1
|
Fu Y, Wang C, Sun W, Kong H, Liang W, Shi T, Li Q, Jia M, Zhao W, Song H. MINT3 promotes STING activation and facilitates antiviral immune responses. Cell Signal 2025; 132:111825. [PMID: 40254147 DOI: 10.1016/j.cellsig.2025.111825] [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: 10/24/2024] [Revised: 04/02/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
Stimulator-of-interferon genes (STING) translocation is the rate-limiting step in the cGAS-STING signaling which detects cytosolic DNA and produces type I interferons. However, the mechanism by which this process is modulated remains to be further clarified. In the present study, we identified munc18-1-interacting protein 3 (MINT3) as a positive regulator of STING signaling. MINT3 promotes type I interferons production induced by herpes simplex virus-1 (HSV-1) infection and ISD or cGAMP stimulation in mouse peritoneal macrophages. Deficiency of Mint3 greatly inhibited STING and IRF3 activation in macrophages. Mint3 knockdown also attenuated STING and IRF3 activation in macrophages, human THP-1 cells, and RAW264.7 cells. Mechanistically, MINT3 interacted with STING, selectively enhanced its K63-linked polyubiquitination and facilitated STING translocation to the Golgi, resulting in the enhancement of the STING and TBK1 interaction. Furthermore, MINT3 also facilitated HSV-1-induced innate antiviral immune responses and impaired HSV-1 replication in vitro and in vivo. Interestingly, we showed that the expression of MINT3 was dramatically elevated during HSV-1 infection, and ISD stimulation in macrophages. Thus, we have revealed a feedback mechanism for the regulation of the cGAS-STING pathway, providing a promising therapeutic target for the treatment of disorders triggered by aberrant STING activity.
Collapse
Affiliation(s)
- Yue Fu
- Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Caiwei Wang
- Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Wenyue Sun
- Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Hongyi Kong
- Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Wenbo Liang
- Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Tongrui Shi
- Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Qizhao Li
- Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Mutian Jia
- Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Wei Zhao
- Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Hui Song
- Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
2
|
Tian X, Wang Z, Yi C, Shi Y, Jia C, Li X, Jiang F, Wang Z. Effects of basal division of posterior pallial amygdala on the motor behaviors in pigeons based on transcriptome analysis. Brain Res Bull 2025; 224:111322. [PMID: 40154927 DOI: 10.1016/j.brainresbull.2025.111322] [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: 10/24/2024] [Revised: 03/13/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
The basal division of posterior pallial amygdala (PoAb) was one important part of the amygdala in birds. PoAb mainly mediated turning behavior. However, the regulating neuromechanisms of PoAb in motor behavior was not clear yet. In this study, we selected septalis lateralis (SL) as the stimulated nucleus because it was closely associated with PoAb and had clear neuroregulatory functions, and we also used unrelated nuclei (entopallium) and unstimulated blank treatment (CK) as controls. We aim to study the neuroregulatory mechanisms of PoAb by investigating the differences of transcriptome level in different groups. A total of 622 differentially expressed genes (DEGs) were obtained from PoAb after comparing the SL stimulating group with the CK control group. GO functional annotation and KEGG pathway enrichment analysis showed that the upregulated 608 DEGs mainly involved energy supply and fluid balance. A total of 345 DEGs were obtained from the PoAb when comparing SL stimulation group and entopallium stimulation group. The upregulated 187 DEGs were mainly involved in cell communication and signal transductions. The study indicated that PoAb may modulate motor behaviour mainly by increasing ATP production and facilitating synaptic transmission, in which genes such as SMAD3, TMED3, GRIA2, HTR1B and SNCG play an important role. We revealed the mechanisms of brain regulation behaviour from gene level, and provided the theoretical foundation for understanding the avian brain.
Collapse
Affiliation(s)
- Xinmao Tian
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; School of Medicine and Health, Harbin Institute of Technology, Harbin, Heilongjiang, 150001, China; Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou, Henan 450000, China.
| | - Zishi Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Chunzhi Yi
- School of Medicine and Health, Harbin Institute of Technology, Harbin, Heilongjiang, 150001, China; Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou, Henan 450000, China.
| | - Yuhua Shi
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Chongchong Jia
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; School of Electrical and Information Engineering, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Xiujuan Li
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Feng Jiang
- School of Artificial Intelligence (School of Future Technology), Nanjing University of Information Science & Technology, Nanjing, Jiangsu, 210000, China.
| | - Zhenlong Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| |
Collapse
|
3
|
Bueno D, Schäfer MK, Wang S, Schmeisser MJ, Methner A. NECAB family of neuronal calcium-binding proteins in health and disease. Neural Regen Res 2025; 20:1236-1243. [PMID: 38934399 PMCID: PMC11624857 DOI: 10.4103/nrr.nrr-d-24-00094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/18/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
The N-terminal EF-hand calcium-binding proteins 1-3 (NECAB1-3) constitute a family of predominantly neuronal proteins characterized by the presence of at least one EF-hand calcium-binding domain and a functionally less well characterized C-terminal antibiotic biosynthesis monooxygenase domain. All three family members were initially discovered due to their interactions with other proteins. NECAB1 associates with synaptotagmin-1, a critical neuronal protein involved in membrane trafficking and synaptic vesicle exocytosis. NECAB2 interacts with predominantly striatal G-protein-coupled receptors, while NECAB3 partners with amyloid-β A4 precursor protein-binding family A members 2 and 3, key regulators of amyloid-β production. This demonstrates the capacity of the family for interactions with various classes of proteins. NECAB proteins exhibit distinct subcellular localizations: NECAB1 is found in the nucleus and cytosol, NECAB2 resides in endosomes and the plasma membrane, and NECAB3 is present in the endoplasmic reticulum and Golgi apparatus. The antibiotic biosynthesis monooxygenase domain, an evolutionarily ancient component, is akin to atypical heme oxygenases in prokaryotes but is not well-characterized in vertebrates. Prokaryotic antibiotic biosynthesis monooxygenase domains typically form dimers, suggesting that calcium-mediated conformational changes in NECAB proteins may induce antibiotic biosynthesis monooxygenase domain dimerization, potentially activating some enzymatic properties. However, the substrate for this enzymatic activity remains uncertain. Alternatively, calcium-mediated conformational changes might influence protein interactions or the subcellular localization of NECAB proteins by controlling the availability of protein-protein interaction domains situated between the EF hands and the antibiotic biosynthesis monooxygenase domain. This review summarizes what is known about genomic organization, tissue expression, intracellular localization, interaction partners, and the physiological and pathophysiological role of the NECAB family.
Collapse
Affiliation(s)
- Diones Bueno
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Michael K.E. Schäfer
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Sudena Wang
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Michael J. Schmeisser
- Institute of Anatomy, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Axel Methner
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
4
|
Nishitani M, Okada H, Nio K, Hayashi T, Terashima T, Iida N, Shimakami T, Takatori H, Honda M, Kaneko S, Sakamoto T, Yamashita T. Mint3 as a Molecular Target Activated in the Early Stage of Hepatocarcinogenesis. Int J Mol Sci 2025; 26:1430. [PMID: 40003897 PMCID: PMC11855386 DOI: 10.3390/ijms26041430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 01/30/2025] [Accepted: 02/02/2025] [Indexed: 02/27/2025] Open
Abstract
Mint3 enhances aerobic ATP production with subsequent nuclear translocation of hypoxia-inducible factor-1 (HIF-1) and activation of angiogenesis-related genes. It remains unclear if and when Mint3 is activated and whether it is involved in hepatocarcinogenesis. We explored the expression of Mint3 in surgically resected hepatocellular carcinoma (HCC) tissues. We evaluated the effects of Mint3 knockdown on spheroid formation capacity and subcutaneous tumor growth in immune-deficient mice. We used Mint3 knockout mice to evaluate the effects of chemically induced HCC development. Mint3 was overexpressed in well-differentiated HCC with the activation of HIF-1 target genes irrespective of the absence of hypervascularization. Mint3 knockdown ameliorated the expression of HIF-1 target genes in patient-derived HCC cell lines and suppressed spheroid formation. Mint3 knockdown further inhibited subcutaneous tumor formation in vivo in immune-deficient mice. Chemical HCC development induced by N-nitrosodiethylamine (DEN) or DEN/CCl4 was dramatically suppressed in Mint3 knockout mice compared to control mice. Mint3 plays a crucial role in early-stage HCC development before hypervascularization by activating HIF-1 target genes before the tumor becomes hypoxic. Mint3 is a molecular target that prevents HCC development in the early stages.
Collapse
Affiliation(s)
- Masaki Nishitani
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8641, Ishikawa, Japan; (M.N.); (H.O.); (K.N.); (T.H.); (T.T.); (N.I.); (T.S.); (H.T.); (M.H.); (S.K.)
| | - Hikari Okada
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8641, Ishikawa, Japan; (M.N.); (H.O.); (K.N.); (T.H.); (T.T.); (N.I.); (T.S.); (H.T.); (M.H.); (S.K.)
| | - Kouki Nio
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8641, Ishikawa, Japan; (M.N.); (H.O.); (K.N.); (T.H.); (T.T.); (N.I.); (T.S.); (H.T.); (M.H.); (S.K.)
| | - Tomoyuki Hayashi
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8641, Ishikawa, Japan; (M.N.); (H.O.); (K.N.); (T.H.); (T.T.); (N.I.); (T.S.); (H.T.); (M.H.); (S.K.)
| | - Takeshi Terashima
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8641, Ishikawa, Japan; (M.N.); (H.O.); (K.N.); (T.H.); (T.T.); (N.I.); (T.S.); (H.T.); (M.H.); (S.K.)
| | - Noriho Iida
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8641, Ishikawa, Japan; (M.N.); (H.O.); (K.N.); (T.H.); (T.T.); (N.I.); (T.S.); (H.T.); (M.H.); (S.K.)
| | - Tetsuro Shimakami
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8641, Ishikawa, Japan; (M.N.); (H.O.); (K.N.); (T.H.); (T.T.); (N.I.); (T.S.); (H.T.); (M.H.); (S.K.)
| | - Hajime Takatori
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8641, Ishikawa, Japan; (M.N.); (H.O.); (K.N.); (T.H.); (T.T.); (N.I.); (T.S.); (H.T.); (M.H.); (S.K.)
| | - Masao Honda
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8641, Ishikawa, Japan; (M.N.); (H.O.); (K.N.); (T.H.); (T.T.); (N.I.); (T.S.); (H.T.); (M.H.); (S.K.)
| | - Shuichi Kaneko
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8641, Ishikawa, Japan; (M.N.); (H.O.); (K.N.); (T.H.); (T.T.); (N.I.); (T.S.); (H.T.); (M.H.); (S.K.)
| | - Takeharu Sakamoto
- Department of Cancer Biology, Institute of Biomedical Science, Kansai Medical University, Hirakata 573-1010, Osaka, Japan
| | - Taro Yamashita
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8641, Ishikawa, Japan; (M.N.); (H.O.); (K.N.); (T.H.); (T.T.); (N.I.); (T.S.); (H.T.); (M.H.); (S.K.)
| |
Collapse
|
5
|
Schaub AM, Gonzalez TL, Dorfman AE, Novoa AG, Hussaini RA, Harakuni PM, Khan MH, Shabani BJ, Swarna A, Wang ET, Chan JL, Williams J, Pisarska MD. A systematic review of genome-wide analyses of methylation changes associated with assisted reproductive technologies in various tissues. Fertil Steril 2024; 121:80-94. [PMID: 37827482 PMCID: PMC11262788 DOI: 10.1016/j.fertnstert.2023.10.007] [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: 06/02/2023] [Revised: 09/29/2023] [Accepted: 10/01/2023] [Indexed: 10/14/2023]
Abstract
IMPORTANCE Because analytic technologies improve, increasing amounts of data on methylation differences between assisted reproductive technology (ART) and unassisted conceptions are available. However, various studies use different tissue types and different populations in their analyses, making data comparison and integration difficult. OBJECTIVE To compare and integrate data on genome-wide analyses of methylation differences due to ART, allowing exposure of overarching themes. EVIDENCE REVIEW All studies undertaking genome-wide analysis of human methylation differences due to ART or infertility in any tissue type across the lifespan were assessed for inclusion. FINDINGS Seventeen studies were identified that met the inclusion criteria. One study assessed trophectoderm biopsies, 2 first-trimester placenta, 1 first-trimester fetal tissue, 2 term placenta, 7 cord blood, 3 newborn dried blood spots, 1 childhood buccal smears, 1 childhood peripheral blood, and 2 adult peripheral blood. Eleven studies compared tissues from in vitro fertilization (IVF) conceptions with those of unassisted conceptions, 4 compared intracytoplasmic sperm injection with unassisted conceptions, 4 compared non-IVF fertility treatment (NIFT) with unassisted conceptions, 4 compared NIFT with IVF, and 5 compared an infertile population (conceiving via various methods) with an unassisted presumably fertile population. In studies assessing placental tissue, 1 gene with potential methylation changes due to IVF when compared with unassisted conceptions was identified by 2 studies. In blood, 11 potential genes with methylation changes due to IVF compared with unassisted conceptions were identified by 2 studies, 1 of which was identified by 3 studies. Three potentially affected genes were identified by 2 studies involving blood between intracytoplasmic sperm injection and unassisted populations. There were no overlapping genes identified in any tissue type between NIFT and unassisted populations, between NIFT and IVF, or the infertility combined population when compared with the unassisted fertile population. CONCLUSIONS Comparing studies is challenging due to differing variables between analyses. However, even in similar tissue types and populations, overlapping methylation changes are limited, suggesting that differences due to ART are minimal. RELEVANCE Information from this systematic review is significant for providers and patients who provide and use ART to understand methylation risks that may be associated with the technology.
Collapse
Affiliation(s)
- Amelia M Schaub
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Cedars Sinai Medical Center, Los Angeles, California
| | - Tania L Gonzalez
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Cedars Sinai Medical Center, Los Angeles, California
| | - Anna E Dorfman
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Cedars Sinai Medical Center, Los Angeles, California
| | - Allynson G Novoa
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Cedars Sinai Medical Center, Los Angeles, California
| | - Rimsha A Hussaini
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Cedars Sinai Medical Center, Los Angeles, California
| | - Paige M Harakuni
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Cedars Sinai Medical Center, Los Angeles, California
| | - Mayaal H Khan
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Cedars Sinai Medical Center, Los Angeles, California
| | - Brandon J Shabani
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Cedars Sinai Medical Center, Los Angeles, California
| | - Akhila Swarna
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Cedars Sinai Medical Center, Los Angeles, California
| | - Erica T Wang
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Cedars Sinai Medical Center, Los Angeles, California
| | - Jessica L Chan
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Cedars Sinai Medical Center, Los Angeles, California
| | - John Williams
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Cedars Sinai Medical Center, Los Angeles, California; David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Margareta D Pisarska
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Cedars Sinai Medical Center, Los Angeles, California; David Geffen School of Medicine at UCLA, Los Angeles, California.
| |
Collapse
|
6
|
Tanaka N, Okada H, Yamaguchi K, Seki M, Matsubara D, Gotoh N, Suzuki Y, Furukawa Y, Yamashita T, Inoue JI, Kaneko S, Sakamoto T. Mint3-depletion-induced energy stress sensitizes triple-negative breast cancer to chemotherapy via HSF1 inactivation. Cell Death Dis 2023; 14:815. [PMID: 38081808 PMCID: PMC10713533 DOI: 10.1038/s41419-023-06352-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 11/23/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023]
Abstract
Given the lack of therapeutic targets, the conventional approach for managing triple-negative breast cancer (TNBC) involves the utilization of cytotoxic chemotherapeutic agents. However, most TNBCs acquire resistance to chemotherapy, thereby lowering the therapeutic outcome. In addition to oncogenic mutations in TNBC, microenvironment-induced mechanisms render chemoresistance more complex and robust in vivo. Here, we aimed to analyze whether depletion of Munc18-1 interacting protein 3 (Mint3), which activates hypoxia-inducible factor 1 (HIF-1) during normoxia, sensitizes TNBC to chemotherapy. We found that Mint3 promotes the chemoresistance of TNBC in vivo. Mint3 depletion did not affect the sensitivity of human TNBC cell lines to doxorubicin and paclitaxel in vitro but sensitized tumors of these cells to chemotherapy in vivo. Transcriptome analyses revealed that the Mint3-HIF-1 axis enhanced heat shock protein 70 (HSP70) expression in tumors of TNBC cells. Administering an HSP70 inhibitor enhanced the antitumor activity of doxorubicin in TNBC tumors, similar to Mint3 depletion. Mint3 expression was also correlated with HSP70 expression in human TNBC specimens. Mechanistically, Mint3 depletion induces glycolytic maladaptation to the tumor microenvironment in TNBC tumors, resulting in energy stress. This energy stress by Mint3 depletion inactivated heat shock factor 1 (HSF-1), the master regulator of HSP expression, via the AMP-activated protein kinase/mechanistic target of the rapamycin pathway following attenuated HSP70 expression. In conclusion, Mint3 is a unique regulator of TNBC chemoresistance in vivo via metabolic adaptation to the tumor microenvironment, and a combination of Mint3 inhibition and chemotherapy may be a good strategy for TNBC treatment.
Collapse
Affiliation(s)
- Noritaka Tanaka
- Department of Cancer Biology, Institute of Biomedical Science, Kansai Medical University, Osaka, Japan
| | - Hikari Okada
- Information-Based Medicine Development, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Kiyoshi Yamaguchi
- Division of Clinical Genome Research, the Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masahide Seki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | | | - Noriko Gotoh
- Division of Cancer Cell Biology, Cancer Research Institute, Kanazawa University, Ishikawa, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Yoichi Furukawa
- Division of Clinical Genome Research, the Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Taro Yamashita
- Department of System Biology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Ishikawa, Japan
| | - Jun-Ichiro Inoue
- The University of Tokyo Pandemic Preparedness, Infection and Advanced Research Center (UTOPIA), Tokyo, Japan
| | - Shuichi Kaneko
- Information-Based Medicine Development, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Takeharu Sakamoto
- Department of Cancer Biology, Institute of Biomedical Science, Kansai Medical University, Osaka, Japan.
- Department of System Biology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Ishikawa, Japan.
| |
Collapse
|
7
|
Tian Y, Shao L, Wang Q, Ru G, Zhai C, Zhou L. NECAB3 promotes the migration and invasion of liver cancer cells through HIF-1α/RIT1 signaling pathway. Open Med (Wars) 2023; 18:20230700. [PMID: 37215053 PMCID: PMC10193407 DOI: 10.1515/med-2023-0700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 03/28/2023] [Accepted: 04/03/2023] [Indexed: 05/24/2023] Open
Abstract
Liver cancer is a prevalent malignant tumor with high mortality worldwide, making it urgent to explore new targets for liver cancer therapy. N-terminal EF-hand calcium binding protein 3 (NECAB3) is a new recognized regulator of cancer, while its role in liver cancer remained elusive. Thus, the study clarified the action of NECAB3 on liver cancer development and explored the detailed mechanism. We found that NECAB3 was enhanced in liver cancer. Knockdown of NECAB3 restrained liver cancer cell migration and invasion. Besides, knockdown of NECAB3 suppressed the activation of the hypoxia-inducible factor 1-alpha (HIF-1α)/Ras like without CAAX 1 (RIT1) pathway. Furthermore, NECAB3 regulated liver cancer migration and invasion through modulating RIT1 expression. Moreover, downregulation of NECAB3 suppressed liver cancer tumor growth in vivo. In conclusion, NECAB3 was upregulated in liver cancer. Knockdown of NECAB3 suppressed aggressive phenotype of liver cancer via modulating the HIF-1α/RIT1 axis, providing a possible target for liver cancer therapy.
Collapse
Affiliation(s)
- Yicheng Tian
- Department of General Surgery, Suzhou Science & Technology Town Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, 215153, China
| | - Longjiang Shao
- Department of General Surgery, Suzhou Science & Technology Town Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, 215153, China
| | - Qi Wang
- Department of General Surgery, Suzhou Science & Technology Town Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, 215153, China
| | - Gan Ru
- Department of General Surgery, Suzhou Science & Technology Town Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, 215153, China
| | - Chuntao Zhai
- Department of General Surgery, Suzhou Science & Technology Town Hospital, Gusu School, Nanjing Medical University, No. 1 Lijiang Road, Huqiu District, Suzhou, Jiangsu, 215153, China
| | - Lihui Zhou
- Department of General Surgery, Suzhou Science & Technology Town Hospital, Gusu School, Nanjing Medical University, No. 1 Lijiang Road, Huqiu District, Suzhou, Jiangsu, 215153, China
| |
Collapse
|
8
|
Tanaka N, Sakamoto T. Mint3 as a Potential Target for Cooling Down HIF-1α-Mediated Inflammation and Cancer Aggressiveness. Biomedicines 2023; 11:biomedicines11020549. [PMID: 36831085 PMCID: PMC9953510 DOI: 10.3390/biomedicines11020549] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/11/2023] [Accepted: 02/12/2023] [Indexed: 02/16/2023] Open
Abstract
Hypoxia-inducible factor-1α (HIF-1α) is a transcription factor that plays a crucial role in cells adapting to a low-oxygen environment by facilitating a switch from oxygen-dependent ATP production to glycolysis. Mediated by membrane type-1 matrix metalloproteinase (MT1-MMP) expression, Munc-18-1 interacting protein 3 (Mint3) binds to the factor inhibiting HIF-1 (FIH-1) and inhibits its suppressive effect, leading to HIF-1α activation. Defects in Mint3 generally lead to improved acute inflammation, which is regulated by HIF-1α and subsequent glycolysis, as well as the suppression of the proliferation and metastasis of cancer cells directly through its expression in cancer cells and indirectly through its expression in macrophages or fibroblasts associated with cancer. Mint3 in inflammatory monocytes enhances the chemotaxis into metastatic sites and the production of vascular endothelial growth factors, which leads to the expression of E-selectin at the metastatic sites and the extravasation of cancer cells. Fibroblasts express L1 cell adhesion molecules in a Mint3-dependent manner and enhance integrin-mediated cancer progression. In pancreatic cancer cells, Mint3 directly promotes cancer progression. Naphthofluorescein, a Mint3 inhibitor, can disrupt the interaction between FIH-1 and Mint3 and potently suppress Mint3-mediated inflammation, cancer progression, and metastasis without causing marked adverse effects. In this review, we will introduce the potential of Mint3 as a therapeutic target for inflammatory diseases and cancers.
Collapse
|
9
|
Qiao X, Ding Y, Wu D, Zhang A, Yin Y, Wang Q, Wang W, Kang J. The roles of long noncoding RNA-mediated macrophage polarization in respiratory diseases. Front Immunol 2023; 13:1110774. [PMID: 36685535 PMCID: PMC9849253 DOI: 10.3389/fimmu.2022.1110774] [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: 11/29/2022] [Accepted: 12/16/2022] [Indexed: 01/06/2023] Open
Abstract
Macrophages play an essential role in maintaining the normal function of the innate and adaptive immune responses during host defence. Macrophages acquire diverse functional phenotypes in response to various microenvironmental stimuli, and are mainly classified into classically activated macrophages (M1) and alternatively activated macrophages (M2). Macrophage polarization participates in the inflammatory, fibrotic, and oncogenic processes of diverse respiratory diseases by changing phenotype and function. In recent decades, with the advent of broad-range profiling methods such as microarrays and next-generation sequencing, the discovery of RNA transcripts that do not encode proteins termed "noncoding RNAs (ncRNAs)" has become more easily accessible. As one major member of the regulatory ncRNA family, long noncoding RNAs (lncRNAs, transcripts >200 nucleotides) participate in multiple pathophysiological processes, including cell proliferation, differentiation, and apoptosis, and vary with different stimulants and cell types. Emerging evidence suggests that lncRNAs account for the regulation of macrophage polarization and subsequent effects on respiratory diseases. In this review, we summarize the current published literature from the PubMed database concerning lncRNAs relevant to macrophage polarization and the underlying molecular mechanisms during the occurrence and development of respiratory diseases. These differentially expressed lncRNAs are expected to be biomarkers and targets for the therapeutic regulation of macrophage polarization during disease development.
Collapse
|
10
|
Kim DH, Binder AM, Zhou H, Jung SY. DNA methylation patterns associated with breast cancer prognosis that are specific to tumor subtype and menopausal status. Front Genet 2023; 14:1133443. [PMID: 36936429 PMCID: PMC10018014 DOI: 10.3389/fgene.2023.1133443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
Tumor subtype and menopausal status are strong predictors of breast cancer (BC) prognosis. We aimed to find and validate subtype- or menopausal-status-specific changes in tumor DNA methylation (DNAm) associated with all-cause mortality or BC progression. Associations between site-specific tumor DNAm and BC prognosis were estimated among The Cancer Genome Atlas participants (n = 692) with Illumina Infinium HumanMethylation450 BeadChip array data. All-cause mortality and BC progression were modeled using Cox proportional hazards models stratified by tumor subtypes, adjusting for age, race, stage, menopausal status, tumor purity, and cell type proportion. Effect measure modification by subtype and menopausal status were evaluated by incorporating a product term with DNAm. Site-specific inference was used to identify subtype- or menopausal-status-specific differentially methylated regions (DMRs) and functional pathways. The validation of the results was carried out on an independent dataset (GSE72308; n = 180). We identified a total of fifteen unique CpG probes that were significantly associated ( P ≤ 1 × 10 - 7 with survival outcomes in subtype- or menopausal-status-specific manner. Seven probes were associated with overall survival (OS) or progression-free interval (PFI) for women with luminal A subtype, and four probes were associated with PFI for women with luminal B subtype. Five probes were associated with PFI for post-menopausal women. A majority of significant probes showed a lower risk of OS or BC progression with higher DNAm. We identified subtype- or menopausal-status-specific DMRs and functional pathways of which top associated pathways differed across subtypes or menopausal status. None of significant probes from site-specific analyses met genome-wide significant level in validation analyses while directions and magnitudes of coefficients showed consistent pattern. We have identified subtype- or menopausal-status-specific DNAm biomarkers, DMRs and functional pathways associated with all-cause mortality or BC progression, albeit with limited validation. Future studies with larger independent cohort of non-post-menopausal women with non-luminal A subtypes are warranted for identifying subtype- and menopausal-status-specific DNAm biomarkers for BC prognosis.
Collapse
Affiliation(s)
- Do Hyun Kim
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA, United States
| | - Alexandra M. Binder
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, United States
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA, United States
- *Correspondence: Alexandra M. Binder,
| | - Hua Zhou
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Computational Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Su Yon Jung
- Translational Sciences Section, School of Nursing, University of California, Los Angeles, Los Angeles, CA, United States
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
11
|
Liang ZZ, Zhang YX, Zhu RM, Li YL, Jiang HM, Li RB, Chen QX, Wang Q, Tang LY, Ren ZF. Identification of epigenetic modifications mediating the antagonistic effect of selenium against cadmium-induced breast carcinogenesis. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:22056-22068. [PMID: 34773240 DOI: 10.1007/s11356-021-17355-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 10/31/2021] [Indexed: 06/13/2023]
Abstract
The antagonistic effect of selenium (Se) against cadmium (Cd)-induced breast carcinogenesis was reported, but underlying mechanisms were unclear. The aim of this study was to identify the epigenetically regulated genes and biological pathways mediating the antagonistic effect. We exposed MCF-7 cells to Cd and Se alone or simultaneously. Cell proliferation was assessed by MTT assay, and differential epigenome (DNA methylation, microRNA, and long non-coding RNA) was obtained by microarrays. We cross-verified the epigenetic markers with differential transcriptome, and the ones modulated by Cd and Se in opposite directions were regarded to mediate the antagonistic effect. The epigenetically regulated genes were validated by using gene expression data in human breast tissues. We further assessed the biological functions of these validated genes. Our results showed that Se alleviated the proliferative effect of Cd on MCF-7 cell. A total of 10 epigenetically regulated genes were regarded to mediate the antagonistic effect, including APBA2, KIAA0895, DHX35, CPEB3, SVIL, MYLK, ZFYVE28, ABLIM2, GRB10, and PCDH9. Biological function analyses suggested that these epigenetically regulated genes were involved in multiple cancer-related pathways, such as focal adhesion and PI3K/Akt pathway. In conclusion, we provided evidence that Se antagonized the Cd-induced breast carcinogenesis via epigenetic modification and revealed the critical pathways.
Collapse
Affiliation(s)
- Zhuo-Zhi Liang
- School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Yi-Xin Zhang
- School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Rui-Mei Zhu
- School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, China
- The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Yue-Lin Li
- School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, China
- The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Hong-Mei Jiang
- School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Ruo-Bi Li
- School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Qian-Xin Chen
- School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Qing Wang
- School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Lu-Ying Tang
- The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Ze-Fang Ren
- School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, China.
| |
Collapse
|
12
|
Domingo-Fernández D, Gadiya Y, Patel A, Mubeen S, Rivas-Barragan D, Diana CW, Misra BB, Healey D, Rokicki J, Colluru V. Causal reasoning over knowledge graphs leveraging drug-perturbed and disease-specific transcriptomic signatures for drug discovery. PLoS Comput Biol 2022; 18:e1009909. [PMID: 35213534 PMCID: PMC8906585 DOI: 10.1371/journal.pcbi.1009909] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 03/09/2022] [Accepted: 02/09/2022] [Indexed: 12/29/2022] Open
Abstract
Network-based approaches are becoming increasingly popular for drug discovery as they provide a systems-level overview of the mechanisms underlying disease pathophysiology. They have demonstrated significant early promise over other methods of biological data representation, such as in target discovery, side effect prediction and drug repurposing. In parallel, an explosion of -omics data for the deep characterization of biological systems routinely uncovers molecular signatures of disease for similar applications. Here, we present RPath, a novel algorithm that prioritizes drugs for a given disease by reasoning over causal paths in a knowledge graph (KG), guided by both drug-perturbed as well as disease-specific transcriptomic signatures. First, our approach identifies the causal paths that connect a drug to a particular disease. Next, it reasons over these paths to identify those that correlate with the transcriptional signatures observed in a drug-perturbation experiment, and anti-correlate to signatures observed in the disease of interest. The paths which match this signature profile are then proposed to represent the mechanism of action of the drug. We demonstrate how RPath consistently prioritizes clinically investigated drug-disease pairs on multiple datasets and KGs, achieving better performance over other similar methodologies. Furthermore, we present two case studies showing how one can deconvolute the predictions made by RPath as well as predict novel targets.
Collapse
Affiliation(s)
| | - Yojana Gadiya
- Enveda Biosciences, Boulder, Colorado, United States of America
| | - Abhishek Patel
- Enveda Biosciences, Boulder, Colorado, United States of America
| | - Sarah Mubeen
- Bonn-Aachen International Center for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | | | - Chris W. Diana
- Enveda Biosciences, Boulder, Colorado, United States of America
| | | | - David Healey
- Enveda Biosciences, Boulder, Colorado, United States of America
| | - Joe Rokicki
- Enveda Biosciences, Boulder, Colorado, United States of America
| | - Viswa Colluru
- Enveda Biosciences, Boulder, Colorado, United States of America
| |
Collapse
|
13
|
Zheng L, Gu X, Zheng G, Li X, He M, Liu L, Zhou X. Prediction of early recurrence and response to adjuvant Sorafenib for hepatocellular carcinoma after resection. PeerJ 2021; 9:e12554. [PMID: 34900444 PMCID: PMC8628622 DOI: 10.7717/peerj.12554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 11/05/2021] [Indexed: 01/27/2023] Open
Abstract
Background Early recurrence of hepatocellular carcinoma (HCC) is a major obstacle to improving the prognosis, and no widely accepted adjuvant therapy guideline for patients post-liver resection is available. Currently, all available methods and biomarkers are insufficient to accurately predict post-operation HCC patients’ risk of early recurrence and their response to adjuvant therapy. Methods In this study, we downloaded four gene expression datasets (GSE14520, GSE54236, GSE87630, and GSE109211) from the Gene Expression Omnibus database and identified 34 common differentially expressed genes associated with HCC dysregulation and response to adjuvant sorafenib. Then, we constructed a novel 11-messenger RNA predictive model by using ROC curves analysis, univariate Cox regression analysis, and LASSO Cox regression analysis. Furthermore, we validated the predictive values of the risk model in GSE14520 and TCGA-LIHC cohorts by using Kaplan–Meier survival analysis, multivariable Cox regression analysis, and decision curve analysis, respectively. Results The risk score model could identify patients with a high risk of HCC recurrence at the early stage and could predict the response of patients to adjuvant sorafenib. Patients with a high risk score had a worse recurrence rate in training cohorts (2-year: p < 0.0001, hazard ratio (HR): 4.658, confidence interval 95% CI [2.895–7.495]; 5-year: p < 0.0001, HR: 3.251, 95% CI [2.155–4.904]) and external validation cohorts (2-year: p < 0.001, HR: 3.65, 95% CI [2.001–6.658]; 5-year: p < 0.001, HR: 3.156, 95% CI [1.78–5.596]). The AUC values of the risk score model for predicting tumor early recurrence were 0.746 and 0.618, and that of the risk score model for predicting the response to adjuvant sorafenib were 0.722 and 0.708 in the different cohort, respectively. Multivariable Cox regression analysis and decision curve analysis also showed that the risk score model was superior to and independent of other clinicopathologic characteristics. Moreover, the risk score model had excellent abilities to predict the overall survival and HCC recurrence of patients with the same tumor stage category. Conclusions Our risk model is a reliable and superior predictive tool. With this model, we could optimize the risk stratification based on early tumor recurrence and could evaluate the response of patients to adjuvant sorafenib after liver resection.
Collapse
Affiliation(s)
- Liming Zheng
- Central Laboratory, The Third People's Hospital of Changzhou, Changzhou, China
| | - Xi Gu
- Department of General Surgery, The Third People's Hospital of Changzhou, Changzhou, China
| | - Guojun Zheng
- Central Laboratory, The Third People's Hospital of Changzhou, Changzhou, China
| | - Xin Li
- Central Laboratory, The Third People's Hospital of Changzhou, Changzhou, China
| | - Meifang He
- Central Laboratory, The Third People's Hospital of Changzhou, Changzhou, China
| | - Longgen Liu
- Central Laboratory, The Third People's Hospital of Changzhou, Changzhou, China
| | - Xike Zhou
- Clinical Lab, Wuxi No. 5 People's Hospital, Wuxi, China
| |
Collapse
|
14
|
Wang J, Nakato R. HiC1Dmetrics: framework to extract various one-dimensional features from chromosome structure data. Brief Bioinform 2021; 23:6446983. [PMID: 34850813 PMCID: PMC8769930 DOI: 10.1093/bib/bbab509] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/03/2021] [Accepted: 11/05/2021] [Indexed: 01/06/2023] Open
Abstract
Eukaryotic genomes are organized in a three-dimensional spatial structure. In this regard, the development of chromosome conformation capture methods has enabled studies of chromosome organization on a genomic scale. Hi-C, the high-throughput chromosome conformation capture method, can reveal a population-averaged, hierarchical chromatin structure. The typical Hi-C analysis uses a two-dimensional (2D) contact matrix that indicates contact frequencies between all possible genomic position pairs. Oftentimes, however, such a 2D matrix is not amenable to handling quantitative comparisons, visualizations and integrations across multiple datasets. Although several one-dimensional (1D) metrics have been proposed to depict structural information in Hi-C data, their effectiveness is still underappreciated. Here, we first review the currently available 1D metrics for individual Hi-C samples or two-sample comparisons and then discuss their validity and suitable analysis scenarios. We also propose several new 1D metrics to identify additional unique features of chromosome structures. We highlight that the 1D metrics are reproducible and robust for comparing and visualizing multiple Hi-C samples. Moreover, we show that 1D metrics can be easily combined with epigenome tracks to annotate chromatin states in greater details. We develop a new framework, called HiC1Dmetrics, to summarize all 1D metrics discussed in this study. HiC1Dmetrics is open-source (github.com/wangjk321/HiC1Dmetrics) and can be accessed from both command-line and web-based interfaces. Our tool constitutes a useful resource for the community of chromosome-organization researchers.
Collapse
Affiliation(s)
- Jiankang Wang
- Institute for Quantitative Biosciences, The University of Tokyo, Japan.,Graduate School of Medicine, The University of Tokyo, Japan
| | - Ryuichiro Nakato
- Institute for Quantitative Biosciences, The University of Tokyo, Japan.,Graduate School of Medicine, The University of Tokyo, Japan
| |
Collapse
|
15
|
Structural and thermodynamical insights into the binding and inhibition of FIH-1 by the N-terminal disordered region of Mint3. J Biol Chem 2021; 297:101304. [PMID: 34655613 PMCID: PMC8571082 DOI: 10.1016/j.jbc.2021.101304] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 10/09/2021] [Accepted: 10/12/2021] [Indexed: 11/29/2022] Open
Abstract
Mint3 is known to enhance aerobic ATP production, known as the Warburg effect, by binding to FIH-1. Since this effect is considered to be beneficial for cancer cells, the interaction is a promising target for cancer therapy. However, previous research has suggested that the interacting region of Mint3 with FIH-1 is intrinsically disordered, which makes investigation of this interaction challenging. Therefore, we adopted thermodynamic and structural studies in solution to clarify the structural and thermodynamical changes of Mint3 binding to FIH-1. First, using a combination of circular dichroism, nuclear magnetic resonance, and hydrogen/deuterium exchange–mass spectrometry (HDX-MS), we confirmed that the N-terminal half, which is the interacting part of Mint3, is mostly disordered. Next, we revealed a large enthalpy and entropy change in the interaction of Mint3 using isothermal titration calorimetry (ITC). The profile is consistent with the model that the flexibility of disordered Mint3 is drastically reduced upon binding to FIH-1. Moreover, we performed a series of ITC experiments with several types of truncated Mint3s, an effective approach since the interacting part of Mint3 is disordered, and identified amino acids 78 to 88 as a novel core site for binding to FIH-1. The truncation study of Mint3 also revealed the thermodynamic contribution of each part of Mint3 to the interaction with FIH-1, where the core sites contribute to the affinity (ΔG), while other sites only affect enthalpy (ΔH), by forming noncovalent bonds. This insight can serve as a foothold for further investigation of intrinsically disordered regions (IDRs) and drug development for cancer therapy.
Collapse
|
16
|
Pharmacological inhibition of Mint3 attenuates tumour growth, metastasis, and endotoxic shock. Commun Biol 2021; 4:1165. [PMID: 34621018 PMCID: PMC8497560 DOI: 10.1038/s42003-021-02701-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 09/20/2021] [Indexed: 11/21/2022] Open
Abstract
Hypoxia-inducible factor-1 (HIF-1) plays essential roles in human diseases, though its central role in oxygen homoeostasis hinders the development of direct HIF-1-targeted pharmacological approaches. Here, we surveyed small-molecule compounds that efficiently inhibit the transcriptional activity of HIF-1 without affecting body homoeostasis. We focused on Mint3, which activates HIF-1 transcriptional activity in limited types of cells, such as cancer cells and macrophages, by suppressing the factor inhibiting HIF-1 (FIH-1). We identified naphthofluorescein, which inhibited the Mint3–FIH-1 interaction in vitro and suppressed Mint3-dependent HIF-1 activity and glycolysis in cancer cells and macrophages without evidence of cytotoxicity in vitro. In vivo naphthofluorescein administration suppressed tumour growth and metastasis without adverse effects, similar to the genetic depletion of Mint3. Naphthofluorescein attenuated inflammatory cytokine production and endotoxic shock in mice. Thus, Mint3 inhibitors may present a new targeted therapeutic option for cancer and inflammatory diseases by avoiding severe adverse effects. Sakomoto et al. identify naphthofluorescein as a mint3 inhibitor that disrupts the Mint3–FIH-1 interaction and attenuates HIF-1 activity. In vivo experiments in mice reveal a reduction in tumor growth with attenuated inflammatory cytokine production and endotoxic shock, presenting an option for targeted therapies for cancer and inflammatory diseases that avoid severe adverse effects.
Collapse
|
17
|
Huang JY, Cai S, Huang Z, Tint MT, Yuan WL, Aris IM, Godfrey KM, Karnani N, Lee YS, Chan JKY, Chong YS, Eriksson JG, Chan SY. Analyses of child cardiometabolic phenotype following assisted reproductive technologies using a pragmatic trial emulation approach. Nat Commun 2021; 12:5613. [PMID: 34556649 PMCID: PMC8460697 DOI: 10.1038/s41467-021-25899-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 08/06/2021] [Indexed: 11/08/2022] Open
Abstract
Assisted reproductive technologies (ART) are increasingly used, however little is known about the long-term health of ART-conceived offspring. Weak selection of comparison groups and poorly characterized mechanisms impede current understanding. In a prospective cohort (Growing Up in Singapore Towards healthy Outcomes; GUSTO; Clinical Trials ID: NCT01174875) including 83 ART-conceived and 1095 spontaneously-conceived singletons, we estimate effects of ART on anthropometry, blood pressure, serum metabolic biomarkers, and cord tissue DNA methylation by emulating a pragmatic trial supported by machine learning-based estimators. We find ART-conceived children to be shorter (-0.5 SD [95% CI: -0.7, -0.2]), lighter (-0.6 SD [-0.9, -0.3]) and have lower skinfold thicknesses (e.g. -14% [-24%, -3%] suprailiac), and blood pressure (-3 mmHg [-6, -0.5] systolic) at 6-6.5 years, with no strong differences in metabolic biomarkers. Differences are not explained by parental anthropometry or comorbidities, polygenic risk score, breastfeeding, or illnesses. Our simulations demonstrate ART is strongly associated with lower NECAB3 DNA methylation, with negative control analyses suggesting these estimates are unbiased. However, methylation changes do not appear to mediate observed differences in child phenotype.
Collapse
Affiliation(s)
- Jonathan Yinhao Huang
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology, and Research (A*STAR), Singapore, Singapore.
| | - Shirong Cai
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology, and Research (A*STAR), Singapore, Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Zhongwei Huang
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology, and Research (A*STAR), Singapore, Singapore
| | - Mya Thway Tint
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology, and Research (A*STAR), Singapore, Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wen Lun Yuan
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology, and Research (A*STAR), Singapore, Singapore
- Université de Paris, CRESS, Inserm, Paris, France
| | - Izzuddin M Aris
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Keith M Godfrey
- MRC Lifecourse Epidemiology Centre and NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton, Southampton, UK
| | - Neerja Karnani
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology, and Research (A*STAR), Singapore, Singapore
| | - Yung Seng Lee
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology, and Research (A*STAR), Singapore, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jerry Kok Yen Chan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore
- Academic Clinical Program in Obstetrics and Gynaecology, Duke-NUS Medical School, Singapore, Singapore
| | - Yap Seng Chong
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology, and Research (A*STAR), Singapore, Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Johan Gunnar Eriksson
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology, and Research (A*STAR), Singapore, Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- University of Helsinki, Department of General Practise and Primary Health Care, Helsinki University Hospital, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
| | - Shiao-Yng Chan
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology, and Research (A*STAR), Singapore, Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
18
|
Hidden Treasures: Macrophage Long Non-Coding RNAs in Lung Cancer Progression. Cancers (Basel) 2021; 13:cancers13164127. [PMID: 34439281 PMCID: PMC8392679 DOI: 10.3390/cancers13164127] [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: 07/10/2021] [Revised: 08/11/2021] [Accepted: 08/13/2021] [Indexed: 12/16/2022] Open
Abstract
Ever since RNA sequencing of whole genomes and transcriptomes became available, numerous RNA transcripts without having the classic function of encoding proteins have been discovered. Long non-coding RNAs (lncRNAs) with a length greater than 200 nucleotides were considered as "junk" in the beginning, but it has increasingly become clear that lncRNAs have crucial roles in regulating a variety of cellular mechanisms and are often deregulated in several diseases, such as cancer. Lung cancer is the leading cause of cancer-related deaths and has a survival rate of less than 10%. Immune cells infiltrating the tumor microenvironment (TME) have been shown to have a great effect on tumor development with macrophages being the major cell type within the TME. Macrophages can inherit an inflammatory M1 or an anti-inflammatory M2 phenotype. Tumor-associated macrophages, which are predominantly polarized to M2, favor tumor growth, angiogenesis, and metastasis. In this review, we aimed to describe the complex roles and functions of lncRNAs in macrophages and their influence on lung cancer development and progression through the TME.
Collapse
|
19
|
Guo H, Li B, Diao L, Wang H, Chen P, Jiang M, Zhao L, He Y, Zhou C. An immune-based risk-stratification system for predicting prognosis in pulmonary sarcomatoid carcinoma (PSC). Oncoimmunology 2021; 10:1947665. [PMID: 34290908 PMCID: PMC8279095 DOI: 10.1080/2162402x.2021.1947665] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Pulmonary sarcomatoid carcinoma (PSC) is an uncommon subtype of lung cancer, and immune checkpoint blockade promises in clinical benefit. However, virtually nothing is known about the expression of common immune checkpoints in PSC. Here, we performed immunohistochemistry (IHC) to detect nine immune-related proteins in 97 PSC patients. Based on the univariable Cox regression, random forests were used to establish risk models for OS and DFS. Moreover, we used the GSEA, CIBERSORT, and ImmuCellAI to analyze the enriched pathways and microenvironment. Univariable analysis revealed that CD4 (P = 0.008), programmed cell death protein 1 (PD-1; P = 0.003), galectin-9 (Gal-9) on tumor cells (TCs; P = 0.021) were independent for DFS, while CD4 (P = 0.020), PD-1 (P = 0.004), Gal-9 (P = 0.033), and HLA on TILs (P = 0.031) were significant for OS. Meanwhile, the expression level of CD8 played a marginable role in DFS (P = 0.061), limited by the number of patients. The combination of Gal-9 on TC with CD4 and PD-1 on TILs demonstrated the most accurate prediction for DFS (AUC: 0.636-0.791, F1-score: 0.635–0.799), and a dramatic improvement to TNM-stage (P < 0.001 for F1-score of 1-y, 3-y, and 5-yDFS). A similar finding was also observed in the predictive ability of CD4 for OS (AUC: 0.602-0.678, F1-score: 0.635–0.679). CD4 was negatively associated with the infiltration of neutrophils (P = 0.015). PDCD1 (coding gene of PD-1) was positively correlated to the number of exhausted T cells (Texs; P = 0.020) and induced regulatory T cells (iTregs; P = 0.021), and LGALS9 (coding gene of Gal-9) was positively related to the level of dendritic cells (DCs; P = 0.021). Further, a higher combinational level of CD4, PDCD1 on TILs, and LAGLS9 on TCs were proved to be infiltrated with more M1-type macrophages (P < 0.05). We confirmed the expression status of nine immune-related proteins and established a TNM-Immune system for OS and DFS in PSC to assist clinical risk-stratification.
Collapse
Affiliation(s)
- Haoyue Guo
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,School of Medicine, Tongji University, Shanghai, China
| | - Binglei Li
- Department of Computer Science and Technology, College of Electronic and Information Engineering, Tongji University, Shanghai, China
| | - Li Diao
- Department of Automation, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Hao Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,School of Medicine, Tongji University, Shanghai, China
| | - Peixin Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,School of Medicine, Tongji University, Shanghai, China
| | - Minlin Jiang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,School of Medicine, Tongji University, Shanghai, China
| | - Lishu Zhao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,School of Medicine, Tongji University, Shanghai, China
| | - Yayi He
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,School of Medicine, Tongji University, Shanghai, China
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
20
|
Uematsu T, Tsuchiya K, Kobayashi N, Seiki M, Inoue JI, Kaneko S, Sakamoto T. Mint3 depletion-mediated glycolytic and oxidative alterations promote pyroptosis and prevent the spread of Listeria monocytogenes infection in macrophages. Cell Death Dis 2021; 12:404. [PMID: 33854054 PMCID: PMC8046764 DOI: 10.1038/s41419-021-03691-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 04/02/2021] [Accepted: 04/02/2021] [Indexed: 02/07/2023]
Abstract
Listeria monocytogenes (LM) infection induces pyroptosis, a form of regulated necrosis, in host macrophages via inflammasome activation. Here, we examined the role of Mint3 in macrophages, which promotes glycolysis via hypoxia-inducible factor-1 activation, during the initiation of pyroptosis following LM infection. Our results showed that Mint3-deficient mice were more resistant to lethal listeriosis than wild-type (WT) mice. Additionally, the mutant mice showed higher levels of IL-1β/IL-18 in the peritoneal fluid during LM infection than WT mice. Moreover, ablation of Mint3 markedly increased the activation of caspase-1, maturation of gasdermin D, and pyroptosis in macrophages infected with LM in vitro, suggesting that Mint3 depletion promotes pyroptosis. Further analyses revealed that Mint3 depletion upregulates inflammasome assembly preceding pyroptosis via glycolysis reduction and reactive oxygen species production. Pharmacological inhibition of glycolysis conferred resistance to listeriosis in a Mint3-dependent manner. Moreover, Mint3-deficient mice treated with the caspase-1 inhibitor VX-765 were as susceptible to LM infection as WT mice. Taken together, these results suggest that Mint3 depletion promotes pyroptosis in host macrophages, thereby preventing the spread of LM infection. Mint3 may serve as a target for treating severe listeriosis by inducing pyroptosis in LM-infected macrophages.
Collapse
Affiliation(s)
- Takayuki Uematsu
- Biomedical Laboratory, Division of Biomedical Research, Kitasato University Medical Center, Arai, Kitamoto, Saitama, Japan.
| | - Kohsuke Tsuchiya
- Division of Immunology and Molecular Biology, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa, Japan
| | - Noritada Kobayashi
- Biomedical Laboratory, Division of Biomedical Research, Kitasato University Medical Center, Arai, Kitamoto, Saitama, Japan
| | - Motoharu Seiki
- Division of Cancer Cell Research, Institute of Medical Science, The University of Tokyo, Shirokanedai, Minato-ku, Tokyo, Japan
| | - Jun-Ichiro Inoue
- Division of Cellular and Molecular Biology, Institute of Medical Science, The University of Tokyo, Shirokanedai, Minato-ku, Tokyo, Japan
| | - Shuichi Kaneko
- Department of System Biology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Takara-machi, Takara-machi, Kanazawa, Ishikawa, Japan
| | - Takeharu Sakamoto
- Division of Cellular and Molecular Biology, Institute of Medical Science, The University of Tokyo, Shirokanedai, Minato-ku, Tokyo, Japan.
- Department of System Biology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Takara-machi, Takara-machi, Kanazawa, Ishikawa, Japan.
| |
Collapse
|
21
|
Mint3 is dispensable for pancreatic and kidney functions in mice. Biochem Biophys Rep 2020; 24:100872. [PMID: 33319072 PMCID: PMC7725678 DOI: 10.1016/j.bbrep.2020.100872] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 11/06/2020] [Accepted: 11/24/2020] [Indexed: 02/07/2023] Open
Abstract
Munc-18 interacting protein 3 (Mint3) is an activator of hypoxia-inducible factor-1 in cancer cells, macrophages, and cancer-associated fibroblasts under pathological conditions. However, exactly which cells highly express Mint3 in vivo and whether Mint3 depletion affects their physiological functions remain unclear. Here, we surveyed mouse tissues for specific expression of Mint3 by comparing Mint3 expression in wild-type and Mint3-knockout mice. Interestingly, immunohistochemical analyses revealed that Mint3 was highly expressed in islet cells of the pancreas, distal tubular epithelia of the kidney, choroid plexus ependymal cells of the cerebrum, medullary cells of the adrenal gland, and epithelial cells of the seminal gland. We also studied whether Mint3 depletion affects the physiological functions of the islets and kidneys. Mint3-knockout mice did not show any abnormalities in glucose-tolerance and urine-biochemical tests, indicating that Mint3 depletion was compensated for in these organs. Thus, loss of Mint3 might be compensated in the islets and kidneys under physiological conditions in mice. Specific expression of Mint3 in mouse tissues is surveyed. Mint3 is highly expressed in islet cells of the pancreas. Mint3 is highly expressed in distal tubular epithelia of the kidney. Mint3 KO mice do not show any abnormalities in glucose-tolerance tests. Mint3 KO mice do not show any abnormalities in urine-biochemical tests.
Collapse
|
22
|
Mint3 depletion restricts tumor malignancy of pancreatic cancer cells by decreasing SKP2 expression via HIF-1. Oncogene 2020; 39:6218-6230. [PMID: 32826949 PMCID: PMC7515798 DOI: 10.1038/s41388-020-01423-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 07/18/2020] [Accepted: 08/07/2020] [Indexed: 12/20/2022]
Abstract
Pancreatic cancer is one of the most fatal cancers without druggable molecular targets. Hypoxia inducible factor-1 (HIF-1) is a heterodimeric transcriptional factor that promotes malignancy in various cancers including pancreatic cancer. Herein, we found that HIF-1 is accumulated in normoxic or moderate hypoxic areas of pancreatic cancer xenografts in vivo and is active even during normoxia in pancreatic cancer cells in vitro. This prompted us to analyze whether the HIF-1 activator Mint3 contributes to malignant features of pancreatic cancer. Mint3 depletion by shRNAs attenuated HIF-1 activity during normoxia and cell proliferation concomitantly with accumulated p21 and p27 protein in pancreatic cancer cells. Further analyses revealed that Mint3 increased transcription of the oncogenic ubiquitin ligase SKP2 in pancreatic cancer cells via HIF-1. This Mint3-HIF-1-SKP2 axis also promoted partial epithelial-mesenchymal transition, stemness features, and chemoresistance in pancreatic cancer cells. Even in vivo, Mint3 depletion attenuated tumor growth of orthotopically inoculated human pancreatic cancer AsPC-1 cells. Database and tissue microarray analyses showed that Mint3 expression is correlated with SKP2 expression in human pancreatic cancer specimens and high Mint3 expression is correlated with poor prognosis of pancreatic cancer patients. Thus, targeting Mint3 may be useful for attenuating the malignant features of pancreatic cancer.
Collapse
|
23
|
GNAS-AS1/miR-4319/NECAB3 axis promotes migration and invasion of non-small cell lung cancer cells by altering macrophage polarization. Funct Integr Genomics 2019; 20:17-28. [PMID: 31267263 DOI: 10.1007/s10142-019-00696-x] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 06/11/2019] [Accepted: 06/19/2019] [Indexed: 12/13/2022]
Abstract
Non-small cell lung cancer (NSCLC) represents for approximately 85% of all lung cancers, which is the most common cancer worldwide. Tumor-associated macrophages (TAM) are crucial for tumor progression, which was widely believed to be mediated by long non-coding RNAs (LncRNAs). We aimed to explore the effect of one LncRNA, GNAS-AS1, in TAM-associated NSCLC progression. Relative mRNA levels were determined by qRT-PCR. Western blot and ELISA were used to detect protein levels. Proliferation in vitro was assessed by MTT and clone formation assays. Migration and invasion of cell lines were evaluated by transwell-based assays. Interaction between molecules was detected by luciferase report assay. GNAS-AS1 expression was dramatically enhanced in TAM, NSCLC cell lines, and clinical tumor tissues, and negatively correlated with overall survival of NSCLC patients. GNAS-AS1 promoted macrophage M2 polarization and NSCLC cell progression via directly inhibiting miR-4319, which could target N-terminal EF-hand calcium binding protein 3 (NECAB3) to inhibit its expression. GNAS-AS1/miR-4319/NECAB3 axis promotes tumor progression of NSCLC by altering macrophage polarization. This novel mechanism may provide potential strategy for NSCLC treatment.
Collapse
|
24
|
Hara T, Murakami Y, Seiki M, Sakamoto T. Mint3 in bone marrow-derived cells promotes lung metastasis in breast cancer model mice. Biochem Biophys Res Commun 2017. [PMID: 28634075 DOI: 10.1016/j.bbrc.2017.06.102] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Breast cancer is one of the most common cancers in women in the world. Although breast cancer is well treatable at the early stage, patients with distant metastases show a poor prognosis. Data from recent studies using transplantation models indicate that Mint3/APBA3 might promote breast cancer malignancy. However, whether Mint3 indeed contributes to tumor development, progression, or metastasis in vivo remains unclear. To address this, here we examined whether Mint3 depletion affects tumor malignancy in MMTV-PyMT breast cancer model mice. In MMTV-PyMT mice, Mint3 depletion did not affect tumor onset and tumor growth, but attenuated lung metastases. Experimental lung metastasis of breast cancer Met-1 cells derived from MMTV-PyMT mice also decreased in Mint3-depleted mice, indicating that host Mint3 expression affected lung metastasis of MMTV-PyMT-derived breast cancer cells. Further bone marrow transplant experiments revealed that Mint3 in bone marrow-derived cells promoted lung metastasis in MMTV-PyMT mice. Thus, targeting Mint3 in bone marrow-derived cells might be a good strategy for preventing metastasis and improving the prognosis of breast cancer patients.
Collapse
Affiliation(s)
- Toshiro Hara
- Division of Cancer Cell Research, Institute of Medical Science, The University of Tokyo, Shirokanedai, Minato-ku, 108-8639 Tokyo, Japan
| | - Yoshinori Murakami
- Division of Molecular Pathology, Institute of Medical Science, The University of Tokyo, Shirokanedai, Minato-ku, 108-8639 Tokyo, Japan
| | - Motoharu Seiki
- Division of Cancer Cell Research, Institute of Medical Science, The University of Tokyo, Shirokanedai, Minato-ku, 108-8639 Tokyo, Japan; Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Takara-machi, 920-8641 Kanazawa, Japan
| | - Takeharu Sakamoto
- Division of Cancer Cell Research, Institute of Medical Science, The University of Tokyo, Shirokanedai, Minato-ku, 108-8639 Tokyo, Japan; Division of Molecular Pathology, Institute of Medical Science, The University of Tokyo, Shirokanedai, Minato-ku, 108-8639 Tokyo, Japan.
| |
Collapse
|
25
|
Sakamoto T, Seiki M. Integrated functions of membrane-type 1 matrix metalloproteinase in regulating cancer malignancy: Beyond a proteinase. Cancer Sci 2017; 108:1095-1100. [PMID: 28267240 PMCID: PMC5480062 DOI: 10.1111/cas.13231] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 03/01/2017] [Accepted: 03/02/2017] [Indexed: 12/19/2022] Open
Abstract
Membrane‐type 1 matrix metalloproteinase (MT1‐MMP) is expressed in different types of invasive and proliferative cells, including cancer cells and stromal cells. MT1‐MMP cleaves extracellular matrix proteins, membrane proteins and other pericellular proteins, thereby changing the cellular microenvironment and regulating signal activation. Critical roles of protease activity in cancer cell proliferation, invasion and metastasis have been demonstrated by many groups. MT1‐MMP also has a non‐protease activity in that it inhibits the oxygen‐dependent suppression of hypoxia‐inducible factors (HIFs) via Munc18‐1‐interacting protein 3 (Mint3) and thereby enhances the expression of HIF target genes. Elevated HIF activity in MT1‐MMP‐expressing cancer cells is a fundamental mechanism underlying the Warburg effect, a well‐known phenomenon where malignant cancer cells exhibit a higher rate of glucose metabolism. Because specific intervention of HIF activation by MT1‐MMP suppresses tumor formation by cancer cells in mice, both the proteolytic and non‐proteolytic activities of MT1‐MMP are important for tumor malignancy and function in an integrated manner. In this review, we summarize recent findings relating to how MT1‐MMP activates HIF and its effects on cancer cells and stromal cells.
Collapse
Affiliation(s)
- Takeharu Sakamoto
- Division of Molecular PathologyThe Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Motoharu Seiki
- Faculty of MedicineInstitute of Medical, Pharmaceutical and Health Sciences, Kanazawa UniversityKanazawaJapan
| |
Collapse
|
26
|
Mint3-mediated L1CAM expression in fibroblasts promotes cancer cell proliferation via integrin α5β1 and tumour growth. Oncogenesis 2017; 6:e334. [PMID: 28504692 PMCID: PMC5523060 DOI: 10.1038/oncsis.2017.27] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 03/20/2017] [Indexed: 12/12/2022] Open
Abstract
Fibroblasts are some of the major cells in tumour tissues that influence tumour progression and drug resistance. However, our understanding on fibroblast-mediated tumour malignancy remains incomplete. Munc18-1-interacting protein 3 (Mint3) is known as an activator of hypoxia-inducible factor-1 (HIF-1) even during normoxia in cancer cells, macrophages and fibroblasts. Although Mint3 promotes ATP production via glycolysis by activating HIF-1 in cancer cells and macrophages, the biological role of Mint3-mediated HIF-1 activation in fibroblasts remains unclear. To address this, we examined whether Mint3 in fibroblasts contributes to tumour growth. Mint3 depletion in mouse embryonic fibroblasts (MEFs) decreased tumour growth of co-injected human breast cancer cells, MDA-MB-231 and epidermoid carcinoma A431 cells in mice. In MEFs, Mint3 also promoted cancer cell proliferation in vitro in a cell–cell contact-dependent manner. Mint3-mediated cancer cell proliferation depended on HIF-1, and further gene expression analysis revealed that the cell adhesion molecule, L1 cell adhesion molecule (L1CAM), was induced by Mint3 and HIF-1 in fibroblasts. Mint3-mediated L1CAM expression in fibroblasts stimulated the ERK signalling pathway via integrin α5β1 in cancer cells, and promoted cancer cell proliferation in vitro and tumour growth. In cancer-associated fibroblasts (CAFs), knockdown of MT1-MMP, which promotes Mint3-mediated HIF-1 activation, or Mint3 decreased L1CAM expression. As MEFs, CAFs also promoted cancer cell proliferation in vitro, and tumour growth via Mint3 and L1CAM. In human breast cancer specimens, the number of fibroblasts expressing L1CAM, Mint3 and MT1-MMP was higher in cancer regions than in adjacent benign regions. In addition, more phospho-ERK1/2-positive cancer cells existed in the peripheral region surrounded by the stroma than in the central region of solid breast cancer nest. Thus, Mint3 in fibroblasts might be a good target for cancer therapy by regulating cancer cell-stromal cell communication.
Collapse
|
27
|
Zhang J, Chen JH, Liu XD, Wang HY, Liu XL, Li XY, Wu ZF, Zhu MJ, Zhao SH. Genomewide association studies for hematological traits and T lymphocyte subpopulations in a Duroc × Erhualian F resource population. J Anim Sci 2017; 94:5028-5041. [PMID: 28046140 DOI: 10.2527/jas.2016-0924] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
It has been shown that hematological traits can act as important indicators of immune function in both humans and livestock. T lymphocytes are key components of the adaptive immune system, playing a critical role in immune response. To identify genomic regions affecting hematological traits and T lymphocyte subpopulations, we performed both a SNP-based genomewide association study (GWAS) and a haplotype analysis for 20 hematological traits and 8 T cell subpopulations at 3 different time points (d 20, 33, and 35) in a Duroc × Erhualian F intercross population. Bonferroni correction was used to calculate the threshold -values for suggestive and 5% genomewide significance levels. In total, for SNP-based GWAS, we detected 96 significant SNP, including 15 genomewide-significant SNP, associated with 23 hematological traits and 234 significant SNP, including 27 genomewide-significant SNP, associated with 8 T cell subpopulations. Meanwhile, we identified 563 significant SNP, including 7 genomewide-significant SNP, associated with 5 hematological traits and 2,407 significant SNP, including 1,261 genomewide-significant SNP, associated with 8 T cell subpopulations by haplotype analysis. Among the significant regions detected, we propose both the () gene and the () gene on SSC3 as plausible candidate genes associated with CD/CD T lymphocytes at d 20. The findings provide insights into the basis of molecular mechanisms that are involved with immune response in the domestic pig and would aid further identification of causative mutations.
Collapse
|
28
|
Martin AR, Ronco C, Demange L, Benhida R. Hypoxia inducible factor down-regulation, cancer and cancer stem cells (CSCs): ongoing success stories. MEDCHEMCOMM 2017; 8:21-52. [PMID: 30108689 PMCID: PMC6071925 DOI: 10.1039/c6md00432f] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 10/10/2016] [Indexed: 12/12/2022]
Abstract
In cancers, hypoxia inducible factor 1 (HIF-1) is an over-expressed transcription factor, which regulates a large set of genes involved in tumour vascularization, metastases, and cancer stem cells (CSCs) formation and self-renewal. This protein has been identified as a relevant target in oncology and several HIF-1 modulators are now marketed or in advanced clinical trials. The purpose of this review is to summarize the advances in the understanding of its regulation and its inhibition, from the medicinal chemist point of view. To this end, we selected in the recent literature relevant examples of "hit" compounds, including small-sized organic molecules, pseudopeptides and nano-drugs, exhibiting in vitro and/or in vivo both anti-HIF-1 and anti-tumour activities. Whenever possible, a particular emphasis has been dedicated to compounds that selectively target CSCs.
Collapse
Affiliation(s)
- Anthony R Martin
- Université Côte d'Azur , CNRS , Institut de Chimie de Nice UMR 7272 - 06108 Nice , France . ; ; ; Tel: +33 4 92076143
| | - Cyril Ronco
- Université Côte d'Azur , CNRS , Institut de Chimie de Nice UMR 7272 - 06108 Nice , France . ; ; ; Tel: +33 4 92076143
| | - Luc Demange
- Université Côte d'Azur , CNRS , Institut de Chimie de Nice UMR 7272 - 06108 Nice , France . ; ; ; Tel: +33 4 92076143
- UFR des Sciences Pharmaceutiques , Université Paris Descartes , Sorbonne Paris Cité , 4 avenue de l'Observatoire , Paris Fr-75006 , France
- UFR Biomédicale des Saints Pères , 45 rue des Saints Pères , Paris Fr-75006 , France
| | - Rachid Benhida
- Université Côte d'Azur , CNRS , Institut de Chimie de Nice UMR 7272 - 06108 Nice , France . ; ; ; Tel: +33 4 92076143
| |
Collapse
|
29
|
Mint3/Apba3 depletion ameliorates severe murine influenza pneumonia and macrophage cytokine production in response to the influenza virus. Sci Rep 2016; 6:37815. [PMID: 27883071 PMCID: PMC5121658 DOI: 10.1038/srep37815] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 11/02/2016] [Indexed: 01/06/2023] Open
Abstract
Influenza virus (IFV) infection is a common cause of severe pneumonia. Studies have suggested that excessive activation of the host immune system including macrophages is responsible for the severe pathologies mediated by IFV infection. Here, we focused on the X11 protein family member Mint3/Apba3, known to promote ATP production via glycolysis by activating hypoxia inducible factor-1 (HIF-1) in macrophages, and examined its roles in lung pathogenesis and anti-viral defence upon IFV infection. Mint3-deficient mice exhibited improved influenza pneumonia with reduced inflammatory cytokines/chemokine levels and neutrophil infiltration in the IFV-infected lungs without alteration in viral burden, type-I interferon production, or acquired immunity. In macrophages, Mint3 depletion attenuated NF-κB signalling and the resultant cytokine/chemokine production in response to IFV infection by increasing IκBα and activating the cellular energy sensor AMPK, respectively. Thus, Mint3 might represent one of the likely therapeutic targets for the treatment of severe influenza pneumonia without affecting host anti-viral defence through suppressing macrophage cytokine/chemokine production.
Collapse
|