Gold Affinity Interactions. ELECTROANAL 2017. [DOI: 10.1002/elan.201700039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Md. Hakimul Haque
- Cancer Molecular Pathology Laboratory in School of Medicine, Menzies Health Institute Queensland; Griffith University; Gold Coast Campus Australia
- School of Natural Sciences; Griffith University; Nathan Campus QLD 4111 Australia
- Queensland Micro and Nanotechnology Centre; Griffith University; Nathan Campus QLD 4111 Australia
| | - Md. Nazmul Islam
- School of Natural Sciences; Griffith University; Nathan Campus QLD 4111 Australia
- Queensland Micro and Nanotechnology Centre; Griffith University; Nathan Campus QLD 4111 Australia
| | - Farhadul Islam
- Cancer Molecular Pathology Laboratory in School of Medicine, Menzies Health Institute Queensland; Griffith University; Gold Coast Campus Australia
| | - Vinod Gopalan
- Cancer Molecular Pathology Laboratory in School of Medicine, Menzies Health Institute Queensland; Griffith University; Gold Coast Campus Australia
| | - Nam-Trung Nguyen
- Queensland Micro and Nanotechnology Centre; Griffith University; Nathan Campus QLD 4111 Australia
| | - Alfred K. Lam
- Cancer Molecular Pathology Laboratory in School of Medicine, Menzies Health Institute Queensland; Griffith University; Gold Coast Campus Australia
| | - Muhammad J. A. Shiddiky
- School of Natural Sciences; Griffith University; Nathan Campus QLD 4111 Australia
- Queensland Micro and Nanotechnology Centre; Griffith University; Nathan Campus QLD 4111 Australia
| |
Collapse
|
39
|
Haque MH, Gopalan V, Yadav S, Islam MN, Eftekhari E, Li Q, Carrascosa LG, Nguyen NT, Lam AK, Shiddiky MJA. Detection of regional DNA methylation using DNA-graphene affinity interactions. Biosens Bioelectron 2017; 87:615-621. [PMID: 27616287 DOI: 10.1016/j.bios.2016.09.016] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 08/29/2016] [Accepted: 09/04/2016] [Indexed: 12/26/2022]
Abstract
We report a new method for the detection of regional DNA methylation using base-dependent affinity interaction (i.e., adsorption) of DNA with graphene. Due to the strongest adsorption affinity of guanine bases towards graphene, bisulfite-treated guanine-enriched methylated DNA leads to a larger amount of the adsorbed DNA on the graphene-modified electrodes in comparison to the adenine-enriched unmethylated DNA. The level of the methylation is quantified by monitoring the differential pulse voltammetric current as a function of the adsorbed DNA. The assay is sensitive to distinguish methylated and unmethylated DNA sequences at single CpG resolution by differentiating changes in DNA methylation as low as 5%. Furthermore, this method has been used to detect methylation levels in a collection of DNA samples taken from oesophageal cancer tissues.
Collapse
Affiliation(s)
- Md Hakimul Haque
- Cancer Molecular Pathology Laboratory in School of Medicine, Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Australia; School of Natural Sciences, Griffith University, Nathan Campus, QLD 4111, Australia
| | - Vinod Gopalan
- Cancer Molecular Pathology Laboratory in School of Medicine, Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Australia.
| | - Sharda Yadav
- School of Natural Sciences, Griffith University, Nathan Campus, QLD 4111, Australia; Queensland Micro and Nanotechnology Centre, Griffith University, Nathan Campus, QLD 4111, Australia
| | - Md Nazmul Islam
- School of Natural Sciences, Griffith University, Nathan Campus, QLD 4111, Australia; Queensland Micro and Nanotechnology Centre, Griffith University, Nathan Campus, QLD 4111, Australia
| | - Ehsan Eftekhari
- Queensland Micro and Nanotechnology Centre, Griffith University, Nathan Campus, QLD 4111, Australia; School of Engineering, Griffith University, Nathan, QLD 4111, Australia
| | - Qin Li
- Queensland Micro and Nanotechnology Centre, Griffith University, Nathan Campus, QLD 4111, Australia; School of Engineering, Griffith University, Nathan, QLD 4111, Australia
| | | | - Nam-Trung Nguyen
- Queensland Micro and Nanotechnology Centre, Griffith University, Nathan Campus, QLD 4111, Australia
| | - Alfred K Lam
- Cancer Molecular Pathology Laboratory in School of Medicine, Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Australia.
| | - Muhammad J A Shiddiky
- School of Natural Sciences, Griffith University, Nathan Campus, QLD 4111, Australia; Queensland Micro and Nanotechnology Centre, Griffith University, Nathan Campus, QLD 4111, Australia.
| |
Collapse
|
40
|
Islam F, Gopalan V, Wahab R, Smith RA, Qiao B, Lam AKY. Stage dependent expression and tumor suppressive function of FAM134B( JK1) in colon cancer. Mol Carcinog 2017; 56:238-249. [DOI: 10.1002/mc.22488] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Farhadul Islam
- Cancer Molecular Pathology; School of Medicine and Griffith Health Institute; Griffith University; Gold Coast Queensland Australia
| | - Vinod Gopalan
- Cancer Molecular Pathology; School of Medicine and Griffith Health Institute; Griffith University; Gold Coast Queensland Australia
| | - Riajul Wahab
- Cancer Molecular Pathology; School of Medicine and Griffith Health Institute; Griffith University; Gold Coast Queensland Australia
| | - Robert A. Smith
- Cancer Molecular Pathology; School of Medicine and Griffith Health Institute; Griffith University; Gold Coast Queensland Australia
| | - Bin Qiao
- Department of Stomatology; The First Affiliated Hospital of Zhengzhou University; Zhengzhou Henan Province China
| | - Alfred King-Yin Lam
- Cancer Molecular Pathology; School of Medicine and Griffith Health Institute; Griffith University; Gold Coast Queensland Australia
- Department of Stomatology; The First Affiliated Hospital of Zhengzhou University; Zhengzhou Henan Province China
| |
Collapse
|
41
|
Haque MH, Gopalan V, Chan KW, Shiddiky MJA, Smith RA, Lam AKY. Identification of Novel FAM134B (JK1) Mutations in Oesophageal Squamous Cell Carcinoma. Sci Rep 2016; 6:29173. [PMID: 27373372 PMCID: PMC4931577 DOI: 10.1038/srep29173] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 06/09/2016] [Indexed: 01/02/2023] Open
Abstract
Mutation of FAM134B (Family with Sequence Similarity 134, Member B) leading to loss of function of its encoded Golgi protein and has been reported induce apoptosis in neurological disorders. FAM134B mutation is still unexplored in cancer. Herein, we studied the DNA copy number variation and novel mutation sites of FAM134B in a large cohort of freshly collected oesophageal squamous cell carcinoma (ESCC) tissue samples. In ESCC tissues, 37% (38/102) showed increased FAM134B DNA copies whereas 35% (36/102) showed loss of FAM134B copies relative to matched non-cancer tissues. Novel mutations were detected in exons 4, 5, 7, 9 as well as introns 2, 4-8 of FAM134B via HRM (High-Resolution Melt) and Sanger sequencing analysis. Overall, thirty-seven FAM134B mutations were noted in which most (31/37) mutations were homozygous. FAM134B mutations were detected in all the cases with metastatic ESCC in the lymph node tested and in 14% (8/57) of the primary ESCC. Genetic alteration of FAM134B is a frequent event in the progression of ESCCs. These findings imply that mutation might be the major driving source of FAM134B genetic modulation in ESCCs.
Collapse
Affiliation(s)
- Md Hakimul Haque
- Cancer Molecular Pathology in Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia
| | - Vinod Gopalan
- Cancer Molecular Pathology in Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia
| | - Kwok-Wah Chan
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | | | - Robert Anthony Smith
- Cancer Molecular Pathology in Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia.,Genomics Research Centre, Institute of Health and Biomedical Innovation, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Alfred King-Yin Lam
- Cancer Molecular Pathology in Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia
| |
Collapse
|