1
|
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
|
2
|
Aktar S, Islam F, Cheng T, Gamage SMK, Choudhury IN, Islam MS, Lu CT, Hamid FB, Ishida H, Abe I, Xie N, Gopalan V, Lam AK. Correlation between KRAS Mutation and CTLA-4 mRNA Expression in Circulating Tumour Cells: Clinical Implications in Colorectal Cancer. Genes (Basel) 2023; 14:1808. [PMID: 37761948 PMCID: PMC10530465 DOI: 10.3390/genes14091808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/07/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Combination strategies of KRAS inhibition with immunotherapy in treating advanced or recurrent colorectal carcinoma (CRC) may need to be assessed in circulating tumour cells (CTCs) to achieve better clinical outcomes. This study aimed to investigate the genomic variations of KRAS in CTCs and matched CRC tissues and compared mRNA expression of KRAS and CTLA-4 between wild-type and KRAS-mutated CTCs and CRC tissues. Clinicopathological correlations were also compared. Six known mutations of KRAS were identified at both codon 12 and codon 13 (c.35G>T/G12V, c.35G>A7/G12D, c.35G>C/G12A, c.34G>A/G12S, c.38G>C/G13A, and c.38G>A/G13D). Three CTC samples harboured the identified mutations (16.7%; 3/18), while fifteen matched primary tumour tissues (65.2%, 15/23) showed the mutations. CTCs harbouring the KRAS variant were different from matched CRC tissue. All the mutations were heterozygous. Though insignificant, CTLA-4 mRNA expression was higher in patients carrying KRAS mutations. Patients harbouring KRAS mutations in CTCs were more likely to have poorly differentiated tumours (p = 0.039) and with lymph node metastasis (p = 0.027) and perineural invasion (p = 0.014). KRAS mutations in CTCs were also significantly correlated with overall pathological stages (p = 0.027). These findings imply the genetic basis of KRAS with immunotherapeutic target molecules based on a real-time platform. This study also suggests the highly heterogeneous nature of cancer cells, which may facilitate the assessment of clonal dynamics across a single patient's disease.
Collapse
Affiliation(s)
- Sharmin Aktar
- Cancer Molecular Pathology, School of Medicine and Dentistry, Griffith University, Gold Coast, QLD 4222, Australia; (S.A.); (T.C.); (S.M.K.G.); (M.S.I.); (F.B.H.); (H.I.); (I.A.); (N.X.)
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD 4222, Australia;
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| | - Farhadul Islam
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi 6205, Bangladesh;
| | - Tracie Cheng
- Cancer Molecular Pathology, School of Medicine and Dentistry, Griffith University, Gold Coast, QLD 4222, Australia; (S.A.); (T.C.); (S.M.K.G.); (M.S.I.); (F.B.H.); (H.I.); (I.A.); (N.X.)
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD 4222, Australia;
| | - Sujani Madhurika Kodagoda Gamage
- Cancer Molecular Pathology, School of Medicine and Dentistry, Griffith University, Gold Coast, QLD 4222, Australia; (S.A.); (T.C.); (S.M.K.G.); (M.S.I.); (F.B.H.); (H.I.); (I.A.); (N.X.)
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD 4222, Australia;
- Faculty of Health Sciences & Medicine, Bond University, Gold Coast, QLD 4229, Australia
| | - Indra Neil Choudhury
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD 4222, Australia;
| | - Md Sajedul Islam
- Cancer Molecular Pathology, School of Medicine and Dentistry, Griffith University, Gold Coast, QLD 4222, Australia; (S.A.); (T.C.); (S.M.K.G.); (M.S.I.); (F.B.H.); (H.I.); (I.A.); (N.X.)
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD 4222, Australia;
- Department of Biochemistry & Biotechnology, University of Barishal, Barishal 8254, Bangladesh
| | - Cu Tai Lu
- Department of Surgery, Gold Coast University Hospital, Gold Coast, QLD 4215, Australia;
| | - Faysal Bin Hamid
- Cancer Molecular Pathology, School of Medicine and Dentistry, Griffith University, Gold Coast, QLD 4222, Australia; (S.A.); (T.C.); (S.M.K.G.); (M.S.I.); (F.B.H.); (H.I.); (I.A.); (N.X.)
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD 4222, Australia;
| | - Hirotaka Ishida
- Cancer Molecular Pathology, School of Medicine and Dentistry, Griffith University, Gold Coast, QLD 4222, Australia; (S.A.); (T.C.); (S.M.K.G.); (M.S.I.); (F.B.H.); (H.I.); (I.A.); (N.X.)
| | - Ichiro Abe
- Cancer Molecular Pathology, School of Medicine and Dentistry, Griffith University, Gold Coast, QLD 4222, Australia; (S.A.); (T.C.); (S.M.K.G.); (M.S.I.); (F.B.H.); (H.I.); (I.A.); (N.X.)
| | - Nan Xie
- Cancer Molecular Pathology, School of Medicine and Dentistry, Griffith University, Gold Coast, QLD 4222, Australia; (S.A.); (T.C.); (S.M.K.G.); (M.S.I.); (F.B.H.); (H.I.); (I.A.); (N.X.)
| | - Vinod Gopalan
- Cancer Molecular Pathology, School of Medicine and Dentistry, Griffith University, Gold Coast, QLD 4222, Australia; (S.A.); (T.C.); (S.M.K.G.); (M.S.I.); (F.B.H.); (H.I.); (I.A.); (N.X.)
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD 4222, Australia;
| | - Alfred K. Lam
- Cancer Molecular Pathology, School of Medicine and Dentistry, Griffith University, Gold Coast, QLD 4222, Australia; (S.A.); (T.C.); (S.M.K.G.); (M.S.I.); (F.B.H.); (H.I.); (I.A.); (N.X.)
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD 4222, Australia;
- Pathology Queensland, Gold Coast University Hospital, Southport, QLD 4215, Australia
| |
Collapse
|
3
|
Xie B, Xie Y, Fang C, Zhong B, Ye R, Zhang J, Liu Q, Li H. Elevated FAM134B expression induces radiation-sensitive in hepatocellular carcinoma. BMC Cancer 2023; 23:671. [PMID: 37460952 PMCID: PMC10353116 DOI: 10.1186/s12885-023-11030-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 05/30/2023] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND Previous studies have shown that Family with sequence similarity 134 member B (FAM134B) was involved in the occurrence and development of malignancy, however, the function and molecular mechanism of FAM134B in Hepatocellular Carcinoma (HCC) radiotherapy resistance remain unclear. Therefore, it may clinical effective to clarify the molecular mechanism and identify novel biomarker to overcome radiotherapy resistance in HCC. METHODS The protein and mRNA expression of FAM134B were determined using Real-time PCR and Western blot, respectively. IHC assay was performed to investigate the association between FAM134B expression and the clinicopathological characteristics of 132 HCC patients. Functional assays, such as in situ model, colon formation, FACS, and Tunel assay were used to determine the oncogenic role of FAM134B in human HCC progression. Furthermore, western blotting and luciferase assay were used to determine the mechanism of FAM134B promotes radiation-sensitive in HCC cells. RESULTS We noted that FAM134B was downregulated in HCC, which was correlated with the radiation resistance in patients with HCC. Overexpression of FAM134B contribute to radiation sensitive in HCC; however, inhibition of FAM134B confers HCC cell lines to radiation resistance both in vitro and in vivo. Moreover, we found that FAM134B interacts with FMS related receptor tyrosine kinase 3 (FLT3) and downregulation of FAM134B activated JAK/Stat3 signaling pathway. Importantly, pharmacological inhibition of JAK/Stat3 signaling pathway significantly counteracted downregulation of FAM134B-induced radiation resistance and enhanced radiation therapeutic efficacy in HCC. CONCLUSIONS Our findings suggest that FAM134B may be a potential therapeutic biomarker for the treatment of HCC patients with radiotherapy tolerance.
Collapse
Affiliation(s)
- Binhui Xie
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Gannan Medical University, 341000, Ganzhou, P R China
| | - Yuankang Xie
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Gannan Medical University, 341000, Ganzhou, P R China
| | - Cuifu Fang
- Department of general surgery III, the First Affiliated Hospital of Gannan Medical University, 341000, Ganzhou, P R China
| | - Baiyin Zhong
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Gannan Medical University, 341000, Ganzhou, P R China
| | - Rong Ye
- Department of general surgery III, the First Affiliated Hospital of Gannan Medical University, 341000, Ganzhou, P R China
| | - Jianhong Zhang
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Gannan Medical University, 341000, Ganzhou, P R China
| | - Qingquan Liu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Gannan Medical University, 341000, Ganzhou, P R China
| | - Heping Li
- Department of Medical Oncology, the First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, P R China.
| |
Collapse
|
4
|
Daraghmi MM, Miller JM, Bailey CG, Doss EM, Kalinski AL, Smaldino PJ, Rubenstein EM. Macro-ER-phagy receptors Atg39p and Atg40p confer resistance to aminoglycoside hygromycin B in S. cerevisiae. MICROPUBLICATION BIOLOGY 2023; 2023:10.17912/micropub.biology.000738. [PMID: 36818312 PMCID: PMC9932795 DOI: 10.17912/micropub.biology.000738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 01/24/2023] [Accepted: 01/26/2023] [Indexed: 02/24/2023]
Abstract
Receptor-mediated autophagic turnover of portions of the endoplasmic reticulum (ER) is mediated by macro-ER-phagy. We hypothesized macro-ER-phagy promotes proteotoxic stress resistance. We predicted Saccharomyces cerevisiae lacking macro-ER-phagy receptors would exhibit enhanced sensitivity to hygromycin B, which reduces translational fidelity and is expected to globally disrupt protein homeostasis, including at the ER. We observed that loss of either of two yeast macro-ER-phagy receptors (Atg39p or Atg40p) compromised cellular resistance to hygromycin B to a similar extent as loss of ER-associated degradation (ERAD) ubiquitin ligases Hrd1p and Doa10p. Our data are consistent with a model whereby macro-ER-phagy and ERAD collaborate to mediate ER protein quality control. Disruptions of macro-ER-phagy have been linked to neuropathy, dementia, and cancer. A dampened capacity to mediate protein quality control may contribute to these conditions.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Eric M. Rubenstein
- Department of Biology, Ball State University
,
Correspondence to: Eric M. Rubenstein (
)
| |
Collapse
|
5
|
Cherubini A, Zito E. ER stress as a trigger of UPR and ER-phagy in cancer growth and spread. Front Oncol 2022; 12:997235. [PMID: 36408145 PMCID: PMC9667062 DOI: 10.3389/fonc.2022.997235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 10/12/2022] [Indexed: 01/25/2023] Open
Abstract
Tumors can survive environmental and metabolic stress by triggering homeostatic responses that re-establish the pre-stress status and permit them to grow and thrive. The endoplasmic reticulum (ER) is the organelle where proteins undergo post-translational modifications and are folded and exported to the secretory pathway. Its environment and activity are therefore fundamental for proteostasis, i.e., the plethora of mechanisms controlling protein formation, folding, degradation, and secretion, needed to assure protein balance and cellular health. In different tumor-related conditions, such as after the activation of oncogenes or under hypoxia and nutrient deprivation, the ER experiences stress, triggered by a high load of proteins to be folded compared to the limited folding capacity of the organelle. As a consequence, three ER membrane sensors and the related unfolded protein response (UPR) are activated. The UPR comprises a complex interconnection between signal transduction pathways that promote a homeostatic response that acts by increasing the amount of protein chaperones and of proteins involved in ER-associated protein degradation (ERAD) on one hand and attenuating protein translation on the other. ER-phagy, literally "eating" the ER, is part of another homeostatic response consisting of the clearance of non-functional ER portions including misfolded proteins. This response is also activated by a set of dedicated ER-phagy receptors after ER stimuli, which overlap the stimuli generating ER stress. Thus, the UPR and ER-phagy are two closely related homeostatic mechanisms that cooperate in re-establishing ER homeostasis. However, while the role of the UPR in favoring cancer growth and thriving by promoting angiogenesis, metastasis, chemotherapy resistance, and epithelial-to-mesenchymal transition is consolidated, that of ER-phagy is still in its infancy. This essay provides an overview of emerging concepts on ER stress, the UPR, and ER-phagy and their crosstalk in tumorigenesis. We also critically review new findings on their pharmacological targeting in cancer.
Collapse
Affiliation(s)
- Alessandro Cherubini
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy,Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Ester Zito
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy,Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy,*Correspondence: Ester Zito,
| |
Collapse
|
6
|
Ravichandran R, PriyaDharshini LC, Sakthivel KM, Rasmi RR. Role and regulation of autophagy in cancer. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166400. [PMID: 35341960 DOI: 10.1016/j.bbadis.2022.166400] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 02/07/2023]
Abstract
Autophagy is an intracellular self-degradative mechanism which responds to cellular conditions like stress or starvation and plays a key role in regulating cell metabolism, energy homeostasis, starvation adaptation, development and cell death. Numerous studies have stipulated the participation of autophagy in cancer, but the role of autophagy either as tumor suppressor or tumor promoter is not clearly understood. However, mechanisms by which autophagy promotes cancer involves a diverse range of modifications of autophagy associated proteins such as ATGs, Beclin-1, mTOR, p53, KRAS etc. and autophagy pathways like mTOR, PI3K, MAPK, EGFR, HIF and NFκB. Furthermore, several researches have highlighted a context-dependent, cell type and stage-dependent regulation of autophagy in cancer. Alongside this, the interaction between tumor cells and their microenvironment including hypoxia has a great potential in modulating autophagy response in favour to substantiate cancer cell metabolism, self-proliferation and metastasis. In this review article, we highlight the mechanism of autophagy and their contribution to cancer cell proliferation and development. In addition, we discuss about tumor microenvironment interaction and their consequence on selective autophagy pathways and the involvement of autophagy in various tumor types and their therapeutic interventions concentrated on exploiting autophagy as a potential target to improve cancer therapy.
Collapse
Affiliation(s)
- Rakesh Ravichandran
- Department of Biotechnology, PSG College of Arts and Science, Civil Aerodrome Post, Coimbatore 641 014, Tamil Nadu, India
| | | | - Kunnathur Murugesan Sakthivel
- Department of Biochemistry, PSG College of Arts and Science, Civil Aerodrome Post, Coimbatore 641 014, Tamil Nadu, India
| | - Rajan Radha Rasmi
- Department of Biotechnology, PSG College of Arts and Science, Civil Aerodrome Post, Coimbatore 641 014, Tamil Nadu, India.
| |
Collapse
|
7
|
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: 66] [Impact Index Per Article: 33.0] [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
|
8
|
HFE variants in colorectal cancer and their clinicopathological correlations. Hum Pathol 2021; 117:9-30. [PMID: 34371060 DOI: 10.1016/j.humpath.2021.07.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 07/30/2021] [Accepted: 07/30/2021] [Indexed: 01/29/2023]
Abstract
The study aimed to screen mutation of human homeostatic iron regulator (HFE) in colorectal carcinoma (CRC) and detect their associations with clinicopathological parameters. Expression of HFE was determined by quantitative polymerase chain reaction in matched CRC and non neoplastic colorectal mucosal tissue of 76 patients. Genomic DNA extracted were subjected to high high-resolution melt curve analysis and Sanger sequencing to detect mutations in HFE. The associations of the identified mutations with a variety of clinical features were determined. Approximately 60% of CRC showed low HFE expression. Of the ten 10 mutations identified in exons 2 and 4, c.187C>G (H63D), c845G>A (C282Y), c.193A>T (S65C), g.3828T>C, g.5795T>C, and g.5728G>A were known mutations. Four novel mutations were discovered; : c.184G>A, c.220T>G, c.322A>C, and c.324T>C. Heterozygous H63D and C282Y mutations were seen in 71% and 49% of cancer tissue, respectively. Tumour site (p = 0.048) and gender (p = 0.039) were significantly associated with H63D and C282Y mutation status, respectively. Local spread of cancer was significantly associated with C282Y mutations in CRC cancer and adjacent non-neoplastic tissue (p = 0.029 & and p = 0.004, respectively). There was a statistically significant association between H63D and C282Y negativity in matched non-neoplastic colorectal mucosa tissue and pathological staging of cancer (p = 0.047 & and p = 0.001, respectively). Patients with H63D and C282Y mutations in cancer tissue tend to have higher survival rates. Hence HFE mutations are common in CRC and are associated with clinicopathological parameters, implying the potential clinical significance of HFE mutations in colorectal carcinogenesis.
Collapse
|
9
|
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: 2.7] [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
|
10
|
Roles of Pathological Assessments of Frozen Sections in Esophageal Squamous Cell Carcinoma. Methods Mol Biol 2021. [PMID: 32056171 DOI: 10.1007/978-1-0716-0377-2_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Pathological assessment of frozen sections of tissues is important in the clinical management (intraoperative consultation) and research in patients with esophageal squamous cell carcinoma. Frozen sections may be used in the assessment of status of resection margins, extent of cancer metastasis (pathological staging), confirmation of the pathology, and increased volume of cancer cells for tissue banking. However, the applications of frozen sections have many technical limitations. Thus, interpretation of frozen sections needs expertise, collaborations, and attention to proper technical skills in the sectioning.
Collapse
|
11
|
Perrotta I. ER-phagy in human atherosclerosis: an exploratory ultrastructural study. Ultrastruct Pathol 2020; 44:489-495. [PMID: 33118423 DOI: 10.1080/01913123.2020.1840468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Autophagy is a vacuolar self-digesting mechanism responsible for the removal of damaged organelles, indigestible aggregates, and nonfunctional long-lived proteins by lysosome. Autophagy is dynamically connected to the endoplasmic reticulum (ER) in several ways. It is capable to counteract the possible harmful effects linked to the impairment of protein folding in the ER; the ER has been proposed as the source for autophagosomal membranes. Also, the ER itself can undergo a selective form of autophagy (called ER-phagy) which ensures the maintenance of ER's morphology and function. Autophagy has been widely investigated in the cardiovascular system however there is no evidence to date regarding the occurrence of ER-phagy into the blood vessel wall. This study has been undertaken to explore the existence of this selective control mechanism in the cells of human atherosclerotic plaques. Transmission Electron Microscopy (TEM) analysis revealed that in the plaque cells the smooth ER profiles reorganized into concentric whorls and closely packed membranes arranged in curved and parallel arrays. Circular, often ring-shaped, ER membranes studded with ribosomes and enclosed in a sequestering vesicle have been also frequently observed. This preliminary study demonstrates the existence of a distinct machinery for the specific turnover of ER membranes in human atherosclerosis and provides the first ultrastructural description of ER-phagy in the diseased vascular tissue. These results may open new perspectives for future investigation in the cardiovascular field.
Collapse
Affiliation(s)
- Ida Perrotta
- Centre for Microscopy and Microanalysis, Transmission Electron Microscopy Laboratory - Department of Biology, Ecology and Earth Sciences (Di.B.E.S.T.), University of Calabria , Cosenza, Italy
| |
Collapse
|
12
|
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: 29] [Impact Index Per Article: 7.3] [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
|
13
|
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 DOI: 10.1080/15384047.2020.1810535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [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 Kin-Yan Lam
- Cancer Molecular Pathology, School of Medicine, Griffith University , Gold Coast, Australia
| |
Collapse
|
14
|
Ishaq M, Ojha R, Sharma AP, Singh SK. Autophagy in cancer: Recent advances and future directions. Semin Cancer Biol 2020; 66:171-181. [PMID: 32201367 DOI: 10.1016/j.semcancer.2020.03.010] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 02/10/2020] [Accepted: 03/16/2020] [Indexed: 02/06/2023]
Abstract
Autophagy is being explored as a potential therapeutic target for enhancing the cytotoxic effects of chemotherapeutic regimens in various malignancies. Autophagy plays a very important role in cancer pathogenesis. Here, we discuss the updates on the modulation of autophagy via dynamic interactions with different organelles and the exploitation of selective autophagy for exploring therapeutic strategies. We further discuss the role of autophagy inhibitors in cancer preclinical and clinical trials, novel autophagy inhibitors, and challenges likely to be faced by clinicians while inducting autophagy modulators in clinical practice.
Collapse
Affiliation(s)
- Mohd Ishaq
- School of Medicine, Department of Pathology, Stanford University, CA, USA.
| | - Rani Ojha
- School of Medicine, Department of Pathology, Stanford University, CA, USA.
| | - Aditya P Sharma
- Department of Urology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| | - Shrawan K Singh
- Department of Urology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| |
Collapse
|
15
|
Abstract
Macroscopic examination of the surgical specimen of esophageal squamous cell carcinoma by pathologist is important for quality clinical management, research, as well as education purposes. The process includes dissection of the specimen, identification of the lesion, measurements, and taking appropriate samples for histopathological examination. The basic principle of the examination is to study the characteristics and extent of the cancer. In addition, examination of proximal resection margin and circumferential resection margin are important in the cancer. A standardized approach for macroscopic examination by professionals is needed for accurate diagnosis and to optimize the use of the surgical specimen with esophageal squamous cell carcinoma.
Collapse
|
16
|
Zhang J, Sun M, Hao M, Diao K, Wang J, Li S, Cao Q, Mi X. FAM53A Affects Breast Cancer Cell Proliferation, Migration, and Invasion in a p53-Dependent Manner. Front Oncol 2019; 9:1244. [PMID: 31799197 PMCID: PMC6874147 DOI: 10.3389/fonc.2019.01244] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 10/29/2019] [Indexed: 12/12/2022] Open
Abstract
Family with sequence similarity 53-member A (FAM53A) is an uncharacterized protein with a suspected but unclear role in tumorigenesis. In this study, we examined its role in breast cancer. Immunohistochemical staining of specimens from 199 cases of breast cancer demonstrated that FAM53A levels were negatively correlated with p53 status. In the p53 wild-type breast cancer cell line MCF-7, FAM53A overexpression inhibited cell migration, invasion, and proliferation, downregulated the expression of Snail, cyclin D1, RhoA, RhoC, and MMP9, and decreased mitogen-activated protein kinase kinase (MEK) and extracellular-signal regulated kinase (ERK) phosphorylation. Concurrently, it upregulated E-cadherin and p21 expression levels. Interestingly, opposite trends were observed in the p53-null breast cancer cell line MDA-MB-231. The MEK inhibitor PD98059 reduced the biological effects of FAM53A knockdown in MCF-7 cells and FAM53A overexpression in MDA-MB-231 cells, suggesting that FAM53A affects breast cancer through the MEK-ERK pathway. Silencing TP53 in MCF-7 cells and stably expressing wild-type p53 in MDA-MB-231 cells confirmed that the effects of FAM53A signaling through the MEK/ERK pathway depended on the p53 status of the cells. These results suggest that FAM53A acts as a tumor suppressor in p53-positive breast cancer by modulating the MEK-ERK pathway, but may be a potential candidate for targeted anticancer therapies in p53-negative breast cancer.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Pathology, College of Basic Medical Sciences, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Mingfang Sun
- Department of Pathology, College of Basic Medical Sciences, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Miaomiao Hao
- Department of Pathology, College of Basic Medical Sciences, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Kexin Diao
- Department of Pathology, College of Basic Medical Sciences, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Jian Wang
- Department of Pathology, College of Basic Medical Sciences, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Shiping Li
- Department of Pathology, College of Basic Medical Sciences, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Qixue Cao
- Department of Pathology, College of Basic Medical Sciences, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Xiaoyi Mi
- Department of Pathology, College of Basic Medical Sciences, First Affiliated Hospital, China Medical University, Shenyang, China
| |
Collapse
|
17
|
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: 72] [Impact Index Per Article: 14.4] [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
|
18
|
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: 3.6] [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
|
19
|
Amengual-Batle P, Rusbridge C, José-López R, Golini L, Shelton GD, Mellersh CS, Gutierrez-Quintana R. Two mixed breed dogs with sensory neuropathy are homozygous for an inversion disrupting FAM134B previously identified in Border Collies. J Vet Intern Med 2018; 32:2082-2087. [PMID: 30307654 PMCID: PMC6272042 DOI: 10.1111/jvim.15312] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 05/19/2018] [Accepted: 07/31/2018] [Indexed: 01/21/2023] Open
Abstract
Two unrelated 8‐month‐old male mixed breed dogs were presented for evaluation of progressive ataxia, knuckling, and lack of pain perception in the distal limbs. Because of the similarity in age of onset, progression, and clinical findings with previously described sensory neuropathy in Border Collies, the affected dogs were screened for an FAM134B mutation and were determined to be homozygous for the mutation. Despite few phenotypic similarities with other breeds, genetic testing for specific diseases should be considered in mixed breed dogs with compatible clinical signs, especially if ancestry is unknown.
Collapse
Affiliation(s)
- Pablo Amengual-Batle
- Small Animal Hospital, School of Veterinary Medicine, University of Glasgow, Glasgow, United Kingdom
| | - Clare Rusbridge
- Fitzpatrick Referrals, Eashing, Surrey, United Kingdom.,Faculty of Health & Medical Sciences, University of Surrey, School of Veterinary Medicine, Vet School Main Building, Daphne Jackson Road, Guildford, GU2 7AL, United Kingdom
| | - Roberto José-López
- Small Animal Hospital, School of Veterinary Medicine, University of Glasgow, Glasgow, United Kingdom
| | - Lorenzo Golini
- Northwest Veterinary Specialists, Cheshire, United Kingdom
| | - G Diane Shelton
- Department of Pathology, School of Medicine, University of California, San Diego, San Diego, California
| | | | | |
Collapse
|
20
|
Daskalaki I, Gkikas I, Tavernarakis N. Hypoxia and Selective Autophagy in Cancer Development and Therapy. Front Cell Dev Biol 2018; 6:104. [PMID: 30250843 PMCID: PMC6139351 DOI: 10.3389/fcell.2018.00104] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 08/13/2018] [Indexed: 01/07/2023] Open
Abstract
Low oxygen availability, a condition known as hypoxia, is a common feature of various pathologies including stroke, ischemic heart disease, and cancer. Hypoxia adaptation requires coordination of intricate pathways and mechanisms such as hypoxia-inducible factors (HIFs), the unfolded protein response (UPR), mTOR, and autophagy. Recently, great effort has been invested toward elucidating the interplay between hypoxia-induced autophagy and cancer cell metabolism. Although novel types of selective autophagy have been identified, including mitophagy, pexophagy, lipophagy, ERphagy and nucleophagy among others, their potential interface with hypoxia response mechanisms remains poorly understood. Autophagy activation facilitates the removal of damaged cellular compartments and recycles components, thus promoting cell survival. Importantly, tumor cells rely on autophagy to support self-proliferation and metastasis; characteristics related to poor disease prognosis. Therefore, a deeper understanding of the molecular crosstalk between hypoxia response mechanisms and autophagy could provide important insights with relevance to cancer and hypoxia-related pathologies. Here, we survey recent findings implicating selective autophagy in hypoxic responses, and discuss emerging links between these pathways and cancer pathophysiology.
Collapse
Affiliation(s)
- Ioanna Daskalaki
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
- Department of Biology, University of Crete, Heraklion, Greece
| | - Ilias Gkikas
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
- Department of Biology, University of Crete, Heraklion, Greece
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
- Department of Basic Sciences, Medical School, University of Crete, Heraklion, Greece
| |
Collapse
|
21
|
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: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 06/25/2018] [Accepted: 06/28/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Farhadul Islam
- Cancer Molecular PathologySchool of Medicine Menzies Health Institute QueenslandGriffith UniversityGold CoastQueenslandAustralia
- Department of Biochemistry and Molecular BiologyUniversity of RajshahiRajshahiBangladesh
| | - Stephanie Chaousis
- Australian Rivers Institute and School of EnvironmentGriffith UniversityGold CoastQueenslandAustralia
| | - Riajul Wahab
- Cancer Molecular PathologySchool of Medicine Menzies Health Institute QueenslandGriffith UniversityGold CoastQueenslandAustralia
| | - Vinod Gopalan
- Cancer Molecular PathologySchool of Medicine Menzies Health Institute QueenslandGriffith UniversityGold CoastQueenslandAustralia
- School of Medical ScienceMenzies Health Institute QueenslandGriffith UniversityGold CoastQueenslandAustralia
| | - Alfred K.‐Y. Lam
- Cancer Molecular PathologySchool of Medicine Menzies Health Institute QueenslandGriffith UniversityGold CoastQueenslandAustralia
| |
Collapse
|
22
|
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: 3.2] [Reference Citation Analysis] [Abstract] [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
|
23
|
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: 6.7] [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
|
24
|
Dai X, Hua T, Hong T. Integrated diagnostic network construction reveals a 4-gene panel and 5 cancer hallmarks driving breast cancer heterogeneity. Sci Rep 2017; 7:6827. [PMID: 28754978 PMCID: PMC5533795 DOI: 10.1038/s41598-017-07189-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 06/23/2017] [Indexed: 12/26/2022] Open
Abstract
Breast cancer encompasses a group of heterogeneous diseases, each associated with distinct clinical implications. Dozens of molecular biomarkers capable of categorizing tumors into clinically relevant subgroups have been proposed which, though considerably contribute in precision medicine, complicate our understandings toward breast cancer subtyping and its clinical translation. To decipher the networking of markers with diagnostic roles on breast carcinomas, we constructed the diagnostic networks by incorporating 6 publically available gene expression datasets with protein interaction data retrieved from BioGRID on previously identified 1015 genes with breast cancer subtyping roles. The Greedy algorithm and mutual information were used to construct the integrated diagnostic network, resulting in 37 genes enclosing 43 interactions. Four genes, FAM134B, KIF2C, ALCAM, KIF1A, were identified having comparable subtyping efficacies with the initial 1015 genes evaluated by hierarchical clustering and cross validations that deploy support vector machine and k nearest neighbor algorithms. Pathway, Gene Ontology, and proliferation marker enrichment analyses collectively suggest 5 primary cancer hallmarks driving breast cancer differentiation, with those contributing to uncontrolled proliferation being the most prominent. Our results propose a 37-gene integrated diagnostic network implicating 5 cancer hallmarks that drives breast cancer heterogeneity and, in particular, a 4-gene panel with clinical diagnostic translation potential.
Collapse
Affiliation(s)
- Xiaofeng Dai
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China.
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China.
| | - Tongyan Hua
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Tingting Hong
- Department of medical oncology, the affiliated hospital of Jiangnan University, the fourth people's hospital of Wuxi, Wuxi, China
| |
Collapse
|
25
|
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: 37] [Impact Index Per Article: 5.3] [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
|
26
|
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: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [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
|
27
|
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: 3.3] [Reference Citation Analysis] [Abstract] [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
|
28
|
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
|
29
|
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: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 01/21/2017] [Indexed: 12/13/2022]
|