1
|
Bustos G, Ahumada-Castro U, Silva-Pavez E, Huerta H, Puebla A, Quezada C, Morgado-Cáceres P, Casanova-Canelo C, Smith-Cortinez N, Podunavac M, Oyarce C, Lladser A, Farias P, Lovy A, Molgó J, Torres VA, Zakarian A, Cárdenas JC. The IP 3R inhibitor desmethylxestospongin B reduces tumor cell migration, invasion and metastasis by impairing lysosome acidification and β1-integrin recycling. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167557. [PMID: 39486657 DOI: 10.1016/j.bbadis.2024.167557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 09/26/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024]
Abstract
Cancer is the second leading cause of death worldwide. >90 % of cancer-related deaths are due to metastasis, a process that depends on the ability of cancer cells to leave the primary tumor, migrate, and colonize different tissues. Inositol 1,4,5-trisphosphate receptor (IP3R)-mediated Ca2+ signaling plays an essential role in maintaining the homeostasis of cancer cells and the sustained proliferation. Desmethylxestospongin B (dmXeB) is a specific inhibitor of the IP3R that selectively arrests cell proliferation and promotes cancer cell death at high concentrations. However, whether migration, invasion and metastasis can be affected by this drug is unknown. Here, by using the highly metastatic triple negative breast cancer (TNBC) cell line MDA-MB-231, we demonstrate that a prolonged inhibition of IP3R-mediated Ca2+ signals with dmXeB significantly reduces cell migration and invasion in vitro and metastasis in vivo. We found that this phenomenon was independent of the bioenergetic control of IP3R over the mitochondria and AMPK activation. Furthermore, employing a tandem LC3-GFP-mcherry assay, we found that prolonged inhibition of IP3R with dmXeB leads to diminished autophagic flux. This reduction can be attributed to impaired lysosomal acidification, as evidenced by assessments using DQ-BSA and pHrodo. Since cell migration requires appropriate assembly and disassembly of focal adhesions, along with the internalization and recycling of integrins via autophagy, we explored the dependency of integrin recycling from autophagosomes, finding that IP3R inhibition with dmXeB impaired the recycling of β1-integrins, which accumulated within autophagosomes. Our findings reveal an unexpected effect of IP3R inhibition with dmXeB in cancer cells that could represent a novel therapeutic strategy for the treatment of cancer metastasis.
Collapse
Affiliation(s)
- Galdo Bustos
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile
| | - Ulises Ahumada-Castro
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile
| | - Eduardo Silva-Pavez
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile
| | - Hernán Huerta
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile
| | - Andrea Puebla
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile
| | - Camila Quezada
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile
| | - Pablo Morgado-Cáceres
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile
| | - César Casanova-Canelo
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile
| | - Natalia Smith-Cortinez
- Department of Gastroenterology and Hepatology, UMCG, University of Groningen, Netherlands
| | - Maša Podunavac
- Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Cesar Oyarce
- Department of Medical Microbiology and Infection Prevention, Tumor Virology and Cancer Immunotherapy, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Alvaro Lladser
- Centro Cientifico y Tecnologico de Excelencia Ciencia & Vida, Fundación Ciencia and Vida, Chile; Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Paula Farias
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile
| | - Alenka Lovy
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile
| | - Jordi Molgó
- Université Paris-Saclay, CEA, Département Médicaments et Technologies pour la Santé, Service d'Ingénierie Moléculaire pour la Santé (SIMoS), Equipe Mixte de Recherche CNRS 9004, F-91191 Gif-sur-Yvette, France
| | - Vicente A Torres
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Chile; Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santiago 8380453, Chile; Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, Santiago, Chile
| | - Armen Zakarian
- Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - J César Cárdenas
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile; Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, CA 93106, USA; The Buck Institute for Research on Aging, Novato, USA.
| |
Collapse
|
2
|
Wang D, Zhang L, Nan J, Wan S, Luo J, Li X, Chen W. High glucose elevates intracellular calcium level and induces ferroptosis in glomerular endothelial cells through the miR-223-3p/ITPR3 pathway. Mol Cell Endocrinol 2024; 594:112384. [PMID: 39426490 DOI: 10.1016/j.mce.2024.112384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 10/10/2024] [Accepted: 10/12/2024] [Indexed: 10/21/2024]
Abstract
We investigated the link between ferroptosis and the miR-223-3p/inositol 1,4,5-trisphosphate receptor type 3 (ITPR3) pathway in diabetic kidney disease (DKD). Blood samples from DKD patients and healthy controls were analysed for iron ions, calcium ions, and lipid peroxidation. High-glucose-induced glomerular endothelial cells were used to simulate DKD. MiR-223-3p overexpression or silencing was achieved using adenoviruses, affecting ferroptosis regulators (glutathione peroxidase 4 [GPX4], cystine/glutamate transporter (xCT), and long-chain acyl-CoA synthetase 4 [ACSL4]) and ITPR3. DKD patients showed elevated levels of iron ions, calcium ions, and lipid peroxidation. High glucose downregulated miR-223-3p, reducing xCT and GPX4 expression and increasing ACSL4 expression. MiR-223-3p was confirmed to target ITPR3 through luciferase reporter assay. MiR-223-3p overexpression reversed high-glucose-induced effects on ferroptosis markers and ITPR3 expression. In summary, high glucose levels decreased miR-223-3p expression, leading to increased calcium ion levels and ferroptosis, potentially through ITPR3 modulation. These findings provide insights into the mechanisms underlying DKD and its potential therapeutic targets.
Collapse
Affiliation(s)
- Dekai Wang
- Department of Endocrinology, The Third Clinical Medical College of China Three Gorges University, Gezhouba Central Hospital of Sinopharm, Yichang, 443002, Hubei, China; Department of General Practice, The First Affiliated Hospital of Kunming Medical University, Kunming, 650031, China
| | - Lihua Zhang
- Department of General Practice, The First Affiliated Hospital of Kunming Medical University, Kunming, 650031, China.
| | - Juanli Nan
- Department of General Practice, The First Affiliated Hospital of Kunming Medical University, Kunming, 650031, China
| | - Shengbi Wan
- Department of General Practice, The First Affiliated Hospital of Kunming Medical University, Kunming, 650031, China
| | - Jingmei Luo
- Department of General Practice, The First Affiliated Hospital of Kunming Medical University, Kunming, 650031, China
| | - Xueqiong Li
- Department of General Practice, The First Affiliated Hospital of Kunming Medical University, Kunming, 650031, China
| | - Wei Chen
- Department of Radiology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650031, China.
| |
Collapse
|
3
|
Pan D, Jiang M, Tao G, Shi J, Song Z, Chen R, Wang D. The role of Ca 2+ signalling and InsP3R in the pathogenesis of intrahepatic cholestasis of pregnancy. J OBSTET GYNAECOL 2024; 44:2345276. [PMID: 38685831 DOI: 10.1080/01443615.2024.2345276] [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: 09/20/2023] [Accepted: 04/14/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND In order to contribute new insights for future prevention and treatment of intrahepatic cholestasis of pregnancy (ICP), and to promote positive pregnancy outcomes, we evaluated serum Ca2+ levels and inositol 1,4,5-trisphosphate receptor (InsP3R) expression in the liver tissue of a rat ICP model. METHODS After establishing the model by injection of oestradiol benzoate and progesterone into pregnant rats, animals were divided into normal control (n = 5) and ICP model groups (n = 5). The expression of InsP3R protein in the liver, and serum levels of Ca2+, glycocholic acid and bile acid were detected. RESULTS InsP3R mRNA and protein were significantly lower in the ICP model group compared to the normal group, as determined by qPCR and immunohistochemistry, respectively. Serum enzyme-linked immunosorbent assay results revealed significantly higher levels of glycocholic acid and bile acid in the ICP model group compared to the normal group, while Ca2+ levels were significantly lower. The levers of Ca2+ were significantly and negatively correlated with the levels of glycocholic acid. The observed decrease in Ca2+ was associated with an increase in total bile acids, but there was no significant correlation. CONCLUSIONS Our results revealed that the expression of InsP3R and serum Ca2+ levels was significantly decreased in the liver tissue of ICP model rats. Additionally, Ca2+ levels were found to be negatively correlated with the level of glycocholic acid.
Collapse
Affiliation(s)
- Dan Pan
- Department of Obstetrics and Gynecology, Taizhou Municipal Hospital affiliated with Taizhou University, Taizhou, China
| | - Mengting Jiang
- Department of Obstetrics and Gynecology, Taizhou Municipal Hospital affiliated with Taizhou University, Taizhou, China
| | - Guoxian Tao
- Department of Obstetrics and Gynecology, Taizhou Municipal Hospital affiliated with Taizhou University, Taizhou, China
| | - Jinmei Shi
- Department of Obstetrics and Gynecology, Taizhou Municipal Hospital affiliated with Taizhou University, Taizhou, China
| | - Zhiwei Song
- Department of Medical Laboratory, Taizhou Municipal Hospital affiliated with Taizhou University, Taizhou, China
| | - Ren Chen
- Department of Obstetrics and Gynecology, Taizhou Municipal Hospital affiliated with Taizhou University, Taizhou, China
| | - Dongguo Wang
- Department of Central Laboratory, Taizhou Municipal Hospital affiliated with Taizhou University, Taizhou, China
| |
Collapse
|
4
|
Cui C, Lu C, Cai Y, Xiong Y, Duan Y, Lan K, Fan Y, Zhou X, Wei X. PTH1R Suppressed Apoptosis of Mesenchymal Progenitors in Mandibular Growth. Int J Mol Sci 2024; 25:12607. [PMID: 39684319 DOI: 10.3390/ijms252312607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/14/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Genetic abnormalities of the parathyroid hormone 1 receptor (PTH1R) lead to profound craniomaxillofacial bone and dentition defects on account of inappropriate tissue metabolism and cellular differentiation. The coordinated activity of differentiation and viability in bone cells is indispensable for bone metabolism. Recent research demonstrates mesenchymal progenitors are responsive to PTH1R signaling for osteogenic differentiation, whereas the effect of PTH1R on cellular survival remains incompletely understood. Here, we report that mice with deletion of PTH1R in Prx1-positive mesenchymal cells (Prx1Cre;PTH1Rfl/fl) exhibit decreased alveolar bone mass due in part to apoptotic response activation. The exploration of oral bone-derived mesenchymal stem cells (OMSCs) with PTH1R deficiency suggests PTH1R signaling modulates OMSCs' apoptosis by interfering mitochondrial function and morphology. The underlying molecular mechanisms are studied by transcriptome sequencing analysis, finding that inositol trisphosphate receptor-3 (IP3R-3), an endoplasmic reticulum calcium channel protein, serves as a modulator of pro-apoptosis in OMSCs. Furthermore, we find PTH1R and its downstream protein kinase A (PKA) pathway dampen IP3R-3's expression. Of note, OMSCs with IP3R-3 overexpression recapitulate the PTH1R-deletion phenotypes, while IP3R-3 silence rescues mitochondrial dysfunction. Altogether, our study uncovers the anti-apoptotic function of PTH1R signaling in OMSCs and proves that excess apoptosis partly contributes to a weakening potential of osteogenic differentiation and aberrant mandibular development.
Collapse
Affiliation(s)
- Chen Cui
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Chuang Lu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Yanling Cai
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Yuhua Xiong
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Yihong Duan
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Kaiwen Lan
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Yi Fan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xi Wei
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| |
Collapse
|
5
|
Su M, Zheng S, Liu H, Tang TS, Hu Y. Ca 2+ homeostasis: a potential target for cancer therapies. BIOPHYSICS REPORTS 2024; 10:283-292. [PMID: 39539289 PMCID: PMC11554574 DOI: 10.52601/bpr.2024.230023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 04/19/2024] [Indexed: 11/16/2024] Open
Abstract
Calcium ions (Ca2+) play a crucial role as secondary messengers in both excitable and non-excitable cells. A complex system of proteins and molecules involved in calcium handling allows Ca2+ signals to be transduced. In cancer cells, mutations, aberrant expression, and dysregulation of these calcium handling toolkit proteins disrupt the normal Ca2+ flux between extracellular space, cytosol, endoplasmic reticulum and mitochondria, as well as the spatio-temporal patterns of Ca2+ signalling. This leads to the dysregulation of calcium-dependent effectors that control key signaling pathways involved in cancer cell proliferation, survival and invasion. Although there has been progressing in understanding the remodelling of calcium homeostasis in cancer cells and identifying key calcium transport molecules that promote malignant phenotypes, much work remains to be done to translate these fundamental findings into new tools for diagnosing and treating cancer by targeting Ca2+ homeostasis.
Collapse
Affiliation(s)
- Min Su
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
- Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, Department of Medicine and Health, Zhengzhou Research Institute of Harbin Institute of Technology, Zhengzhou 450000, China
| | - Shanliang Zheng
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
- Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, Department of Medicine and Health, Zhengzhou Research Institute of Harbin Institute of Technology, Zhengzhou 450000, China
| | - Hao Liu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
- Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, Department of Medicine and Health, Zhengzhou Research Institute of Harbin Institute of Technology, Zhengzhou 450000, China
| | - Tie-Shan Tang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Ying Hu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
- Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, Department of Medicine and Health, Zhengzhou Research Institute of Harbin Institute of Technology, Zhengzhou 450000, China
| |
Collapse
|
6
|
Guo M, Liu R, Zhang F, Qu J, Yang Y, Li X. A new perspective on liver diseases: Focusing on the mitochondria-associated endoplasmic reticulum membranes. Pharmacol Res 2024; 208:107409. [PMID: 39284429 DOI: 10.1016/j.phrs.2024.107409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 08/29/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024]
Abstract
The pathogenesis of liver diseases is multifaceted and intricate, posing a persistent global public health challenge with limited therapeutic options. Therefore, further research into liver diseases is imperative for better comprehension and advancement in treatment strategies. Numerous studies have confirmed the endoplasmic reticulum (ER) and mitochondria as key organelles driving liver diseases. Notably, the mitochondrial-associated ER membranes (MAMs) establish a physical and functional connection between the ER and mitochondria, highlighting the importance of inter-organelle communication in maintaining their functional homeostasis. This review delves into the intricate architecture and regulative mechanism of the integrated MAM that facilitate the physiological transfer of signals and substances between organelles. Additionally, we also provide a detailed overview regarding the varied pathogenic roles of malfunctioning MAM in liver diseases, focusing on its involvement in the progression of ER stress and mitochondrial dysfunction, the regulation of mitochondrial dynamics and Ca2+ transfer, as well as the disruption of lipid and glucose homeostasis. Furthermore, the current challenges and prospects associated with MAM in liver disease research are thoroughly discussed. In conclusion, elucidating the specific structure and function of MAM in different liver diseases may pave the way for novel therapeutic strategies.
Collapse
Affiliation(s)
- Mengyu Guo
- School of Life Sciences, Beijing University of Chinese Medicine, 100029, China
| | - Runping Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 100029, China
| | - Fukun Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, 100029, China
| | - Jiaorong Qu
- School of Life Sciences, Beijing University of Chinese Medicine, 100029, China
| | - Yun Yang
- School of Life Sciences, Beijing University of Chinese Medicine, 100029, China
| | - Xiaojiaoyang Li
- School of Life Sciences, Beijing University of Chinese Medicine, 100029, China.
| |
Collapse
|
7
|
Wu L, Chen J. Type 3 IP3 receptor: Its structure, functions, and related disease implications. Channels (Austin) 2023; 17:2267416. [PMID: 37818548 PMCID: PMC10569359 DOI: 10.1080/19336950.2023.2267416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 10/02/2023] [Indexed: 10/12/2023] Open
Abstract
Cell-fate decisions depend on the precise and strict regulation of multiple signaling molecules and transcription factors, especially intracellular Ca2+ homeostasis and dynamics. Type 3 inositol 1,4,5-triphosphate receptor (IP3R3) is an a tetrameric channel that can mediate the release of Ca2+ from the endoplasmic reticulum (ER) in response to extracellular stimuli. The gating of IP3R3 is regulated not only by ligands but also by other interacting proteins. To date, extensive research conducted on the basic structure of IP3R3, as well as its regulation by ligands and interacting proteins, has provided novel perspectives on its biological functions and pathogenic mechanisms. This review aims to discuss recent advancements in the study of IP3R3 and provides a comprehensive overview of the relevant literature pertaining to its structure, biological functions, and pathogenic mechanisms.
Collapse
Affiliation(s)
- Lvying Wu
- Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Jin Chen
- Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
8
|
Parthasarathi KTS, Mandal S, George JP, Gaikwad KB, Sasidharan S, Gundimeda S, Jolly MK, Pandey A, Sharma J. Aberrations in ion channels interacting with lipid metabolism and epithelial-mesenchymal transition in esophageal squamous cell carcinoma. Front Mol Biosci 2023; 10:1201459. [PMID: 37529379 PMCID: PMC10388552 DOI: 10.3389/fmolb.2023.1201459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/27/2023] [Indexed: 08/03/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is the most prevalent malignant gastrointestinal tumor. Ion channels contribute to tumor growth and progression through interactions with their neighboring molecules including lipids. The dysregulation of membrane ion channels and lipid metabolism may contribute to the epithelial-mesenchymal transition (EMT), leading to metastatic progression. Herein, transcriptome profiles of patients with ESCC were analyzed by performing differential gene expression and weighted gene co-expression network analysis to identify the altered ion channels, lipid metabolism- and EMT-related genes in ESCC. A total of 1,081 differentially expressed genes, including 113 ion channels, 487 lipid metabolism-related, and 537 EMT-related genes, were identified in patients with ESCC. Thereafter, EMT scores were correlated with altered co-expressed genes. The altered co-expressed genes indicated a correlation with EMT signatures. Interactions among 22 ion channels with 3 hub lipid metabolism- and 13 hub EMT-related proteins were determined using protein-protein interaction networks. A pathway map was generated to depict deregulated signaling pathways including insulin resistance and the estrogen receptor-Ca2+ signaling pathway in ESCC. The relationship between potential ion channels and 5-year survival rates in ESCC was determined using Kaplan-Meier plots and Cox proportional hazard regression analysis. Inositol 1,4,5-trisphosphate receptor type 3 (ITPR3) was found to be associated with poor prognosis of patients with ESCC. Additionally, drugs interacting with potential ion channels, including GJA1 and ITPR3, were identified. Understanding alterations in ion channels with lipid metabolism and EMT in ESCC pathophysiology would most likely provide potential targets for the better treatment of patients with ESCC.
Collapse
Affiliation(s)
- K. T. Shreya Parthasarathi
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Susmita Mandal
- Center for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - John Philip George
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | | | - Sruthi Sasidharan
- Institute of Bioinformatics, International Technology Park, Bangalore, India
| | - Seetaramanjaneyulu Gundimeda
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Mohit Kumar Jolly
- Center for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Rochester, MN, United States
- Center for Molecular Medicine, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
- Center for Individualized Medicine, Rochester, MN, United States
| | - Jyoti Sharma
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| |
Collapse
|
9
|
Zheng S, Wang X, Zhao D, Liu H, Hu Y. Calcium homeostasis and cancer: insights from endoplasmic reticulum-centered organelle communications. Trends Cell Biol 2023; 33:312-323. [PMID: 35915027 DOI: 10.1016/j.tcb.2022.07.004] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 12/17/2022]
Abstract
Calcium ion (Ca2+) is a ubiquitous and versatile signaling molecule controlling a wide variety of cellular processes, such as proliferation, cell death, migration, and immune response, all fundamental processes essential for the establishment of cancer. In recent decades, the loss of Ca2+ homeostasis has been considered an important driving force in the initiation and progression of malignant diseases. The primary intracellular Ca2+ store, the endoplasmic reticulum (ER), plays an essential role in maintaining Ca2+ homeostasis by coordinating with other organelles and the plasma membrane. Here, we discuss the dysregulation of ER-centered Ca2+ homeostasis in cancer, summarize Ca2+-based anticancer therapeutics, and highlight the significance of furthering our understanding of Ca2+ homeostasis regulation in cancer.
Collapse
Affiliation(s)
- Shanliang Zheng
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province 150001, China
| | - Xingwen Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province 150001, China
| | - Dong Zhao
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province 150001, China
| | - Hao Liu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province 150001, China
| | - Ying Hu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province 150001, China.
| |
Collapse
|
10
|
Means RE, Katz SG. Balancing life and death: BCL-2 family members at diverse ER-mitochondrial contact sites. FEBS J 2022; 289:7075-7112. [PMID: 34668625 DOI: 10.1111/febs.16241] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/11/2021] [Accepted: 10/19/2021] [Indexed: 01/13/2023]
Abstract
The outer mitochondrial membrane is a busy place. One essential activity for cellular survival is the regulation of membrane integrity by the BCL-2 family of proteins. Another critical facet of the outer mitochondrial membrane is its close approximation with the endoplasmic reticulum. These mitochondrial-associated membranes (MAMs) occupy a significant fraction of the mitochondrial surface and serve as key signaling hubs for multiple cellular processes. Each of these pathways may be considered as forming their own specialized MAM subtype. Interestingly, like membrane permeabilization, most of these pathways play critical roles in regulating cellular survival and death. Recently, the pro-apoptotic BCL-2 family member BOK has been found within MAMs where it plays important roles in their structure and function. This has led to a greater appreciation that multiple BCL-2 family proteins, which are known to participate in numerous functions throughout the cell, also have roles within MAMs. In this review, we evaluate several MAM subsets, their role in cellular homeostasis, and the contribution of BCL-2 family members to their functions.
Collapse
Affiliation(s)
- Robert E Means
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Samuel G Katz
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
11
|
Li S, Zhang X, Lou C, Gu Y, Zhao J. Preoperative peripheral blood inflammatory markers especially the fibrinogen-to-lymphocyte ratio and novel FLR-N score predict the prognosis of patients with early-stage resectable extrahepatic cholangiocarcinoma. Front Oncol 2022; 12:1003845. [PMID: 36387142 PMCID: PMC9659886 DOI: 10.3389/fonc.2022.1003845] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/10/2022] [Indexed: 01/27/2023] Open
Abstract
Background Systemic inflammation is important in the development of extrahepatic cholangiocarcinoma (ECC). The aim of this study was to compare the prognostic power of preoperative peripheral blood inflammatory markers and the novel FLR-N score in patients with resectable ECC. Methods A total of 140 patients with resectable ECC and 140 healthy controls (HCs) were recruited for the study. The Mann−Whitney U test was used to evaluate the differences in inflammatory markers between groups. Kaplan−Meier and Cox regression analyses were used to evaluate the prognostic power of preoperative fibrinogen, albumin, prealbumin, bilirubin, neutrophils, lymphocytes, monocytes, platelets, fibrinogen-to-lymphocyte ratio (FLR), fibrinogen-to-albumin ratio (FAR), fibrinogen-to-prealbumin ratio (FPR), neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), monocyte-to-lymphocyte ratio (MLR), FLR-neutrophil (FLR-N) score, and CA19-9 in patients with resectable ECC. Nomogram was developed based on the results of multivariate Cox analyses. Results Patients with resectable ECC had significantly higher levels of neutrophils, monocytes, fibrinogen, FLR, FAR, FPR, NLR, PLR, and MLR and lower levels of lymphocytes, albumin, and prealbumin than HCs (all P < 0.01). Albumin, prealbumin, and FPR had a good ability to distinguish between ECC patients with total bilirubin < 34 µmol/L and HCs (AUCs of 0.820, 0.827, and 0.836, respectively). Kaplan−Meier analysis showed that high neutrophil, fibrinogen, FLR, FAR, PLR, MLR, and FLR-N score values were associated with poor survival in patients with resectable ECC. Multivariate analyses indicated that neutrophils (P = 0.022), FLR (P = 0.040), FLR-N score (P < 0.0001), and positive lymph node metastasis (P = 0.016) were independent factors for overall survival (OS). Nomogram were developed to predict OS for patients with ECC. Conclusion The prognostic roles of inflammatory markers in patients with resectable ECC were different. The preoperative neutrophil count, FLR and FLR-N score could serve as noninvasive markers for predicting the prognosis of resectable ECC.
Collapse
Affiliation(s)
- Shijie Li
- Department of Interventional Radiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xingli Zhang
- Biotherapy Center, Harbin Medical University Cancer Hospital, Harbin, China
| | - Changjie Lou
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Yuanlong Gu
- Biotherapy Center, Harbin Medical University Cancer Hospital, Harbin, China
- Department of Interventional Oncology, Taizhou Municipal Hospital, Taizhou, Zhejiang, China
- *Correspondence: Juan Zhao, ; Yuanlong Gu,
| | - Juan Zhao
- Biotherapy Center, Harbin Medical University Cancer Hospital, Harbin, China
- *Correspondence: Juan Zhao, ; Yuanlong Gu,
| |
Collapse
|
12
|
Recent Developments on the Roles of Calcium Signals and Potential Therapy Targets in Cervical Cancer. Cells 2022; 11:cells11193003. [PMID: 36230965 PMCID: PMC9563098 DOI: 10.3390/cells11193003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 09/14/2022] [Accepted: 09/20/2022] [Indexed: 12/24/2022] Open
Abstract
Intracellular calcium (Ca2+) concentration ([Ca2+]i) is implicated in proliferation, invasion, and metastasis in cancerous tissues. A variety of oncologic therapies and some candidate drugs induce their antitumor effects (in part or in whole) through the modulation of [Ca2+]i. Cervical cancer is one of most common cancers among women worldwide. Recently, major research advances relating to the Ca2+ signals in cervical cancer are emerging. In this review, we comprehensively describe the current progress concerning the roles of Ca2+ signals in the occurrence, development, and prognosis of cervical cancer. It will enhance our understanding of the causative mechanism of Ca2+ signals in cervical cancer and thus provide new sights for identifying potential therapeutic targets for drug discovery.
Collapse
|
13
|
Morgado-Cáceres P, Liabeuf G, Calle X, Briones L, Riquelme JA, Bravo-Sagua R, Parra V. The aging of ER-mitochondria communication: A journey from undifferentiated to aged cells. Front Cell Dev Biol 2022; 10:946678. [PMID: 36060801 PMCID: PMC9437272 DOI: 10.3389/fcell.2022.946678] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/20/2022] [Indexed: 01/10/2023] Open
Abstract
The complex physiology of eukaryotic cells requires that a variety of subcellular organelles perform unique tasks, even though they form highly dynamic communication networks. In the case of the endoplasmic reticulum (ER) and mitochondria, their functional coupling relies on the physical interaction between their membranes, mediated by domains known as mitochondria-ER contacts (MERCs). MERCs act as shuttles for calcium and lipid transfer between organelles, and for the nucleation of other subcellular processes. Of note, mounting evidence shows that they are heterogeneous structures, which display divergent behaviors depending on the cell type. Furthermore, MERCs are plastic structures that remodel according to intra- and extracellular cues, thereby adjusting the function of both organelles to the cellular needs. In consonance with this notion, the malfunction of MERCs reportedly contributes to the development of several age-related disorders. Here, we integrate current literature to describe how MERCs change, starting from undifferentiated cells, and their transit through specialization, malignant transformation (i.e., dedifferentiation), and aging/senescence. Along this journey, we will review the function of MERCs and their relevance for pivotal cell types, such as stem and cancer cells, cardiac, skeletal, and smooth myocytes, neurons, leukocytes, and hepatocytes, which intervene in the progression of chronic diseases related to age.
Collapse
Affiliation(s)
- Pablo Morgado-Cáceres
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas e Instituto de Nutrición y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago, Chile
- Departamento de Bioquímica y Biología Molecular y Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
| | - Gianella Liabeuf
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas e Instituto de Nutrición y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago, Chile
- Laboratorio de Obesidad y Metabolismo Energético (OMEGA), Instituto de Nutrición y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago, Chile
- Facultad de Salud y Ciencias Sociales, Escuela de Nutrición y Dietética, Universidad de las Américas, Santiago, Chile
| | - Ximena Calle
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas e Instituto de Nutrición y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago, Chile
- Departamento de Bioquímica y Biología Molecular y Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Lautaro Briones
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas e Instituto de Nutrición y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago, Chile
- Laboratorio de Obesidad y Metabolismo Energético (OMEGA), Instituto de Nutrición y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago, Chile
- Departamento de Nutrición y Salud Pública, Facultad de Ciencias de la Salud y de los Alimentos, Universidad del Bío-Bío, Chillán, Chile
| | - Jaime A. Riquelme
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas e Instituto de Nutrición y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago, Chile
- Departamento de Bioquímica y Biología Molecular y Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Roberto Bravo-Sagua
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas e Instituto de Nutrición y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago, Chile
- Laboratorio de Obesidad y Metabolismo Energético (OMEGA), Instituto de Nutrición y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago, Chile
- Red de Investigación en Envejecimiento Saludable, Consorcio de Universidades del Estado de Chile, Santiago, Chile
- *Correspondence: Roberto Bravo-Sagua, ; Valentina Parra,
| | - Valentina Parra
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas e Instituto de Nutrición y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago, Chile
- Departamento de Bioquímica y Biología Molecular y Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
- Red para el Estudio de Enfermedades Cardiopulmonares de alta letalidad (REECPAL), Universidad de Chile, Santiago, Chile
- *Correspondence: Roberto Bravo-Sagua, ; Valentina Parra,
| |
Collapse
|
14
|
Marchi S, Pinton P. Mitochondria in the line of fire. Cell Death Differ 2022; 29:1301-1303. [DOI: 10.1038/s41418-022-01034-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 11/09/2022] Open
|
15
|
Zheng W, Bai X, Zhou Y, Yu L, Ji D, Zheng Y, Meng N, Wang H, Huang Z, Chen W, Yam JWP, Xu Y, Cui Y. Transcriptional ITPR3 as potential targets and biomarkers for human pancreatic cancer. Aging (Albany NY) 2022; 14:4425-4444. [PMID: 35580861 PMCID: PMC9186782 DOI: 10.18632/aging.204080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 05/02/2022] [Indexed: 11/26/2022]
Abstract
Inositol 1,4,5-Triphosphate Receptor Family (ITPRs) are necessary intracellular Ca2+-release channel encoders and participate in mammalian cell physiological and pathological processes. Previous studies have suggested that ITPRs participate in tumorigenesis of multiple cancers. Nevertheless, the diverse expression profiles and prognostic significance of three ITPRs in pancreatic cancer have yet to be uncovered. In this work, we examined the expression levels and survival dates of ITPRs in patients with pancreatic cancer. As a result, we identified that ITPR1 and ITPR3 expression levels are significantly elevated in cancerous specimens. Survival data revealed that over-expression of ITPR2 and ITPR3 resulted in unfavourable overall survival and pathological stage. The multivariate Cox logistic regression analysis showed that ITPR3 could be an independent risk factor for PAAD patient survival. Moreover, to investigate how ITPRs work, co-expressed genes, alterations, protein-protein interaction, immune infiltration, methylation, and functional enrichment of ITPRs were also analyzed. Then, we evaluated these findings in clinical samples. Moreover, the gain and loss of function of ITPR3 were also conducted. The electron microscope assay was employed to explore the role of ITPR3 in pancreatic cancer cell lines' endoplasmic reticulum stress. In summary, our findings demonstrated that ITPR3 has the potential to be drug targets and biomarkers for human pancreatic cancer.
Collapse
Affiliation(s)
- Wangyang Zheng
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin 150086, China
- Department II of Gastroenterology, Third Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Xue Bai
- Department of Clinic of Internal Medicine I, Ulm University, Ulm 89081, Germany
| | - Yongxu Zhou
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin 150086, China
| | - Liang Yu
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin 150086, China
| | - Daolin Ji
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin 150086, China
- Department of Hepatopancreatobiliary Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Yuling Zheng
- Department of Pediatric, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Nanfeng Meng
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Hang Wang
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Ziyue Huang
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Wangming Chen
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Judy Wai Ping Yam
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Yi Xu
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin 150086, China
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Yunfu Cui
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| |
Collapse
|
16
|
Moy RH, Nguyen A, Loo JM, Yamaguchi N, Kajba CM, Santhanam B, Ostendorf BN, Wu YG, Tavazoie S, Tavazoie SF. Functional genetic screen identifies ITPR3/calcium/RELB axis as a driver of colorectal cancer metastatic liver colonization. Dev Cell 2022; 57:1146-1159.e7. [PMID: 35487218 PMCID: PMC9446818 DOI: 10.1016/j.devcel.2022.04.010] [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: 09/12/2021] [Revised: 03/02/2022] [Accepted: 04/06/2022] [Indexed: 11/29/2022]
Abstract
Metastatic colonization is the primary cause of death from colorectal cancer (CRC). We employed genome-scale in vivo short hairpin RNA (shRNA) screening and validation to identify 26 promoters of CRC liver colonization. Among these genes, we identified a cluster that contains multiple targetable genes, including ITPR3, which promoted liver-metastatic colonization and elicited similar downstream gene expression programs. ITPR3 is a caffeine-sensitive inositol 1,4,5-triphosphate (IP3) receptor that releases calcium from the endoplasmic reticulum and enhanced metastatic colonization by inducing expression of RELB, a transcription factor that is associated with non-canonical NF-κB signaling. Genetic, cell biological, pharmacologic, and clinical association studies revealed that ITPR3 and RELB drive CRC colony formation by promoting cell survival upon substratum detachment or hypoxic exposure. RELB was sufficient to drive colonization downstream of ITPR3. Our findings implicate the ITPR3/calcium/RELB axis in CRC metastatic colony formation and uncover multiple clinico-pathologically associated targetable proteins as drivers of CRC metastatic colonization.
Collapse
Affiliation(s)
- Ryan H Moy
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY 10065, USA
| | - Alexander Nguyen
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY 10065, USA
| | - Jia Min Loo
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY 10065, USA
| | - Norihiro Yamaguchi
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY 10065, USA
| | - Christina M Kajba
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY 10065, USA
| | - Balaji Santhanam
- Department of Biochemistry and Molecular Biophysics and Department of Systems Biology, Columbia University, New York, NY 10032, USA; Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Benjamin N Ostendorf
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY 10065, USA
| | - Y Gloria Wu
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY 10065, USA
| | - Saeed Tavazoie
- Department of Biochemistry and Molecular Biophysics and Department of Systems Biology, Columbia University, New York, NY 10032, USA; Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Sohail F Tavazoie
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
17
|
Zhang S, Cao S, Gong M, Zhang W, Zhang W, Zhu Z, Wu S, Yue Y, Qian W, Ma Q, Wang S, Wang Z. Mechanically activated ion channel Piezo1 contributes to melanoma malignant progression through AKT/mTOR signaling. Cancer Biol Ther 2022; 23:336-347. [PMID: 36112948 PMCID: PMC9037449 DOI: 10.1080/15384047.2022.2060015] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Melanoma is a highly aggressive cancer that can metastasize at early stage. The aim of this study is to clarify the role of Piezo1 and its potential mechanism in regulating the malignant phenotypes of melanoma. In the present study, we first showed that Piezo1 was abnormally expressed in melanoma, which accelerated the malignant progression by activating AKT/mTOR signaling. Firstly, we found that Piezo1 was upregulated in melanoma and associated with poor survival. Additionally, Piezo1 knockdown significantly weakened intracellular calcium signal and viability of melanoma cells. Furthermore, Piezo1 knockdown inhibited the transendothelial migration and invasion in vitro, as well as metastasis in vivo. Mechanistically, we found that Piezo1 activated AKT/mTOR signaling to maintain malignant phenotypes of melanoma. Therefore, Piezo1 acts as an oncogene in melanoma cells and provides a novel candidate for melanoma diagnosis and treatment.
Collapse
Affiliation(s)
- Simei Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Shuang Cao
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin, Molecular and Translational Kidney Research, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Mengyuan Gong
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Wunai Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Weifan Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Zeen Zhu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Shuai Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yangyang Yue
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Weikun Qian
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Shengpeng Wang
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| |
Collapse
|
18
|
Inositol (1,4,5)-Trisphosphate Receptors in Invasive Breast Cancer: A New Prognostic Tool? Int J Mol Sci 2022; 23:ijms23062962. [PMID: 35328381 PMCID: PMC8955728 DOI: 10.3390/ijms23062962] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/04/2022] [Accepted: 03/06/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary The inositol-trisphosphate receptor (IP3R) is a key player in physiological and pathological intracellular calcium signaling. The objective of the present study was to assess the putative value of the three IP3R subtypes as prognostic biomarkers in breast cancer. We found that IP3R3 is the most strongly expressed subtype in breast cancer tissue. Furthermore, IP3R3 and IP3R1 are significantly more expressed in invasive breast cancer tissue than in non-tumor tissue. In contrast to IP3R1 and IP3R2, the expression of IP3R3 was positively correlated with prognostic factors including tumor size, regional node invasion, histologic grade, proliferation index, and hormonal status. By analyzing public databases, we found that the expression of all IP3R subtypes is significantly correlated with the overall survival and disease-free survival of patients with breast cancer. We conclude that relative to the other two IP3R subtypes, IP3R3 expression is upregulated in breast cancer and is correlated with prognostic factors. We strongly believe that our results will open up new perspectives with regard to the link between IP3Rs and breast cancer aggressiveness. Abstract Breast cancer is the leading cause of cancer death among women in worldwide and France. The disease prognosis and treatment differ from one breast cancer subtype to another, and the disease outcome depends on many prognostic factors. Deregulation of ion flux (especially Ca2+ flux) is involved in many pathophysiology processes, including carcinogenesis. Inside the cell, the inositol-trisphosphate receptor (IP3R) is a major player in the regulation of the Ca2+ flux from the endoplasmic reticulum to the cytoplasm. The IP3Rs (and particularly the IP3R3 subtype) are known to be involved in proliferation, migration, and invasion processes in breast cancer cell lines. The objective of the present study was to evaluate the potential value of IP3Rs as prognostic biomarkers in breast cancer. We found that expression levels of IP3R3 and IP3R1 (but not IP3R2) were significantly higher in invasive breast cancer of no special type than in non-tumor tissue from the same patient. However, the IP3R3 subtype was expressed more strongly than the IP3R1 and IP3R2 subtypes. Furthermore, the expression of IP3R3 (but not of IP3R1 or IP3R2) was positively correlated with prognostic factors such as tumor size, regional node invasion, histologic grade, proliferation index, and hormone receptor status. In an analysis of public databases, we found that all IP3Rs types are significantly associated with overall survival and progression-free survival in patients with breast cancer. We conclude that relative to the other two IP3R subtypes, IP3R3 expression is upregulated in breast cancer and is correlated with prognostic factors.
Collapse
|
19
|
iASPP suppresses Gp78-mediated TMCO1 degradation to maintain Ca 2+ homeostasis and control tumor growth and drug resistance. Proc Natl Acad Sci U S A 2022; 119:2111380119. [PMID: 35121659 PMCID: PMC8832991 DOI: 10.1073/pnas.2111380119] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2021] [Indexed: 12/13/2022] Open
Abstract
Accumulating preclinical and clinical evidence has supported a central role for alterations in Ca2+ homeostasis in the development of cancer. TMCO1 protein is an identified Ca2+-channel protein, while its roles in cancer remain obscure. Here, we found that TMCO1 is increased in colon cancer tissues. In addition, it is a substrate of E3 ligase Gp78. Enhanced oncogene iASPP stabilizes TMCO1 by competitively binding with Gp78. Inhibition of iASPP-TMCO1 sensitizes cancer cells’ response to Ca2+-induced apoptosis. This study has improved our fundamental understanding of the Ca2+ homeostasis in cancer cells. iASPP-TMCO1 axis may present a promising therapeutic target that can combine the conventional drugs to reinforce Ca2+-dependent apoptosis. Ca2+ release from the endoplasmic reticulum (ER) is an essential event in the modulation of Ca2+ homeostasis, which is coordinated by multiple biological processes, ranging from cell proliferation to apoptosis. Deregulated Ca2+ homeostasis is linked with various cancer hallmarks; thus, uncovering the mechanisms underlying Ca2+ homeostasis dynamics may lead to new anticancer treatment strategies. Here, we demonstrate that a reported Ca2+-channel protein TMCO1 (transmembrane and coiled-coil domains 1) is overexpressed in colon cancer tissues at protein levels but not at messenger RNA levels in colon cancer. Further study revealed that TMCO1 is a substrate of ER-associated degradation E3 ligase Gp78. Intriguingly, Gp78-mediated TMCO1 degradation at K186 is under the control of the iASPP (inhibitor of apoptosis-stimulating protein of p53) oncogene. Mechanistically, iASPP robustly reduces ER Ca2+ stores, mainly by competitively binding with Gp78 and interfering with Gp78-mediated TMCO1 degradation. A positive correlation between iASPP and TMCO1 proteins is further validated in human colon tissues. Inhibition of iASPP-TMCO1 axis promotes cytosolic Ca2+ overload–induced apoptotic cell death, reducing tumor growth both in vitro and in vivo. Thus, iASPP-TMCO1 represents a promising anticancer treatment target by modulating Ca2+ homeostasis.
Collapse
|
20
|
Rodrigues MA, Gomes DA, Cosme AL, Sanches MD, Resende V, Cassali GD. Inositol 1,4,5-trisphosphate receptor type 3 (ITPR3) is overexpressed in cholangiocarcinoma and its expression correlates with S100 calcium-binding protein A4 (S100A4). Biomed Pharmacother 2022; 145:112403. [PMID: 34798470 PMCID: PMC8678364 DOI: 10.1016/j.biopha.2021.112403] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/31/2021] [Accepted: 11/03/2021] [Indexed: 01/03/2023] Open
Abstract
Cholangiocarcinoma (CCA) is the second most malignant neoplasm in the liver that arises from the biliary tree. CCA is associated with a poor prognosis, and the key players involved in its pathogenesis are still not well understood. Receptor tyrosine kinases (RTKs), such as epidermal growth factor receptor (EGFR), can mediate intracellular calcium (Ca2+) signaling pathways via inositol 1,4,5-trisphosphate (InsP3), activating inositol 1,4,5-trisphosphate receptors (ITPRs) and regulating tumor growth. ITPR isoform 3 (ITPR3) is the main intracellular Ca2+ release channel in cholangiocytes. The effects of intracellular Ca2+ are mediated by calcium-binding proteins such as Calmodulin and S100 calcium-binding protein A4 (S100A4). However, the clinicopathological and biological significance of EGFR, ITPR3 and S100A4 in CCA remains unclear. Thus, the present work investigates the immunoexpression of these three proteins in 59 CCAs from patients who underwent curative surgical treatment and correlates the data with clinicopathological features and survival. High ITPR3 expression was correlated with CA 19-9 levels, TNM stage and lymph node metastasis (N). Furthermore, ITPR3 expression was increased in distal CCA compared to control bile ducts and intrahepatic and perihilar CCAs. These observations were confirmed by proteomic analysis. ITPR3 and S100A4 clinical scores were significantly correlated. Furthermore, it was demonstrated that EGF induces calcium signaling in a cholangiocarcinoma cell line and ITPR3 colocalizes with nonmuscle myosin IIA (NMIIA). In summary, ITPR3 overexpression could contribute to CCA progression and it may represent a potential therapeutic target.
Collapse
Affiliation(s)
- Michele A. Rodrigues
- Department of General Pathology, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, Minas Gerais, CEP: 31270-901, Brazil
| | - Dawidson A. Gomes
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, Minas Gerais, CEP: 31270-901, Brazil
| | - Ana Luiza Cosme
- School of Medicine, Department of Surgery, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena 190, Belo Horizonte, Minas Gerais, CEP: 30130-100, Brazil
| | - Marcelo Dias Sanches
- School of Medicine, Department of Surgery, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena 190, Belo Horizonte, Minas Gerais, CEP: 30130-100, Brazil.,Hepatopancreatobiliary Division, Clinical Hospital, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena 110, Belo Horizonte, Minas Gerais, CEP: 30130-100, Brazil
| | - Vivian Resende
- School of Medicine, Department of Surgery, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena 190, Belo Horizonte, Minas Gerais, CEP: 30130-100, Brazil.,Hepatopancreatobiliary Division, Clinical Hospital, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena 110, Belo Horizonte, Minas Gerais, CEP: 30130-100, Brazil
| | - Geovanni D. Cassali
- Department of General Pathology, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, Minas Gerais, CEP: 31270-901, Brazil.,Corresponding author: Department of General Pathology, Instituto de Ciências Biológicas, Bloco C3, Sala 102, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627 Belo Horizonte–MG, Brazil 31270-901. Tel: +55 31 34092891.
| |
Collapse
|
21
|
Dos Santos ML, França A, Lima Filho ACM, Florentino RM, Diniz PH, Oliveira Lemos F, Gonçalves CAX, Coelho VL, Lima CX, Foureaux G, Nathanson MH, Vidigal PVT, Leite MF. Inositol 1,4,5-trisphosphate receptor type 3 is involved in resistance to apoptosis and maintenance of human hepatocellular carcinoma. Oncol Lett 2022; 23:32. [PMID: 34966448 PMCID: PMC8669656 DOI: 10.3892/ol.2021.13150] [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: 03/02/2021] [Accepted: 09/24/2021] [Indexed: 11/06/2022] Open
Abstract
The expression of the inositol 1,4,5-trisphosphate receptor type 3 (ITRP3) in hepatocytes is a common event in the pathogenesis of hepatocellular carcinoma (HCC), regardless of the type of underlying liver disease. However, it is not known whether ITPR3 expression in hepatocytes is involved in tumor maintenance. The aim of the present study was to determine whether there is an association between ITPR3 expression and clinical and morphological parameters using HCC samples obtained from liver explants from patients (n=53) with different etiologies of underlying chronic liver disease (CLD). ITPR3 expression, mitosis and apoptosis were analyzed in human liver samples by immunohistochemistry. Clinical and event-free survival data were combined to assess the relationship between ITPR3 and liver cancer growth in patients. RNA sequencing analysis was performed to identify apoptotic genes altered by ITPR3 expression in a liver tumor cell line. ITPR3 was highly expressed in HCC tumor cells relative to adjacent CLD tissue and healthy livers. There was an inverse correlation between ITPR3 expression and mitotic and apoptotic indices in HCC, suggesting that ITPR3 contributed to the maintenance of HCC by promoting resistance to apoptosis. This was confirmed by the upregulation of CTSB, CHOP and GADD45, genes involved in the apoptotic pathway in HCC. The expression of ITPR3 in the liver may be a promising prognostic marker of HCC.
Collapse
Affiliation(s)
- Marcone Loiola Dos Santos
- Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Andressa França
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Antônio Carlos Melo Lima Filho
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Rodrigo M. Florentino
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Paulo Henrique Diniz
- Department of Internal Medicine, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Fernanda Oliveira Lemos
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Carlos Alberto Xavier Gonçalves
- Coordination of Biotechnology, SENAI's Innovation Institute for Biosynthetics and Fibers, SENAI CETIQT, Rio de Janeiro 20961-020, Brazil
| | - Vitor Lima Coelho
- Coordination of Biotechnology, SENAI's Innovation Institute for Biosynthetics and Fibers, SENAI CETIQT, Rio de Janeiro 20961-020, Brazil
| | - Cristiano Xavier Lima
- Department of Surgery, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais 30 130-100, Brazil
| | - Giselle Foureaux
- Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Michael H. Nathanson
- Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520-8056, USA
| | - Paula Vieira Teixeira Vidigal
- Department of Pathological Anatomy and Forensic Medicine of Hospital das Clínicas, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais 30 130-100, Brazil
| | - M. Fátima Leite
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| |
Collapse
|
22
|
The basis of nuclear phospholipase C in cell proliferation. Adv Biol Regul 2021; 82:100834. [PMID: 34710785 DOI: 10.1016/j.jbior.2021.100834] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/17/2021] [Accepted: 10/21/2021] [Indexed: 11/23/2022]
Abstract
Ca2+ is a highly versatile intracellular signal that regulates many biological processes such as cell death and proliferation. Broad Ca2+-signaling machinery is used to assemble signaling systems with a precise spatial and temporal resolution to achieve this versatility. Ca2+-signaling components can be organized in different regions of the cell and local increases in Ca2+ within the nucleus can regulate different cellular functions from the increases in cytosolic Ca2+. However, the mechanisms and pathways that promote localized increases in Ca2+ levels in the nucleus are still under investigation. This review presents evidence that the nucleus has its own Ca2+ stores and signaling machinery, which modulate processes such as cell proliferation and tumor growth. We focus on what is known about the functions of nuclear Phospholipase C (PLC) in the generation of nuclear Ca2+ transients that are involved in cell proliferation.
Collapse
|
23
|
Arige V, Terry LE, Malik S, Knebel TR, Wagner II LE, Yule DI. CREB regulates the expression of type 1 inositol 1,4,5-trisphosphate receptors. J Cell Sci 2021; 134:jcs258875. [PMID: 34533188 PMCID: PMC8601716 DOI: 10.1242/jcs.258875] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 09/13/2021] [Indexed: 12/16/2022] Open
Abstract
Inositol 1,4,5-trisphosphate (IP3) receptors (IP3Rs) play a central role in regulating intracellular Ca2+ signals in response to a variety of internal and external cues. Dysregulation of IP3R signaling is the underlying cause for numerous pathological conditions. It is well established that the activities of IP3Rs are governed by several post-translational modifications, including phosphorylation by protein kinase A (PKA). However, the long-term effects of PKA activation on expression of IP3R subtypes remains largely unexplored. In this report, we investigate the effects of chronic stimulation and tonic activity of PKA on the expression of IP3R subtypes. We demonstrate that expression of the type 1 IP3R (IP3R1) is augmented upon prolonged activation of PKA or upon ectopic overexpression of cyclic AMP-response element-binding protein (CREB) without altering IP3R2 and IP3R3 abundance. By contrast, inhibition of PKA or blocking CREB diminished IP3R1 expression. We also demonstrate that agonist-induced Ca2+-release mediated by IP3R1 is significantly attenuated upon blocking of CREB. Moreover, CREB - by regulating the expression of KRAS-induced actin-interacting protein (KRAP) - ensures correct localization and licensing of IP3R1. Overall, we report a crucial role for CREB in governing both the expression and correct localization of IP3R1. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
| | | | | | | | | | - David I. Yule
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642, USA
| |
Collapse
|
24
|
Wei YL, Wen B, Gao JZ, Chen ZZ. Brain transcriptome analysis reveals genes involved in parental care behaviour in discus fish (Symphysodon haraldi). Gen Comp Endocrinol 2021; 309:113793. [PMID: 33887271 DOI: 10.1016/j.ygcen.2021.113793] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 04/14/2021] [Accepted: 04/16/2021] [Indexed: 10/21/2022]
Abstract
Parental care is common in mammals and allows offspring to obtain milk, a substance rich in a range of nutritional and non-nutritional factors crucial to the survival of newborns. The discus fish Symphysodon spp., an Amazonian cichlid, shows an unusual behaviour: Free-swimming fry bite on their parents' skin mucus for growth and development during the first month after hatching. This is similar to the breastfeeding behaviour of mammals, but little is known about the regulatory mechanism by which discus secrete 'milk' and the related genes involved in parental care. Here, transcriptome sequencing was performed by using the brain tissues of female discus fish in parental and non-parental care. The results showed that a total of 86 differentially expressed genes (71 up-regulated genes and 15 down-regulated genes) were obtained by comparing parental with non-parental discus fish, including up-regulated LAPTM, FOXB, SOX1S, OTX2 and NR1F2, and down-regulated EDNRB, PRKCD, H1-5 and HBE. Through functional enrichment analysis, a total of 20 pathways were identified, e.g., estrogen signaling pathway, inflammatory mediator regulation of TRP channels, vascular smooth muscle contraction, GnRH signaling pathway, neurotrophin signaling pathway, NOD-like receptor signaling pathway, Jak-STAT signaling pathway, Fc gamma R-mediated phagocytosis, serotonergic synapse, autophagy-animal and cytokine-cytokine receptor interaction. These pathways and related genes might play important roles in the regulation of discus 'milk' secretion.
Collapse
Affiliation(s)
- Yu-Ling Wei
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai 201306, China; Shanghai Collaborative Innovation for Aquatic Animal Genetics and Breeding, Shanghai Ocean University, Shanghai 201306, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China
| | - Bin Wen
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai 201306, China; Shanghai Collaborative Innovation for Aquatic Animal Genetics and Breeding, Shanghai Ocean University, Shanghai 201306, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China.
| | - Jian-Zhong Gao
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai 201306, China; Shanghai Collaborative Innovation for Aquatic Animal Genetics and Breeding, Shanghai Ocean University, Shanghai 201306, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China
| | - Zai-Zhong Chen
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai 201306, China; Shanghai Collaborative Innovation for Aquatic Animal Genetics and Breeding, Shanghai Ocean University, Shanghai 201306, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China.
| |
Collapse
|
25
|
Woll KA, Van Petegem F. Calcium Release Channels: Structure and Function of IP3 Receptors and Ryanodine Receptors. Physiol Rev 2021; 102:209-268. [PMID: 34280054 DOI: 10.1152/physrev.00033.2020] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Ca2+-release channels are giant membrane proteins that control the release of Ca2+ from the endoplasmic and sarcoplasmic reticulum. The two members, ryanodine receptors (RyRs) and inositol-1,4,5-trisphosphate Receptors (IP3Rs), are evolutionarily related and are both activated by cytosolic Ca2+. They share a common architecture, but RyRs have evolved additional modules in the cytosolic region. Their massive size allows for the regulation by tens of proteins and small molecules, which can affect the opening and closing of the channels. In addition to Ca2+, other major triggers include IP3 for the IP3Rs, and depolarization of the plasma membrane for a particular RyR subtype. Their size has made them popular targets for study via electron microscopic methods, with current structures culminating near 3Å. The available structures have provided many new mechanistic insights int the binding of auxiliary proteins and small molecules, how these can regulate channel opening, and the mechanisms of disease-associated mutations. They also help scrutinize previously proposed binding sites, as some of these are now incompatible with the structures. Many questions remain around the structural effects of post-translational modifications, additional binding partners, and the higher-order complexes these channels can make in situ. This review summarizes our current knowledge about the structures of Ca2+-release channels and how this informs on their function.
Collapse
Affiliation(s)
- Kellie A Woll
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Filip Van Petegem
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
26
|
Thongchot S, Vidoni C, Ferraresi A, Loilome W, Khuntikeo N, Sangkhamanon S, Titapun A, Isidoro C, Namwat N. Cancer-Associated Fibroblast-Derived IL-6 Determines Unfavorable Prognosis in Cholangiocarcinoma by Affecting Autophagy-Associated Chemoresponse. Cancers (Basel) 2021; 13:cancers13092134. [PMID: 33925189 PMCID: PMC8124468 DOI: 10.3390/cancers13092134] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/22/2021] [Accepted: 04/26/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary We aimed to validate with clinical and molecular data the hypothesis that CAF infiltration and release of IL-6 predict poor prognosis in CCA patients following dysregulation of autophagy in cancer cells. Stromal IL-6 and cancer-cell-associated autophagy proteins were assayed by Tissue MicroArray immunohistochemistry and their expression correlated with overall survival (OS) in a cohort of 70 CCA patients. We found that patients bearing a CCA with low stromal IL-6 and active autophagy flux in the cancer cells have the best prognosis and this correlates with a more effective response to post-operative chemotherapy. A similar trend was observed in CCA patients from the TCGA database. In vitro experiments with primary CAFs isolated from human CCA showed that IL-6 impairs the autophagy-associated apoptotic response to 5-FU in human CCA cells. Stromal IL-6 inhibition of autophagy in cancer cells was confirmed in an animal model of CCA. Our data support a therapeutic strategy that includes drugs limiting the stromal inflammation and enhancing autophagy to improve the survival of CCA patients. Abstract Background: Interleukin-6 (IL-6) released by cancer-associated fibroblasts (CAFs) has been shown to associate with the malignant behavior of cholangiocarcinoma (CCA). Here, we aimed to validate with clinical and molecular data the hypothesis that CAF infiltration and release of IL-6 predict poor prognosis in CCA patients following dysregulation of autophagy in cancer cells. Methods: Stromal IL-6 and cancer-cell-associated autophagy proteins LC3 and p62 were assayed by Tissue MicroArray immunohistochemistry and their expression correlated with overall survival (OS) in a cohort of 70 CCA patients. The 5-FU cytotoxicity and autophagy were determined in CCA cells cultured with CAF-conditioned medium. Results: We show that patients bearing a CCA with low production of stromal IL-6 and active autophagy flux in the cancer cells have the best prognosis and this correlates with a more effective response to post-operative chemotherapy. A similar trend was observed in CCA patients from the TCGA database. In vitro genetic manipulation of IL-6 production by primary CAFs isolated from human CCA showed that IL-6 impairs the autophagy-associated apoptotic response to 5-FU in human CCA cells. Stromal IL-6 inhibition of autophagy in cancer cells was confirmed in an animal model of CCA. Conclusion: Our data support a therapeutic strategy that includes autophagy-enhancing drugs along with adjuvants limiting the stromal inflammation (i.e., the secretion of IL-6) to improve the survival of CCA patients.
Collapse
Affiliation(s)
- Suyanee Thongchot
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, 123 Mitraparp Highway, Khon Kaen 40002, Thailand; (S.T.); (W.L.)
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale “A. Avogadro”, Via Solaroli 17, 28100 Novara, Italy; (C.V.); (A.F.)
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Chiara Vidoni
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale “A. Avogadro”, Via Solaroli 17, 28100 Novara, Italy; (C.V.); (A.F.)
| | - Alessandra Ferraresi
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale “A. Avogadro”, Via Solaroli 17, 28100 Novara, Italy; (C.V.); (A.F.)
| | - Watcharin Loilome
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, 123 Mitraparp Highway, Khon Kaen 40002, Thailand; (S.T.); (W.L.)
- Cholangiocarcinoma Research Institute, Khon Kaen University, 123 Mitraparp Highway, Khon Kaen 40002, Thailand; (N.K.); (S.S.); (A.T.)
| | - Narong Khuntikeo
- Cholangiocarcinoma Research Institute, Khon Kaen University, 123 Mitraparp Highway, Khon Kaen 40002, Thailand; (N.K.); (S.S.); (A.T.)
- Department of Surgery, Faculty of Medicine, Khon Kaen University, 123 Mitraparp Highway, Khon Kaen 40002, Thailand
| | - Sakkarn Sangkhamanon
- Cholangiocarcinoma Research Institute, Khon Kaen University, 123 Mitraparp Highway, Khon Kaen 40002, Thailand; (N.K.); (S.S.); (A.T.)
- Department of Pathology, Faculty of Medicine, Khon Kaen University, 123 Mitraparp Highway, Khon Kaen 40002, Thailand
| | - Attapol Titapun
- Cholangiocarcinoma Research Institute, Khon Kaen University, 123 Mitraparp Highway, Khon Kaen 40002, Thailand; (N.K.); (S.S.); (A.T.)
- Department of Surgery, Faculty of Medicine, Khon Kaen University, 123 Mitraparp Highway, Khon Kaen 40002, Thailand
| | - Ciro Isidoro
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale “A. Avogadro”, Via Solaroli 17, 28100 Novara, Italy; (C.V.); (A.F.)
- Correspondence: (C.I.); (N.N.); Tel.: +39-(0321)-660507 (C.I.); +66-6-3635-2491 (N.N.)
| | - Nisana Namwat
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, 123 Mitraparp Highway, Khon Kaen 40002, Thailand; (S.T.); (W.L.)
- Cholangiocarcinoma Research Institute, Khon Kaen University, 123 Mitraparp Highway, Khon Kaen 40002, Thailand; (N.K.); (S.S.); (A.T.)
- Correspondence: (C.I.); (N.N.); Tel.: +39-(0321)-660507 (C.I.); +66-6-3635-2491 (N.N.)
| |
Collapse
|
27
|
Bustos G, Ahumada-Castro U, Silva-Pavez E, Puebla A, Lovy A, Cesar Cardenas J. The ER-mitochondria Ca 2+ signaling in cancer progression: Fueling the monster. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 363:49-121. [PMID: 34392932 DOI: 10.1016/bs.ircmb.2021.03.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cancer is a leading cause of death worldwide. All major tumor suppressors and oncogenes are now recognized to have fundamental connections with metabolic pathways. A hallmark feature of cancer cells is a reprogramming of their metabolism even when nutrients are available. Increasing evidence indicates that most cancer cells rely on mitochondrial metabolism to sustain their energetic and biosynthetic demands. Mitochondria are functionally and physically coupled to the endoplasmic reticulum (ER), the major calcium (Ca2+) storage organelle in mammalian cells, through special domains known as mitochondria-ER contact sites (MERCS). In this domain, the release of Ca2+ from the ER is mainly regulated by inositol 1,4,5-trisphosphate (IP3) receptors (IP3Rs), a family of Ca2+ release channels activated by the ligand IP3. IP3R mediated Ca2+ release is transferred to mitochondria through the mitochondrial Ca2+ uniporter (MCU). Once in the mitochondrial matrix, Ca2+ activates several proteins that stimulate mitochondrial performance. The role of IP3R and MCU in cancer, as well as the other proteins that enable the Ca2+ communication between these two organelles is just beginning to be understood. Here, we describe the function of the main players of the ER mitochondrial Ca2+ communication and discuss how this particular signal may contribute to the rise and development of cancer traits.
Collapse
Affiliation(s)
- Galdo Bustos
- Faculty of Sciences, Universidad Mayor, Center for Integrative Biology, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Ulises Ahumada-Castro
- Faculty of Sciences, Universidad Mayor, Center for Integrative Biology, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Eduardo Silva-Pavez
- Faculty of Sciences, Universidad Mayor, Center for Integrative Biology, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Andrea Puebla
- Faculty of Sciences, Universidad Mayor, Center for Integrative Biology, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Alenka Lovy
- Faculty of Sciences, Universidad Mayor, Center for Integrative Biology, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile; Department of Neuroscience, Center for Neuroscience Research, Tufts School of Medicine, Boston, MA, United States.
| | - J Cesar Cardenas
- Faculty of Sciences, Universidad Mayor, Center for Integrative Biology, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile; Buck Institute for Research on Aging, Novato, CA, United States; Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA, United States.
| |
Collapse
|
28
|
Jin C, Kumar P, Gracia-Sancho J, Dufour JF. Calcium transfer between endoplasmic reticulum and mitochondria in liver diseases. FEBS Lett 2021; 595:1411-1421. [PMID: 33752262 DOI: 10.1002/1873-3468.14078] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/21/2021] [Accepted: 02/25/2021] [Indexed: 01/07/2023]
Abstract
Calcium (Ca2+ ) is a second messenger essential for cellular homeostasis. Inside the cell, Ca2+ is compartmentalized and exchanged among organelles in response to both external and internal stimuli. Mitochondria-associated membranes (MAMs) provide a platform for proteins and channels involved in Ca2+ transfer between the endoplasmic reticulum (ER) and mitochondria. Deregulated Ca2+ signaling and proteins regulating ER-mitochondria interactions have been linked to liver diseases and intensively investigated in recent years. In this review, we summarize the role of MAM-resident proteins in Ca2+ transfer and their association with different liver diseases.
Collapse
Affiliation(s)
- Chaonan Jin
- Hepatology, Department for BioMedical Research, University of Bern, Switzerland
| | - Pavitra Kumar
- Hepatology, Department for BioMedical Research, University of Bern, Switzerland
| | - Jordi Gracia-Sancho
- Hepatology, Department for BioMedical Research, University of Bern, Switzerland.,Liver Vascular Biology Research Group, CIBEREHD, IDIBAPS Research Institute, Barcelona, Spain
| | - Jean-François Dufour
- Hepatology, Department for BioMedical Research, University of Bern, Switzerland.,University Clinic of Visceral Surgery and Medicine, Inselspital, Bern, Switzerland
| |
Collapse
|
29
|
Zhang M, Wang L, Yue Y, Zhang L, Liu T, Jing M, Liang X, Ma M, Xu S, Wang K, Wang X, Fan J. ITPR3 facilitates tumor growth, metastasis and stemness by inducing the NF-ĸB/CD44 pathway in urinary bladder carcinoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:65. [PMID: 33573671 PMCID: PMC7877014 DOI: 10.1186/s13046-021-01866-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 02/02/2021] [Indexed: 12/13/2022]
Abstract
Background Bladder carcinoma is one of the most common urological cancers. ITPR3, as a ubiquitous endoplasmic reticulum calcium channel protein, was reported to be involved in the development and progression of various types of cancer. However, the potential roles and molecular mechanism of ITPR3 in bladder cancer are still unclear. Herein, we elucidated a novel role of ITPR3 in regulating the proliferation, metastasis, and stemness of bladder cancer cells. Methods The expression of ITPR3 in bladder cancer was analyzed using public databases and bladder cancer tissue microarrays. To demonstrate the role of ITPR3 in regulating the NF-ĸB/CD44 pathway and the progression of bladder cancer, a series of molecular biology and biochemistry methods was performed on clinical tissues, along with in vivo and in vitro experiments. The methods used included western blot assay, quantitative RT-PCR assay, immunofluorescence assay, immunohistochemistry (IHC) assays, wound healing assay, Transwell assay, colony formation assay, tumorsphere formation assay, cell flow cytometry analysis, EdU assay, MTT assay, cell transfection, bisulfite sequencing PCR (BSP), a xenograft tumor model and a tail vein cancer metastasis model. Results Higher ITPR3 expression was found in bladder cancer tissues and bladder cancer cells compared with the corresponding normal peritumor tissues and SV-HUC-1 cells, which was attributed to demethylation in the ITPR3 promoter region. ITPR3 promoted the proliferation of bladder cancer by accelerating cell cycle transformation and promoted local invasion and distant metastasis by inducing epithelial-to-mesenchymal transition (EMT). Meanwhile, ITPR3 maintained the cancer stemness phenotype by regulating CD44 expression. NF-κB, which is upstream of CD44, also played a critical role in this process. Conclusions Our study clarifies that ITPR3 serves as an oncogene in bladder cancer cells and represents a novel candidate for bladder cancer diagnosis and treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-01866-1.
Collapse
Affiliation(s)
- Mengzhao Zhang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, #277 Yanta West Road, Xi'an, 710061, China
| | - Lu Wang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, #277 Yanta West Road, Xi'an, 710061, China
| | - Yangyang Yue
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lu Zhang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, #277 Yanta West Road, Xi'an, 710061, China
| | - Tianjie Liu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, #277 Yanta West Road, Xi'an, 710061, China
| | - Minxuan Jing
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, #277 Yanta West Road, Xi'an, 710061, China
| | - Xiao Liang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, #277 Yanta West Road, Xi'an, 710061, China
| | - Minghai Ma
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, #277 Yanta West Road, Xi'an, 710061, China
| | - Shan Xu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, #277 Yanta West Road, Xi'an, 710061, China
| | - Ke Wang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, #277 Yanta West Road, Xi'an, 710061, China
| | - Xinyang Wang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, #277 Yanta West Road, Xi'an, 710061, China
| | - Jinhai Fan
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, #277 Yanta West Road, Xi'an, 710061, China. .,Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of education, Xi'an, China.
| |
Collapse
|
30
|
Alharbi A, Zhang Y, Parrington J. Deciphering the Role of Ca 2+ Signalling in Cancer Metastasis: From the Bench to the Bedside. Cancers (Basel) 2021; 13:E179. [PMID: 33430230 PMCID: PMC7825727 DOI: 10.3390/cancers13020179] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/01/2021] [Accepted: 01/03/2021] [Indexed: 01/03/2023] Open
Abstract
Metastatic cancer is one of the major causes of cancer-related mortalities. Metastasis is a complex, multi-process phenomenon, and a hallmark of cancer. Calcium (Ca2+) is a ubiquitous secondary messenger, and it has become evident that Ca2+ signalling plays a vital role in cancer. Ca2+ homeostasis is dysregulated in physiological processes related to tumour metastasis and progression-including cellular adhesion, epithelial-mesenchymal transition, cell migration, motility, and invasion. In this review, we looked at the role of intracellular and extracellular Ca2+ signalling pathways in processes that contribute to metastasis at the local level and also their effects on cancer metastasis globally, as well as at underlying molecular mechanisms and clinical applications. Spatiotemporal Ca2+ homeostasis, in terms of oscillations or waves, is crucial for hindering tumour progression and metastasis. They are a limited number of clinical trials investigating treating patients with advanced stages of various cancer types. Ca2+ signalling may serve as a novel hallmark of cancer due to the versatility of Ca2+ signals in cells, which suggests that the modulation of specific upstream/downstream targets may be a therapeutic approach to treat cancer, particularly in patients with metastatic cancers.
Collapse
Affiliation(s)
- Abeer Alharbi
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK;
- Pharmaceutical Sciences Department, College of Pharmacy, King Saud Bin Abdul-Aziz University for Health Sciences, Riyadh 11426, Saudi Arabia
| | - Yuxuan Zhang
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK;
| | - John Parrington
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK;
| |
Collapse
|
31
|
Parys JB, Bultynck G, Vervliet T. IP 3 Receptor Biology and Endoplasmic Reticulum Calcium Dynamics in Cancer. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2021; 59:215-237. [PMID: 34050869 DOI: 10.1007/978-3-030-67696-4_11] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Intracellular Ca2+ signaling regulates a plethora of cellular functions. A central role in these processes is reserved for the inositol 1,4,5-trisphosphate receptor (IP3R), a ubiquitously expressed Ca2+-release channel, mainly located in the endoplasmic reticulum (ER). Three IP3R isoforms (IP3R1, IP3R2 and IP3R3) exist, encoded respectively by ITPR1, ITPR2 and ITPR3. The proteins encoded by these genes are each about 2700 amino acids long and assemble into large tetrameric channels, which form the target of many regulatory proteins, including several tumor suppressors and oncogenes. Due to the important role of the IP3Rs in cell function, their dysregulation is linked to multiple pathologies. In this review, we highlight the complex role of the IP3R in cancer, as it participates in most of the so-called "hallmarks of cancer". In particular, the IP3R directly controls cell death and cell survival decisions via regulation of autophagy and apoptosis. Moreover, the IP3R impacts cellular proliferation, migration and invasion. Typical examples of the role of the IP3Rs in these various processes are discussed. The relative levels of the IP3R isoforms expressed and their subcellular localization, e.g. at the ER-mitochondrial interface, is hereby important. Finally, evidence is provided about how the knowledge of the regulation of the IP3R by tumor suppressors and oncogenes can be exploited to develop novel therapeutic approaches to fight cancer.
Collapse
Affiliation(s)
- Jan B Parys
- Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Cancer Institute, KU Leuven, Leuven, Belgium.
| | - Geert Bultynck
- Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Tim Vervliet
- Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| |
Collapse
|
32
|
Genovese I, Vezzani B, Danese A, Modesti L, Vitto VAM, Corazzi V, Pelucchi S, Pinton P, Giorgi C. Mitochondria as the decision makers for cancer cell fate: from signaling pathways to therapeutic strategies. Cell Calcium 2020; 92:102308. [PMID: 33096320 DOI: 10.1016/j.ceca.2020.102308] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/05/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023]
Abstract
As pivotal players in cellular metabolism, mitochondria have a double-faceted role in the final decision of cell fate. This is true for all cell types, but it is even more important and intriguing in the cancer setting. Mitochondria regulate cell fate in many diverse ways: through metabolism, by producing ATP and other metabolites deemed vital or detrimental for cancer cells; through the regulation of Ca2+ homeostasis, especially by the joint participation of the endoplasmic reticulum in a membranous tethering system for Ca2+ signaling called mitochondria-ER associated membranes (MAMs); and by regulating signaling pathways involved in the survival of cancer cells such as mitophagy. Recent studies have shown that mitochondria can also play a role in the regulation of inflammatory pathways in cancer cells, for example, through the release of mitochondrial DNA (mtDNA) involved in the activation of the cGAS-cGAMP-STING pathway. In this review, we aim to explore the role of mitochondria as decision makers in fostering cancer cell death or survival depending on the tumor cell stage and describe novel anticancer therapeutic strategies targeting mitochondria.
Collapse
Affiliation(s)
- Ilaria Genovese
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Bianca Vezzani
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Alberto Danese
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Lorenzo Modesti
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Veronica Angela Maria Vitto
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Virginia Corazzi
- ENT & Audiology Department, University Hospital of Ferrara, Ferrara, Italy
| | - Stefano Pelucchi
- ENT & Audiology Department, University Hospital of Ferrara, Ferrara, Italy
| | - Paolo Pinton
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Carlotta Giorgi
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy.
| |
Collapse
|
33
|
Loncke J, Kerkhofs M, Kaasik A, Bezprozvanny I, Bultynck G. Recent advances in understanding IP3R function with focus on ER-mitochondrial Ca2+ transfers. CURRENT OPINION IN PHYSIOLOGY 2020. [DOI: 10.1016/j.cophys.2020.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
34
|
Lemos FO, Guerra MT, Leite MDF. Inositol 1,4,5 trisphosphate receptors in secretory epithelial cells of the gastrointestinal tract. CURRENT OPINION IN PHYSIOLOGY 2020. [DOI: 10.1016/j.cophys.2020.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
35
|
Ali ES, Rychkov GY, Barritt GJ. Targeting Ca 2+ Signaling in the Initiation, Promotion and Progression of Hepatocellular Carcinoma. Cancers (Basel) 2020; 12:cancers12102755. [PMID: 32987945 PMCID: PMC7600741 DOI: 10.3390/cancers12102755] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/20/2020] [Accepted: 09/21/2020] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Liver cancer (hepatocellular carcinoma) is a significant health burden worldwide. It is often not detected until at an advanced stage when there are few treatment options available. Changes in calcium concentrations within liver cancer cells are essential for regulating their growth, death, and migration (metastasis). Our aim was to review published papers which have identified proteins involved in calcium signaling as potential drug targets for the treatment of liver cancer. About twenty calcium signaling proteins were identified, including those involved in regulating calcium concentrations in the cytoplasm, endoplasmic reticulum and mitochondria. A few of these have turned out to be sites of action of natural products previously known to inhibit liver cancer. More systematic studies are now needed to determine which calcium signaling proteins might be used clinically for treatment of liver cancer, especially advanced stage cancers and those resistant to inhibition by current drugs. Abstract Hepatocellular carcinoma (HCC) is a considerable health burden worldwide and a major contributor to cancer-related deaths. HCC is often not noticed until at an advanced stage where treatment options are limited and current systemic drugs can usually only prolong survival for a short time. Understanding the biology and pathology of HCC is a challenge, due to the cellular and anatomic complexities of the liver. While not yet fully understood, liver cancer stem cells play a central role in the initiation and progression of HCC and in resistance to drugs. There are approximately twenty Ca2+-signaling proteins identified as potential targets for therapeutic treatment at different stages of HCC. These potential targets include inhibition of the self-renewal properties of liver cancer stem cells; HCC initiation and promotion by hepatitis B and C and non-alcoholic fatty liver disease (principally involving reduction of reactive oxygen species); and cell proliferation, tumor growth, migration and metastasis. A few of these Ca2+-signaling pathways have been identified as targets for natural products previously known to reduce HCC. Promising Ca2+-signaling targets include voltage-operated Ca2+ channel proteins (liver cancer stem cells), inositol trisphosphate receptors, store-operated Ca2+ entry, TRP channels, sarco/endoplasmic reticulum (Ca2++Mg2+) ATP-ase and Ca2+/calmodulin-dependent protein kinases. However, none of these Ca2+-signaling targets has been seriously studied any further than laboratory research experiments. The future application of more systematic studies, including genomics, gene expression (RNA-seq), and improved knowledge of the fundamental biology and pathology of HCC will likely reveal new Ca2+-signaling protein targets and consolidate priorities for those already identified.
Collapse
Affiliation(s)
- Eunus S. Ali
- Department of Medical Biochemistry, College of Medicine and Public Health, Flinders University, Adelaide 5001, South Australia, Australia;
| | - Grigori Y. Rychkov
- School of Medicine, The University of Adelaide, Adelaide 5005, South Australia, Australia;
- South Australian Health and Medical Research Institute, Adelaide 5005, South Australia, Australia
| | - Greg J. Barritt
- Department of Medical Biochemistry, College of Medicine and Public Health, Flinders University, Adelaide 5001, South Australia, Australia;
- Correspondence: ; Tel.: +61-438-204-426
| |
Collapse
|
36
|
Marchi S, Giorgi C, Galluzzi L, Pinton P. Ca 2+ Fluxes and Cancer. Mol Cell 2020; 78:1055-1069. [PMID: 32559424 DOI: 10.1016/j.molcel.2020.04.017] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/14/2020] [Accepted: 04/15/2020] [Indexed: 02/06/2023]
Abstract
Ca2+ ions are key second messengers in both excitable and non-excitable cells. Owing to the rather pleiotropic nature of Ca2+ transporters and other Ca2+-binding proteins, however, Ca2+ signaling has attracted limited attention as a potential target of anticancer therapy. Here, we discuss cancer-associated alterations of Ca2+ fluxes at specific organelles as we identify novel candidates for the development of drugs that selectively target Ca2+ signaling in malignant cells.
Collapse
Affiliation(s)
- Saverio Marchi
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - Carlotta Giorgi
- Department of Medical Sciences, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA; Sandra and Edward Meyer Cancer Center, New York, NY, USA; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA; Department of Dermatology, Yale School of Medicine, New Haven, CT, USA; Université de Paris, Paris, France.
| | - Paolo Pinton
- Department of Medical Sciences, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy.
| |
Collapse
|
37
|
Pascual-Caro C, Orantos-Aguilera Y, Sanchez-Lopez I, de Juan-Sanz J, Parys JB, Area-Gomez E, Pozo-Guisado E, Martin-Romero FJ. STIM1 Deficiency Leads to Specific Down-Regulation of ITPR3 in SH-SY5Y Cells. Int J Mol Sci 2020; 21:ijms21186598. [PMID: 32916960 PMCID: PMC7555297 DOI: 10.3390/ijms21186598] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/05/2020] [Accepted: 09/07/2020] [Indexed: 12/19/2022] Open
Abstract
STIM1 is an endoplasmic reticulum (ER) protein that modulates the activity of a number of Ca2+ transport systems. By direct physical interaction with ORAI1, a plasma membrane Ca2+ channel, STIM1 activates the ICRAC current, whereas the binding with the voltage-operated Ca2+ channel CaV1.2 inhibits the current through this latter channel. In this way, STIM1 is a key regulator of Ca2+ signaling in excitable and non-excitable cells, and altered STIM1 levels have been reported to underlie several pathologies, including immunodeficiency, neurodegenerative diseases, and cancer. In both sporadic and familial Alzheimer’s disease, a decrease of STIM1 protein levels accounts for the alteration of Ca2+ handling that compromises neuronal cell viability. Using SH-SY5Y cells edited by CRISPR/Cas9 to knockout STIM1 gene expression, this work evaluated the molecular mechanisms underlying the cell death triggered by the deficiency of STIM1, demonstrating that STIM1 is a positive regulator of ITPR3 gene expression. ITPR3 (or IP3R3) is a Ca2+ channel enriched at ER-mitochondria contact sites where it provides Ca2+ for transport into the mitochondria. Thus, STIM1 deficiency leads to a strong reduction of ITPR3 transcript and ITPR3 protein levels, a consequent decrease of the mitochondria free Ca2+ concentration ([Ca2+]mit), reduction of mitochondrial oxygen consumption rate, and decrease in ATP synthesis rate. All these values were normalized by ectopic expression of ITPR3 in STIM1-KO cells, providing strong evidence for a new mode of regulation of [Ca2+]mit mediated by the STIM1-ITPR3 axis.
Collapse
Affiliation(s)
- Carlos Pascual-Caro
- Department of Biochemistry and Molecular Biology, School of Life Sciences and Institute of Molecular Pathology and Biomarkers, University of Extremadura, 06006 Badajoz, Spain; (C.P.-C.); (Y.O.-A.); (I.S.-L.)
| | - Yolanda Orantos-Aguilera
- Department of Biochemistry and Molecular Biology, School of Life Sciences and Institute of Molecular Pathology and Biomarkers, University of Extremadura, 06006 Badajoz, Spain; (C.P.-C.); (Y.O.-A.); (I.S.-L.)
| | - Irene Sanchez-Lopez
- Department of Biochemistry and Molecular Biology, School of Life Sciences and Institute of Molecular Pathology and Biomarkers, University of Extremadura, 06006 Badajoz, Spain; (C.P.-C.); (Y.O.-A.); (I.S.-L.)
| | - Jaime de Juan-Sanz
- Sorbonne Universités and Institut du Cerveau et de la Moelle Epinière (ICM) - Hôpital Pitié-Salpêtrière, Inserm, CNRS, 75013 Paris, France;
| | - Jan B. Parys
- Department of Cellular and Molecular Medicine, Leuven Kanker Instituut, KU Leuven, B-3000 Leuven, Belgium;
| | - Estela Area-Gomez
- Department of Neurology, Columbia University Medical Center, New York, NY 10032-3748, USA;
| | - Eulalia Pozo-Guisado
- Department of Cell Biology, School of Medicine and Institute of Molecular Pathology and Biomarkers, University of Extremadura, 06006 Badajoz, Spain;
| | - Francisco Javier Martin-Romero
- Department of Biochemistry and Molecular Biology, School of Life Sciences and Institute of Molecular Pathology and Biomarkers, University of Extremadura, 06006 Badajoz, Spain; (C.P.-C.); (Y.O.-A.); (I.S.-L.)
- Correspondence: ; Tel.: +34-924-489-971
| |
Collapse
|
38
|
Lima Filho ACM, França A, Florentino RM, Dos Santos ML, de Oliveira Lemos F, Missiaggia DG, Fonseca RC, Gustavo Oliveira A, Ananthanarayanan M, Guerra MT, de Castro Fonseca M, Vidigal PVT, Lima CX, Nathanson MH, Fatima Leite M. Inositol 1,4,5-trisphosphate receptor type 3 plays a protective role in hepatocytes during hepatic ischemia-reperfusion injury. Cell Calcium 2020; 91:102264. [PMID: 32957029 DOI: 10.1016/j.ceca.2020.102264] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/05/2020] [Accepted: 08/05/2020] [Indexed: 01/02/2023]
Abstract
Hepatic ischemia-reperfusion injury is seen in a variety of clinical conditions, including hepatic thrombosis, systemic hypotension, and liver transplantation. Calcium (Ca2+) signaling mediates several pathophysiological processes in the liver, but it is not known whether and how intracellular Ca2+ channels are involved in the hepatocellular events secondary to ischemia-reperfusion. Using an animal model of hepatic ischemia-reperfusion injury, we observed a progressive increase in expression of the type 3 isoform of the inositol trisphosphate receptor (ITPR3), an intracellular Ca2+ channel that is not normally expressed in healthy hepatocytes. ITPR3 expression was upregulated, at least in part, by a combination of demethylation of the ITPR3 promoter region and the increased transcriptional activity of the nuclear factor of activated T-cells (NFAT). Additionally, expression of pro-inflammatory interleukins and necrotic surface area were less pronounced in livers of control animals compared to liver-specific ITPR3 KO mice subjected to hepatic damage. Corroborating these findings, ITPR3 expression and activation of NFAT were observed in hepatocytes of liver biopsies from patients who underwent liver ischemia caused by thrombosis after organ transplant. Together, these results are consistent with the idea that ITPR3 expression in hepatocytes plays a protective role during hepatic injury induced by ischemia-reperfusion.
Collapse
Affiliation(s)
| | - Andressa França
- Department of Molecular Medicine, Federal University of Minas Gerais (UFMG), MG, Brazil.
| | - Rodrigo M Florentino
- Department of Biophysics and Physiology, Federal University of Minas Gerais (UFMG), MG, Brazil.
| | | | | | | | | | - André Gustavo Oliveira
- Department of Biophysics and Physiology, Federal University of Minas Gerais (UFMG), MG, Brazil.
| | | | - Mateus T Guerra
- Section of Digestive Disease, Department of Internal Medicine, Yale University School of Medicine, CT, United States.
| | - Matheus de Castro Fonseca
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials, SP, Brazil.
| | | | - Cristiano Xavier Lima
- Department of Surgery, Medicine School of Federal University of Minas Gerais (UFMG), MG, United States.
| | - Michael H Nathanson
- Section of Digestive Disease, Department of Internal Medicine, Yale University School of Medicine, CT, United States.
| | - M Fatima Leite
- Department of Biophysics and Physiology, Federal University of Minas Gerais (UFMG), MG, Brazil.
| |
Collapse
|
39
|
Chung C, Seo W, Silwal P, Jo EK. Crosstalks between inflammasome and autophagy in cancer. J Hematol Oncol 2020; 13:100. [PMID: 32703253 PMCID: PMC7376907 DOI: 10.1186/s13045-020-00936-9] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 07/13/2020] [Indexed: 02/07/2023] Open
Abstract
Both inflammasomes and autophagy have important roles in the intracellular homeostasis, inflammation, and pathology; the dysregulation of these processes is often associated with the pathogenesis of numerous cancers. In addition, they can crosstalk with each other in multifaceted ways to influence various physiological and pathological responses, including cancer. Multiple molecular mechanisms connect the autophagy pathway to inflammasome activation and, through this, may influence the outcome of pro-tumor or anti-tumor responses depending on the cancer types, microenvironment, and the disease stage. In this review, we highlight the rapidly growing literature on the various mechanisms by which autophagy interacts with the inflammasome pathway, to encourage additional applications in the context of tumors. In addition, we provide insight into the mechanisms by which pathogen modulates the autophagy-inflammasome pathway to favor the infection-induced carcinogenesis. We also explore the challenges and opportunities of using multiple small molecules/agents to target the autophagy/inflammasome axis and their effects upon cancer treatment. Finally, we discuss the emerging clinical efforts assessing the potential usefulness of targeting approaches for either autophagy or inflammasome as anti-cancer strategies, although it remains underexplored in terms of their crosstalks.
Collapse
Affiliation(s)
- Chaeuk Chung
- Division of Pulmonary and Critical Care, Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, 35015, Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, 35015, Korea
| | - Wonhyoung Seo
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, 35015, Korea.,Department of Microbiology, Chungnam National University School of Medicine, Daejeon, 35015, Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon, 35015, Korea
| | - Prashanta Silwal
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, 35015, Korea.,Department of Microbiology, Chungnam National University School of Medicine, Daejeon, 35015, Korea
| | - Eun-Kyeong Jo
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, 35015, Korea. .,Department of Microbiology, Chungnam National University School of Medicine, Daejeon, 35015, Korea. .,Department of Medical Science, Chungnam National University School of Medicine, Daejeon, 35015, Korea.
| |
Collapse
|
40
|
Lemos FDO, França A, Lima Filho ACM, Florentino RM, Santos ML, Missiaggia DG, Rodrigues GOL, Dias FF, Souza Passos IB, Teixeira MM, Andrade AMDF, Lima CX, Vidigal PVT, Costa VV, Fonseca MC, Nathanson MH, Leite MF. Molecular Mechanism for Protection Against Liver Failure in Human Yellow Fever Infection. Hepatol Commun 2020; 4:657-669. [PMID: 32363317 PMCID: PMC7193135 DOI: 10.1002/hep4.1504] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 02/18/2020] [Accepted: 02/20/2020] [Indexed: 12/18/2022] Open
Abstract
Yellow fever (YF) is a viral hemorrhagic fever that typically involves the liver. Brazil recently experienced its largest recorded YF outbreak, and the disease was fatal in more than a third of affected individuals, mostly because of acute liver failure. Affected individuals are generally treated only supportively, but during the recent Brazilian outbreak, selected patients were treated with liver transplant. We took advantage of this clinical experience to better characterize the clinical and pathological features of YF-induced liver failure and to examine the mechanism of hepatocellular injury in YF, to identify targets that would be amenable to therapeutic intervention in preventing progression to liver failure and death. Patients with YF liver failure rapidly developed massive transaminase elevations, with jaundice, coagulopathy, thrombocytopenia, and usually hepatic encephalopathy, along with pathological findings that included microvesicular steatosis and lytic necrosis. Hepatocytes began to express the type 3 isoform of the inositol trisphosphate receptor (ITPR3), an intracellular calcium (Ca2+) channel that is not normally expressed in hepatocytes. Experiments in an animal model, isolated hepatocytes, and liver-derived cell lines showed that this new expression of ITPR3 was associated with increased nuclear Ca2+ signaling and hepatocyte proliferation, and reduced steatosis and cell death induced by the YF virus. Conclusion: Yellow fever often induces liver failure characterized by massive hepatocellular damage plus steatosis. New expression of ITPR3 also occurs in YF-infected hepatocytes, which may represent an endogenous protective mechanism that could suggest approaches to treat affected individuals before they progress to liver failure, thereby decreasing the mortality of this disease in a way that does not rely on the costly and limited resource of liver transplantation.
Collapse
Affiliation(s)
| | - Andressa França
- Department of Physiology and BiophysicsUniversidade Federal de Minas GeraisBelo HorizonteBrazil
| | | | - Rodrigo M. Florentino
- Department of Physiology and BiophysicsUniversidade Federal de Minas GeraisBelo HorizonteBrazil
| | - Marcone Loiola Santos
- Department of Physiology and BiophysicsUniversidade Federal de Minas GeraisBelo HorizonteBrazil
| | - Dabny G. Missiaggia
- Department of Physiology and BiophysicsUniversidade Federal de Minas GeraisBelo HorizonteBrazil
| | | | - Felipe Ferraz Dias
- Center of MicroscopyUniversidade Federal de Minas GeraisBelo HorizonteBrazil
| | | | - Mauro M. Teixeira
- Department of Biochemistry and ImmunologyUniversidade Federal de Minas GeraisBelo HorizonteBrazil
| | | | - Cristiano Xavier Lima
- Hepatic Transplant ServiceHospital Felício RochoBelo HorizonteBrazil
- SurgeryUniversidade Federal de Minas GeraisBelo HorizonteBrazil
| | | | | | - Matheus Castro Fonseca
- Brazilian Biosciences National Laboratory (LNBio)Brazilian Center for Research in Energy and MaterialsRua Giuseppe Máximo ScolfaroCampinasBrazil
| | - Michael H. Nathanson
- Section of Digestive DiseasesDepartment of Internal MedicineYale University School of MedicineNew HavenCT
| | - M. Fatima Leite
- Department of Physiology and BiophysicsUniversidade Federal de Minas GeraisBelo HorizonteBrazil
| |
Collapse
|
41
|
Inositol 1,4,5-Trisphosphate Receptors in Human Disease: A Comprehensive Update. J Clin Med 2020; 9:jcm9041096. [PMID: 32290556 PMCID: PMC7231134 DOI: 10.3390/jcm9041096] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 03/30/2020] [Accepted: 04/10/2020] [Indexed: 12/22/2022] Open
Abstract
Inositol 1,4,5-trisphosphate receptors (ITPRs) are intracellular calcium release channels located on the endoplasmic reticulum of virtually every cell. Herein, we are reporting an updated systematic summary of the current knowledge on the functional role of ITPRs in human disorders. Specifically, we are describing the involvement of its loss-of-function and gain-of-function mutations in the pathogenesis of neurological, immunological, cardiovascular, and neoplastic human disease. Recent results from genome-wide association studies are also discussed.
Collapse
|
42
|
Rosa N, Sneyers F, Parys JB, Bultynck G. Type 3 IP 3 receptors: The chameleon in cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 351:101-148. [PMID: 32247578 DOI: 10.1016/bs.ircmb.2020.02.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Inositol 1,4,5-trisphosphate (IP3) receptors (IP3Rs), intracellular calcium (Ca2+) release channels, fulfill key functions in cell death and survival processes, whose dysregulation contributes to oncogenesis. This is essentially due to the presence of IP3Rs in microdomains of the endoplasmic reticulum (ER) in close proximity to the mitochondria. As such, IP3Rs enable efficient Ca2+ transfers from the ER to the mitochondria, thus regulating metabolism and cell fate. This review focuses on one of the three IP3R isoforms, the type 3 IP3R (IP3R3), which is linked to proapoptotic ER-mitochondrial Ca2+ transfers. Alterations in IP3R3 expression have been highlighted in numerous cancer types, leading to dysregulations of Ca2+ signaling and cellular functions. However, the outcome of IP3R3-mediated Ca2+ transfers for mitochondrial function is complex with opposing effects on oncogenesis. IP3R3 can either suppress cancer by promoting cell death and cellular senescence or support cancer by driving metabolism, anabolic processes, cell cycle progression, proliferation and invasion. The aim of this review is to provide an overview of IP3R3 dysregulations in cancer and describe how such dysregulations alter critical cellular processes such as proliferation or cell death and survival. Here, we pose that the IP3R3 isoform is not only linked to proapoptotic ER-mitochondrial Ca2+ transfers but might also be involved in prosurvival signaling.
Collapse
Affiliation(s)
- Nicolas Rosa
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut (LKI), Leuven, Belgium
| | - Flore Sneyers
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut (LKI), Leuven, Belgium
| | - Jan B Parys
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut (LKI), Leuven, Belgium
| | - Geert Bultynck
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut (LKI), Leuven, Belgium.
| |
Collapse
|
43
|
Danese A, Marchi S, Vitto VAM, Modesti L, Leo S, Wieckowski MR, Giorgi C, Pinton P. Cancer-Related Increases and Decreases in Calcium Signaling at the Endoplasmic Reticulum-Mitochondria Interface (MAMs). Rev Physiol Biochem Pharmacol 2020; 185:153-193. [PMID: 32789789 DOI: 10.1007/112_2020_43] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Endoplasmic reticulum (ER)-mitochondria regions are specialized subdomains called also mitochondria-associated membranes (MAMs). MAMs allow regulation of lipid synthesis and represent hubs for ion and metabolite signaling. As these two organelles can module both the amplitude and the spatiotemporal patterns of calcium (Ca2+) signals, this particular interaction controls several Ca2+-dependent pathways well known for their contribution to tumorigenesis, such as metabolism, survival, sensitivity to cell death, and metastasis. Mitochondria-mediated apoptosis arises from mitochondrial Ca2+ overload, permeabilization of the mitochondrial outer membrane, and the release of mitochondrial apoptotic factors into the cytosol. Decreases in Ca2+ signaling at the ER-mitochondria interface are being studied in depth as failure of apoptotic-dependent cell death is one of the predominant characteristics of cancer cells. However, some recent papers that linked MAMs Ca2+ crosstalk-related upregulation to tumor onset and progression have aroused the interest of the scientific community.In this review, we will describe how different MAMs-localized proteins modulate the effectiveness of Ca2+-dependent apoptotic stimuli by causing both increases and decreases in the ER-mitochondria interplay and, specifically, by modulating Ca2+ signaling.
Collapse
Affiliation(s)
- Alberto Danese
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Saverio Marchi
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - Veronica Angela Maria Vitto
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Lorenzo Modesti
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Sara Leo
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Mariusz R Wieckowski
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Carlotta Giorgi
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Paolo Pinton
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy.
| |
Collapse
|
44
|
Lemos FDO, Florentino RM, Lima Filho ACM, Santos MLD, Leite MF. Inositol 1,4,5-trisphosphate receptor in the liver: Expression and function. World J Gastroenterol 2019; 25:6483-6494. [PMID: 31802829 PMCID: PMC6886013 DOI: 10.3748/wjg.v25.i44.6483] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 10/22/2019] [Accepted: 11/13/2019] [Indexed: 02/06/2023] Open
Abstract
The liver is a complex organ that performs several functions to maintain homeostasis. These functions are modulated by calcium, a second messenger that regulates several intracellular events. In hepatocytes and cholangiocytes, which are the epithelial cell types in the liver, inositol 1,4,5-trisphosphate (InsP3) receptors (ITPR) are the only intracellular calcium release channels. Three isoforms of the ITPR have been described, named type 1, type 2 and type 3. These ITPR isoforms are differentially expressed in liver cells where they regulate distinct physiological functions. Changes in the expression level of these receptors correlate with several liver diseases and hepatic dysfunctions. In this review, we highlight how the expression level, modulation, and localization of ITPR isoforms in hepatocytes and cholangiocytes play a role in hepatic homeostasis and liver pathology.
Collapse
Affiliation(s)
- Fernanda de Oliveira Lemos
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Rodrigo M Florentino
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Antônio Carlos Melo Lima Filho
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Marcone Loiola dos Santos
- Department of Cell Biology, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - M Fatima Leite
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| |
Collapse
|
45
|
Type 3 inositol 1,4,5-trisphosphate receptor: A calcium channel for all seasons. Cell Calcium 2019; 85:102132. [PMID: 31790953 DOI: 10.1016/j.ceca.2019.102132] [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: 10/17/2019] [Revised: 11/21/2019] [Accepted: 11/22/2019] [Indexed: 12/18/2022]
Abstract
Inositol 1,4,5 trisphosphate receptors (ITPRs) are a family of endoplasmic reticulum Ca2+ channels essential for the control of intracellular Ca2+ levels in virtually every mammalian cell type. The three isoforms (ITPR1, ITPR2 and ITPR3) are highly homologous in amino acid sequence, but they differ considerably in terms of biophysical properties, subcellular localization, and tissue distribution. Such differences underscore the variety of cellular responses triggered by each isoform and suggest that the expression/activity of specific isoforms might be linked to particular pathophysiological states. Indeed, recent findings demonstrate that changes in expression of ITPR isoforms are associated with a number of human diseases ranging from fatty liver disease to cancer. ITPR3 is emerging as the isoform that is particularly important in the pathogenesis of various human diseases. Here we review the physiological and pathophysiological roles of ITPR3 in various tissues and the mechanisms by which the expression of this isoform is modulated in health and disease.
Collapse
|
46
|
de Miranda MC, Rodrigues MA, de Angelis Campos AC, Faria JAQA, Kunrath-Lima M, Mignery GA, Schechtman D, Goes AM, Nathanson MH, Gomes DA. Epidermal growth factor (EGF) triggers nuclear calcium signaling through the intranuclear phospholipase Cδ-4 (PLCδ4). J Biol Chem 2019; 294:16650-16662. [PMID: 31537645 DOI: 10.1074/jbc.ra118.006961] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 09/10/2019] [Indexed: 02/06/2023] Open
Abstract
Calcium (Ca2+) signaling within the cell nucleus regulates specific cellular events such as gene transcription and cell proliferation. Nuclear and cytosolic Ca2+ levels can be independently regulated, and nuclear translocation of receptor tyrosine kinases (RTKs) is one way to locally activate signaling cascades within the nucleus. Nuclear RTKs, including the epidermal growth factor receptor (EGFR), are important for processes such as transcriptional regulation, DNA-damage repair, and cancer therapy resistance. RTKs can hydrolyze phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) within the nucleus, leading to Ca2+ release from the nucleoplasmic reticulum by inositol 1,4,5-trisphosphate receptors. PI(4,5)P2 hydrolysis is mediated by phospholipase C (PLC). However, it is unknown which nuclear PLC isoform is triggered by EGFR. Here, using subcellular fractionation, immunoblotting and fluorescence, siRNA-based gene knockdowns, and FRET-based biosensor reporter assays, we investigated the role of PLCδ4 in epidermal growth factor (EGF)-induced nuclear Ca2+ signaling and downstream events. We found that EGF-induced Ca2+ signals are inhibited when translocation of EGFR is impaired. Nuclear Ca2+ signals also were reduced by selectively buffering inositol 1,4,5-trisphosphate (InsP3) within the nucleus. EGF induced hydrolysis of nuclear PI(4,5)P2 by the intranuclear PLCδ4, rather than by PLCγ1. Moreover, protein kinase C, a downstream target of EGF, was active in the nucleus of stimulated cells. Furthermore, PLCδ4 and InsP3 modulated cell cycle progression by regulating the expression of cyclins A and B1. These results provide evidence that EGF-induced nuclear signaling is mediated by nuclear PLCδ4 and suggest new therapeutic targets to modulate the proliferative effects of this growth factor.
Collapse
Affiliation(s)
- Marcelo Coutinho de Miranda
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais (UFMG), Av. Antonio Carlos, 6627 Belo Horizonte-MG, 31270-901, Brazil.,Section of Digestive Diseases, Internal Medicine, Yale University, New Haven, Connecticut 06520-8056
| | - Michele Angela Rodrigues
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais (UFMG), Av. Antonio Carlos, 6627 Belo Horizonte-MG, 31270-901, Brazil.,Section of Digestive Diseases, Internal Medicine, Yale University, New Haven, Connecticut 06520-8056
| | - Ana Carolina de Angelis Campos
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais (UFMG), Av. Antonio Carlos, 6627 Belo Horizonte-MG, 31270-901, Brazil.,Section of Digestive Diseases, Internal Medicine, Yale University, New Haven, Connecticut 06520-8056
| | | | - Marianna Kunrath-Lima
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais (UFMG), Av. Antonio Carlos, 6627 Belo Horizonte-MG, 31270-901, Brazil
| | - Gregory A Mignery
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois 60153
| | - Deborah Schechtman
- Department of Biochemistry, University of São Paulo, Av. Professor Lineu Prestes, 748, São Paulo-SP 05508-900, Brazil
| | - Alfredo Miranda Goes
- Department of Pathology, Universidade Federal de Minas Gerais (UFMG), Av. Antonio Carlos, 6627 Belo Horizonte-MG, 31270-901, Brazil
| | - Michael H Nathanson
- Section of Digestive Diseases, Internal Medicine, Yale University, New Haven, Connecticut 06520-8056
| | - Dawidson A Gomes
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais (UFMG), Av. Antonio Carlos, 6627 Belo Horizonte-MG, 31270-901, Brazil .,Section of Digestive Diseases, Internal Medicine, Yale University, New Haven, Connecticut 06520-8056
| |
Collapse
|