1
|
Kohara Y, Yasuda S, Nagai M, Nakamura K, Matsuo Y, Terai T, Doi S, Sakata T, Sho M. Critical Role of Preoperative Hyperglycemia in Association with GFAT-1 Expression in Resected Pancreatic Cancer. Ann Surg Oncol 2024; 31:7713-7721. [PMID: 39107610 DOI: 10.1245/s10434-024-15985-4] [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: 04/26/2024] [Accepted: 07/23/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Hyperglycemia is involved in malignant transformation of pancreatic cancer via the hexosamine biosynthetic pathway (HBP). However, few studies have verified this mechanism based on clinical data. This study investigated the complementary effects of hyperglycemia and HBP on pancreatic cancer prognosis using detailed clinical data. METHODS The study analyzed data of 477 patients with pancreatic cancer who underwent pancreatectomy between 2006 and 2020. The patients were divided into normoglycemia and hyperglycemia groups based on their HbA1c levels. Immunostaining for glutamine fructose-6-phosphate transaminase-1 (GFAT-1), the rate-limiting enzyme in HBP, CD4, CD8, and Foxp3, was performed to evaluate the association between survival outcomes, HBP, and local tumor immunity. RESULTS Overall survival (OS) was significantly poorer in the hyperglycemia group than in the normoglycemia group (mean survival time [MST]: 35.0 vs. 47.9 months; p = 0.007). The patients in the hyperglycemia group with high GFAT-1 expression had significantly poorer OS than those with low GFAT-1 expression (MST, 49.0 vs. 27.6 months; p < 0.001). However, the prognosis did not differ significantly between the patients with high and low GFAT-1 expression in the normoglycemia group. In addition, the patients with hyperglycemia and high GFAT-1 expression had fewer CD4+ (p = 0.015) and CD8+ (p = 0.017) T cells and a lower CD8+/Foxp3+ ratio (p = 0.032) than those with hyperglycemia and low GFAT-1 expression. CONCLUSIONS The patients with hyperglycemia and high GFAT-1 expression levels had an extremely poor prognosis. Furthermore, the tumors in these patients were characterized as immunologically cold tumors.
Collapse
Affiliation(s)
| | - Satoshi Yasuda
- Department of Surgery, Nara Medical University, Nara, Japan
| | - Minako Nagai
- Department of Surgery, Nara Medical University, Nara, Japan
| | - Kota Nakamura
- Department of Surgery, Nara Medical University, Nara, Japan
| | - Yasuko Matsuo
- Department of Surgery, Nara Medical University, Nara, Japan
| | - Taichi Terai
- Department of Surgery, Nara Medical University, Nara, Japan
| | - Shunsuke Doi
- Department of Surgery, Nara Medical University, Nara, Japan
| | - Takeshi Sakata
- Department of Surgery, Nara Medical University, Nara, Japan
| | - Masayuki Sho
- Department of Surgery, Nara Medical University, Nara, Japan.
| |
Collapse
|
2
|
Zhang X, Zhong Y, Yang Q. FOXM1 Upregulates O-GlcNAcylation Level Via The Hexosamine Biosynthesis Pathway to Promote Angiogenesis in Hepatocellular Carcinoma. Cell Biochem Biophys 2024; 82:2767-2785. [PMID: 39031247 DOI: 10.1007/s12013-024-01393-8] [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] [Accepted: 06/29/2024] [Indexed: 07/22/2024]
Abstract
Hepatocellular carcinoma (HCC) presents significant challenges in treatment and prognosis because of its aggressive nature and high metastatic potential. This study aims to investigate the role of the hexosamine biosynthesis pathway (HBP) and its association with HCC progression and prognosis. We identified SPP1 and FOXM1 as hub genes within the HBP pathway, showing their correlation with poor prognosis and late-stage progression. In addition, the analysis uncovered the complex participation of the HBP pathway in nutrients and oxygen reactions, PI3K-AKT signaling, AMPK activation, and angiogenesis regulation. The disruption of these pathways is pivotal in influencing the growth and progression of HCC. Targeting the HBP presents a promising therapeutic approach to modulate the tumor microenvironment, thereby enhancing the efficacy of immunotherapy. In addition, FOXM1 was identified as the HBP pathway regulator, influencing cellular O-GlcNAcylation level and VEGF secretion, thereby promoting angiogenesis in HCC. Inhibition of O-GlcNAcylation significantly hindered angiogenesis, which is suggested as a potential avenue for therapeutic intervention. Our research demonstrates the practicality of using the HBP-related gene as a prognostic marker in liver cancer patients and suggests targeting FOXM1 as a novel avenue for personalized therapy.
Collapse
Affiliation(s)
- Xiaorong Zhang
- Department of Pathogenobiology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, Jilin Province, China
| | - Yifan Zhong
- Department of Pathogenobiology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, Jilin Province, China
| | - Qing Yang
- Department of Pathogenobiology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, Jilin Province, China.
| |
Collapse
|
3
|
Simon‐Molas H, Del Prete R, Kabanova A. Glucose metabolism in B cell malignancies: a focus on glycolysis branching pathways. Mol Oncol 2024; 18:1777-1794. [PMID: 38115544 PMCID: PMC11223612 DOI: 10.1002/1878-0261.13570] [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: 07/17/2023] [Revised: 10/13/2023] [Accepted: 12/15/2023] [Indexed: 12/21/2023] Open
Abstract
Glucose catabolism, one of the essential pathways sustaining cellular bioenergetics, has been widely studied in the context of tumors. Nevertheless, the function of various branches of glucose metabolism that stem from 'classical' glycolysis have only been partially explored. This review focuses on discussing general mechanisms and pathological implications of glycolysis and its branching pathways in the biology of B cell malignancies. We summarize here what is known regarding pentose phosphate, hexosamine, serine biosynthesis, and glycogen synthesis pathways in this group of tumors. Despite most findings have been based on malignant B cells themselves, we also discuss the role of glucose metabolism in the tumor microenvironment, with a focus on T cells. Understanding the contribution of glycolysis branching pathways and how they are hijacked in B cell malignancies will help to dissect the role they have in sustaining the dissemination and proliferation of tumor B cells and regulating immune responses within these tumors. Ultimately, this should lead to deciphering associated vulnerabilities and improve current therapeutic schedules.
Collapse
Affiliation(s)
- Helga Simon‐Molas
- Departments of Experimental Immunology and HematologyAmsterdam UMC location University of AmsterdamThe Netherlands
- Cancer ImmunologyCancer Center AmsterdamThe Netherlands
| | | | - Anna Kabanova
- Fondazione Toscana Life Sciences FoundationSienaItaly
| |
Collapse
|
4
|
Papadatos SS, Mitselou A, Lampri E, Bali C, Betzou A, Varouktsi A, Katsanos K, Galani V. Expression of ten-eleven translocation 2 and glutathione-S-transferase pi in colorectal cancer patients with and without type 2 diabetes mellitus. Folia Med (Plovdiv) 2024; 66:370-379. [PMID: 39365623 DOI: 10.3897/folmed.66.e122713] [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: 03/10/2024] [Accepted: 04/24/2024] [Indexed: 10/05/2024] Open
Abstract
AIMS To highlight possible correlations of type 2 diabetes mellitus (T2DM) with microscopic / macroscopic characteristics of colorectal cancer tissues, along with the expression of Ten-Eleven Translocation 2 (TET2) and glutathione-S-transferase pi (GST-pi) proteins.
Collapse
Affiliation(s)
| | | | | | | | | | - Anna Varouktsi
- Ippokratio Hospital of Thessaloniki, Thessaloniki, Greece
| | | | | |
Collapse
|
5
|
Li F, Wan X, Li Z, Zhou L. High glucose inhibits autophagy and promotes the proliferation and metastasis of colorectal cancer through the PI3K/AKT/mTOR pathway. Cancer Med 2024; 13:e7382. [PMID: 38872380 PMCID: PMC11176572 DOI: 10.1002/cam4.7382] [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: 02/25/2024] [Revised: 05/16/2024] [Accepted: 05/28/2024] [Indexed: 06/15/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) ranks among the most prevalent malignancies worldwide, characterized by its complex etiology and slow research progress. Diabetes, as an independent risk factor for CRC, has been widely certified. Consequently, this study centers on elucidating the intricacies of CRC cells initiation and progression within a high-glucose environment. METHODS A battery of assays was employed to assess the proliferation and metastasis of CRC cells cultured under varying glucose concentrations. Optimal glucose levels conducive to cells' proliferation and migration were identified. Western blot analyses were conducted to evaluate alterations in apoptosis, autophagy, and EMT-related proteins in CRC cells under high-glucose conditions. The expression of PI3K/AKT/mTOR pathway-associated proteins was assessed using western blot. The effect of high glucose on xenograft growth was investigated in vivo by MC38 cells, and changes in inflammatory factors (IL-4, IL-13, TNF-α, IL-5, and IL-12) were measured via serum ELISA. RESULTS Our experiments demonstrated that elevated glucose concentrations promoted both the proliferation and migration of CRC cells; the most favorable glucose dose is 20 mM. Western blot analyses revealed a decrease in apoptotic proteins, such as Bim, Bax, and caspase-3 with increasing glucose levels. Concurrently, the expression of EMT-related proteins, including N-cadherin, vimentin, ZEB1, and MMP9, increased. High-glucose cultured cells exhibited elevated levels of PI3K/AKT/mTOR pathway proteins. In the xenograft model, tumor cells stimulated by high glucose exhibited accelerated growth, larger tumor volumes, and heightened KI67 expression of immunohistochemistry. ELISA experiments revealed higher expression of IL-4 and IL-13 and lower expression of TNF-α and IL-5 in the serum of high-glucose-stimulated mice. CONCLUSION The most favorable dose and time for tumor cells proliferation and migration is 20 mM, 48 h. High glucose fosters CRC cell proliferation and migration while suppressing autophagy through the activation of the PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Feng Li
- Department of Pharmacology, West China School of Basic Science and Forensic Medicine, Sichuan University, Chengdu, China
| | - Xing Wan
- Department of Pharmacology, West China School of Basic Science and Forensic Medicine, Sichuan University, Chengdu, China
| | - Zhigui Li
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Liming Zhou
- Department of Pharmacology, West China School of Basic Science and Forensic Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
6
|
Wang Y, Wang S, Wang Y, Wang C, Tang Y, Zhang C, Hou S, Yu D, Lin N. Glucose regulates the HMGB1 signaling pathway through SIRT1 in glioma. Cell Signal 2024; 118:111137. [PMID: 38467242 DOI: 10.1016/j.cellsig.2024.111137] [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: 01/14/2024] [Revised: 02/28/2024] [Accepted: 03/08/2024] [Indexed: 03/13/2024]
Abstract
BACKGROUND Glucose is a fundamental substance for numerous cancers, including glioma. However, its influence on tumor cells regulatory mechanisms remains uncertain. SIRT1 is a regulator of deacetylation and a key player in the progression of malignant tumors. The objective of this study was to examine the role of glucose and SIRT1 in glioma. METHODS This study investigated the association of SIRT1 expression with clinicopathological features and prognosis in glioma patients using the TCGA database. The Western blotting technique was used to identify the expression of SIRT1 protein in glioma cells. The study also examined the impact of differing glucose concentrations on the biological functions of glioma cells. The study investigated the expression of SIRT1 and HMGB1 signaling pathways in glioma. Additionally, resilience experiments were conducted utilizing SRT1720. RESULTS SIRT1 is a gene that suppresses tumors and is low expressed in gliomas. Low expression of this gene is strongly linked to a poor prognosis in patients with glioma. High concentrations of glucose can promote the proliferation, migration, and invasion of glioma cells, while also inhibiting apoptosis. The findings of this mechanistic study provide evidence that glucose can down-regulate SIRT1 expression, leading to increased levels of acetylated HMGB1. This in turn promotes the ex-nuclear activation of HMGB1 and associated signaling pathways, ultimately driving glioma malignancy. CONCLUSION Glucose has the ability to regulate the HMGB1 associated signaling pathway through SIRT1, thus promoting glioma progression. This holds significant research value.
Collapse
Affiliation(s)
- Yu Wang
- Department of Neurosurgery, The Affiliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou 239000, China
| | - Shuai Wang
- Department of Neurosurgery, The Affiliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou 239000, China
| | - Yuhao Wang
- Department of Neurosurgery, The Affiliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou 239000, China
| | - Chengcheng Wang
- Department of Neurosurgery, The Affiliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou 239000, China
| | - Yuhang Tang
- Department of Neurosurgery, The Affiliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou 239000, China
| | - Chao Zhang
- Department of Neurosurgery, The Affiliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou 239000, China
| | - Shiqiang Hou
- Department of Neurosurgery, The Affiliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou 239000, China
| | - Dong Yu
- Department of Neurosurgery, The Affiliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou 239000, China.
| | - Ning Lin
- Department of Neurosurgery, The Affiliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou 239000, China.
| |
Collapse
|
7
|
Schuurmans F, Wagemans KE, Adema GJ, Cornelissen LAM. Tumor glucose metabolism and the T cell glycocalyx: implication for T cell function. Front Immunol 2024; 15:1409238. [PMID: 38881904 PMCID: PMC11176483 DOI: 10.3389/fimmu.2024.1409238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 05/21/2024] [Indexed: 06/18/2024] Open
Abstract
The T cell is an immune cell subset highly effective in eliminating cancer cells. Cancer immunotherapy empowers T cells and occupies a solid position in cancer treatment. The response rate, however, remains relatively low (<30%). The efficacy of immunotherapy is highly dependent on T cell infiltration into the tumor microenvironment (TME) and the ability of these infiltrated T cells to sustain their function within the TME. A better understanding of the inhibitory impact of the TME on T cells is crucial to improve cancer immunotherapy. Tumor cells are well described for their switch into aerobic glycolysis (Warburg effect), resulting in high glucose consumption and a metabolically distinct TME. Conversely, glycosylation, a predominant posttranslational modification of proteins, also relies on glucose molecules. Proper glycosylation of T cell receptors influences the immunological synapse between T cells and tumor cells, thereby affecting T cell effector functions including their cytolytic and cytostatic activities. This review delves into the complex interplay between tumor glucose metabolism and the glycocalyx of T cells, shedding light on how the TME can induce alterations in the T cell glycocalyx, which can subsequently influence the T cell's ability to target and eliminate tumor cells.
Collapse
Affiliation(s)
| | | | | | - Lenneke A. M. Cornelissen
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
8
|
Wei S, Ai M, Zhan Y, Yu J, Xie T, Hu Q, Fang Y, Huang X, Li Y. TRIM14 suppressed the progression of NSCLC via hexosamine biosynthesis pathway. Carcinogenesis 2024; 45:324-336. [PMID: 38267812 DOI: 10.1093/carcin/bgae005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 01/17/2024] [Accepted: 01/23/2024] [Indexed: 01/26/2024] Open
Abstract
Tripartite Motif 14 (TRIM14) is an oncoprotein that belongs to the E3 ligase TRIM family, which is involved in the progression of various tumors except for non-small cell lung carcinoma (NSCLC). However, little is currently known regarding the function and related mechanisms of TRIM14 in NSCLC. Here, we found that the TRIM14 protein was downregulated in lung adenocarcinoma tissues compared with the adjacent tissues, which can suppress tumor cell proliferation and migration both in vitro and in vivo. Moreover, TRIM14 can directly bind to glutamine fructose-6-phosphate amidotransferase 1 (GFAT1), which in turn results in the degradation of GFAT1 and reduced O-glycosylation levels. GFAT1 is a key enzyme in the rate-limiting step of the hexosamine biosynthetic pathway (HBP). Replenishment of N-acetyl-d-glucosamine can successfully reverse the inhibitory effect of TRIM14 on the NSCLC cell growth and migration as expected. Collectively, our data revealed that TRIM14 suppressed NSCLC cell proliferation and migration through ubiquitination and degradation of GFAT1, providing a new regulatory role for TRIM14 on HBP.
Collapse
Affiliation(s)
- Sisi Wei
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Meiling Ai
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, The Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
- Department of Pharmacy, Jiangxi Maternal and Child Health Hospital, Nanchang 330006, China
| | - Yuan Zhan
- Department of Pathology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Jieqing Yu
- Department of Otorhinolaryngology Head and Neck Surgery, Jiangxi Otorhinolaryngology Head and Neck Surgery Institute, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Tao Xie
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, The Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Qinghua Hu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, The Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Yang Fang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, The Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Xuan Huang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, The Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Yong Li
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| |
Collapse
|
9
|
Drury J, Geisen ME, Tessmann JW, Rychahou PG, Kelson CO, He D, Wang C, Evers BM, Zaytseva YY. Overexpression of Fatty Acid Synthase Upregulates Glutamine-Fructose-6-Phosphate Transaminase 1 and O-Linked N-Acetylglucosamine Transferase to Increase O-GlcNAc Protein Glycosylation and Promote Colorectal Cancer Growth. Int J Mol Sci 2024; 25:4883. [PMID: 38732103 PMCID: PMC11084459 DOI: 10.3390/ijms25094883] [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: 04/03/2024] [Revised: 04/24/2024] [Accepted: 04/28/2024] [Indexed: 05/13/2024] Open
Abstract
Fatty acid synthesis has been extensively investigated as a therapeutic target in cancers, including colorectal cancer (CRC). Fatty acid synthase (FASN), a key enzyme of de novo lipid synthesis, is significantly upregulated in CRC, and therapeutic approaches of targeting this enzyme are currently being tested in multiple clinical trials. However, the mechanisms behind the pro-oncogenic action of FASN are still not completely understood. Here, for the first time, we show that overexpression of FASN increases the expression of glutamine-fructose-6-phosphate transaminase 1 (GFPT1) and O-linked N-acetylglucosamine transferase (OGT), enzymes involved in hexosamine metabolism, and the level of O-GlcNAcylation in vitro and in vivo. Consistently, expression of FASN significantly correlates with expression of GFPT1 and OGT in human CRC tissues. shRNA-mediated downregulation of GFPT1 and OGT inhibits cellular proliferation and the level of protein O-GlcNAcylation in vitro, and knockdown of GFPT1 leads to a significant decrease in tumor growth and metastasis in vivo. Pharmacological inhibition of GFPT1 and OGT leads to significant inhibition of cellular proliferation and colony formation in CRC cells. In summary, our results show that overexpression of FASN increases the expression of GFPT1 and OGT as well as the level of protein O-GlcNAcylation to promote progression of CRC; targeting the hexosamine biosynthesis pathway could be a therapeutic approach for this disease.
Collapse
Affiliation(s)
- James Drury
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA; (J.D.); (M.E.G.); (J.W.T.); (C.O.K.)
| | - Mariah E. Geisen
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA; (J.D.); (M.E.G.); (J.W.T.); (C.O.K.)
| | - Josiane Weber Tessmann
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA; (J.D.); (M.E.G.); (J.W.T.); (C.O.K.)
| | - Piotr G. Rychahou
- Department of Surgery, University of Kentucky, Lexington, KY 40536, USA; (P.G.R.); (B.M.E.)
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Courtney O. Kelson
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA; (J.D.); (M.E.G.); (J.W.T.); (C.O.K.)
| | - Daheng He
- Markey Cancer Center Biostatistics and Bioinformatics Shared Resource Facility, University of Kentucky, Lexington, KY 40536, USA; (D.H.); (C.W.)
| | - Chi Wang
- Markey Cancer Center Biostatistics and Bioinformatics Shared Resource Facility, University of Kentucky, Lexington, KY 40536, USA; (D.H.); (C.W.)
| | - B. Mark Evers
- Department of Surgery, University of Kentucky, Lexington, KY 40536, USA; (P.G.R.); (B.M.E.)
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Yekaterina Y. Zaytseva
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA; (J.D.); (M.E.G.); (J.W.T.); (C.O.K.)
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
10
|
Jun SY, Cho S, Kim MJ, Park JW, Ryoo SB, Jeong SY, Park KJ, Shin A. Glycemic traits and colorectal cancer survival in a cohort of South Korean patients: A Mendelian randomization analysis. Cancer Med 2024; 13:e7084. [PMID: 38477501 DOI: 10.1002/cam4.7084] [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: 09/07/2023] [Revised: 02/13/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Clinical diabetic traits have been reported to be associated with increased colorectal cancer (CRC) risk in observational studies. Using the Mendelian randomization (MR) analysis method, we examined the causal association between glycemic traits, such as fasting glucose (FG), fasting insulin (FI), and glycosylated hemoglobin A1c (HbA1c), and survival in a cohort of CRC patients. METHODS We conducted a two-sample MR analysis among a cohort of patients with locally advanced CRC at Seoul National University Hospital. Single-nucleotide polymorphisms robustly associated (p < 5 × 10-8 ) with the three glycemic traits were obtained from the Meta-Analyses of Glucose and Insulin-related traits Consortium, Asian Genetic Epidemiology Network, and Korea Biobank Array. Three-year and 5-year overall survival (OS) and progression-free survival (PFS) were used as outcomes. Survival analysis was conducted using subgroup analysis by cancer stage and subsite in a multivariate Cox proportional hazards model adjusted for age and sex to examine whether glycemic traits affected survival. RESULTS A total of 509 patients were included in our final analysis. MR analysis showed that HbA1c levels were associated with poor 3-year OS (β = 4.20, p = 0.02). Sensitivity analyses did not show evidence of any violations of the MR assumptions. In the cancer subgroup analysis of the Cox proportional hazards model, pooled hazard ratios for FG were significantly associated with poor 3-year OS and PFS regardless of cancer stage. FI was not significantly associated with any 3-year survival endpoints. Among Stage III patients, three glycemic traits were significantly associated with both 5-year OS and PFS. Location-specific subgroup analysis showed a significant association between three glycemic traits and 5-year PFS in patients with left-sided colon cancer. FG was associated with poor 3-year survival for colon cancer but not rectal cancer. CONCLUSIONS Our results suggest that FG and HbA1c could be used to predict prognosis in CRC patients. Lifestyle and/or pharmacological interventions targeting glycemic traits could help improve survival for CRC patients.
Collapse
Affiliation(s)
- So Yon Jun
- Department of Preventative Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sooyoung Cho
- Department of Preventative Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Medical Research Center, Genomic Medicine Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Min Jung Kim
- Department of Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Ji Won Park
- Department of Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Seung-Bum Ryoo
- Department of Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seung Yong Jeong
- Department of Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Kyu Joo Park
- Department of Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Aesun Shin
- Department of Preventative Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
- Medical Research Center, Genomic Medicine Institute, Seoul National University College of Medicine, Seoul, South Korea
- Integrated Major in Innovative Medical Science, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
11
|
Canha MI, Ramos G, Prata R, Lages Martins P, Viúla Ramos M, Coimbra J. Is Metformin Associated with a Lower Prevalence of Polyps, Adenomas, and Colorectal Carcinoma in Patients with Diabetes Mellitus? J Gastrointest Cancer 2024; 55:435-443. [PMID: 37987968 DOI: 10.1007/s12029-023-00989-2] [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] [Accepted: 11/13/2023] [Indexed: 11/22/2023]
Abstract
PURPOSE Recent studies suggested a protective role of metformin in the development of colorectal cancer (CRC) and its precursors. We aimed to investigate if metformin was associated with a lower prevalence and number of colorectal polyps in diabetic patients and also adenomas, high-risk adenomas, and CRC. METHODS Retrospective study on adult patients with diabetes mellitus followed in our hospital with a total colonoscopy between 2015 and 2019, treated with either metformin for > 5 years or other antidiabetic agent (control group). We assessed the number, size, and histopathology examination of proliferative lesions detected on colonoscopy. RESULTS We included 401 patients aged 69 ± 9 years, 57% males, divided into two groups: treated with metformin (n = 260) and without (n = 141). The number of polyps detected was significantly lower in patients under metformin (p = 0.014). There was a nonsignificant trend towards lower polyp detection rates in the metformin compared to the control group both in unadjusted analysis (50% vs 60%, p = 0.058) and multivariable adjusted analysis (odds ratio [OR] 0.68, 95% confidence interval [CI] 0.43-1.09, p = 0.111). In the latter, we identified male gender (OR 2.24, 95%CI 1.44-3.49, p < 0.001), age (OR 1.35 for every 10 years, 95%CI 1.07-1.71, p = 0.012), glycated hemoglobin value (OR 1.20 for every 1% increase, 95%CI 1.06-1.37, p = 0.005), and hypertension (OR 1.76, 95%CI 1.01-3.08, p = 0.046) as factors associated with a higher prevalence of polyps. We saw no statistically significant differences regarding adenoma (p = 0.231), high-risk adenoma (p = 0.810), and CRC (p = 0.705) diagnoses between groups. CONCLUSION In our study, metformin was associated with less colorectal polyps in diabetic patients compared to other treatment modalities. We observed a nonsignificant trend towards lower polyp detection rates in the metformin group both in unadjusted and adjusted analyses.
Collapse
Affiliation(s)
- Maria Inês Canha
- Gastroenterology Department of Centro Hospitalar Universitário de Lisboa Central, EPE, Lisbon, Portugal.
- NOVA Medical School, Lisbon, Portugal.
| | - Gonçalo Ramos
- Gastroenterology Department of Centro Hospitalar Universitário de Lisboa Central, EPE, Lisbon, Portugal
- NOVA Medical School, Lisbon, Portugal
| | - Rita Prata
- Gastroenterology Department of Centro Hospitalar Universitário de Lisboa Central, EPE, Lisbon, Portugal
| | - Pedro Lages Martins
- Gastroenterology Department of Centro Hospitalar Universitário de Lisboa Central, EPE, Lisbon, Portugal
- NOVA Medical School, Lisbon, Portugal
| | - Marta Viúla Ramos
- Gastroenterology Department of Centro Hospitalar Universitário de Lisboa Central, EPE, Lisbon, Portugal
| | - João Coimbra
- Gastroenterology Department of Centro Hospitalar Universitário de Lisboa Central, EPE, Lisbon, Portugal
- NOVA Medical School, Lisbon, Portugal
| |
Collapse
|
12
|
Liu X, Cai YD, Chiu JC. Regulation of protein O-GlcNAcylation by circadian, metabolic, and cellular signals. J Biol Chem 2024; 300:105616. [PMID: 38159854 PMCID: PMC10810748 DOI: 10.1016/j.jbc.2023.105616] [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: 07/24/2023] [Revised: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 01/03/2024] Open
Abstract
O-linked β-N-acetylglucosamine (O-GlcNAcylation) is a dynamic post-translational modification that regulates thousands of proteins and almost all cellular processes. Aberrant O-GlcNAcylation has been associated with numerous diseases, including cancer, neurodegenerative diseases, cardiovascular diseases, and type 2 diabetes. O-GlcNAcylation is highly nutrient-sensitive since it is dependent on UDP-GlcNAc, the end product of the hexosamine biosynthetic pathway (HBP). We previously observed daily rhythmicity of protein O-GlcNAcylation in a Drosophila model that is sensitive to the timing of food consumption. We showed that the circadian clock is pivotal in regulating daily O-GlcNAcylation rhythms given its control of the feeding-fasting cycle and hence nutrient availability. Interestingly, we reported that the circadian clock also modulates daily O-GlcNAcylation rhythm by regulating molecular mechanisms beyond the regulation of food consumption time. A large body of work now indicates that O-GlcNAcylation is likely a generalized cellular status effector as it responds to various cellular signals and conditions, such as ER stress, apoptosis, and infection. In this review, we summarize the metabolic regulation of protein O-GlcNAcylation through nutrient availability, HBP enzymes, and O-GlcNAc processing enzymes. We discuss the emerging roles of circadian clocks in regulating daily O-GlcNAcylation rhythm. Finally, we provide an overview of other cellular signals or conditions that impact O-GlcNAcylation. Many of these cellular pathways are themselves regulated by the clock and/or metabolism. Our review highlights the importance of maintaining optimal O-GlcNAc rhythm by restricting eating activity to the active period under physiological conditions and provides insights into potential therapeutic targets of O-GlcNAc homeostasis under pathological conditions.
Collapse
Affiliation(s)
- Xianhui Liu
- Department of Entomology and Nematology, College of Agricultural and Environmental Sciences, University of California, Davis, California, USA
| | - Yao D Cai
- Department of Entomology and Nematology, College of Agricultural and Environmental Sciences, University of California, Davis, California, USA
| | - Joanna C Chiu
- Department of Entomology and Nematology, College of Agricultural and Environmental Sciences, University of California, Davis, California, USA.
| |
Collapse
|
13
|
Scheper AF, Schofield J, Bohara R, Ritter T, Pandit A. Understanding glycosylation: Regulation through the metabolic flux of precursor pathways. Biotechnol Adv 2023; 67:108184. [PMID: 37290585 DOI: 10.1016/j.biotechadv.2023.108184] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 05/29/2023] [Accepted: 05/31/2023] [Indexed: 06/10/2023]
Abstract
Glycosylation is how proteins and lipids are modified with complex carbohydrates known as glycans. The post-translational modification of proteins with glycans is not a template-driven process in the same way as genetic transcription or protein translation. Glycosylation is instead dynamically regulated by metabolic flux. This metabolic flux is determined by the concentrations and activities of the glycotransferase enzymes, which synthesise glycans, the metabolites that act as their precursors and transporter proteins. This review provides an overview of the metabolic pathways underlying glycan synthesis. Pathological dysregulation of glycosylation, particularly increased glycosylation occurring during inflammation, is also elucidated. The resulting inflammatory hyperglycosylation acts as a glycosignature of disease, and we report on the changes in the metabolic pathways which feed into glycan synthesis, revealing alterations to key enzymes. Finally, we examine studies in developing metabolic inhibitors targeting these critical enzymes. These results provide the tools for researchers investigating the role of glycan metabolism in inflammation and have helped to identify promising glycotherapeutic approaches to inflammation.
Collapse
Affiliation(s)
- Aert F Scheper
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Ireland
| | - Jack Schofield
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Ireland
| | - Raghvendra Bohara
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Ireland
| | - Thomas Ritter
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Ireland; School of Medicine, University of Galway, Ireland; Regenerative Medicine Institute (REMEDI), University of Galway, Ireland
| | - Abhay Pandit
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Ireland.
| |
Collapse
|
14
|
Leonel AV, Alisson-Silva F, Santos RCM, Silva-Aguiar RP, Gomes JC, Longo GMC, Faria BM, Siqueira MS, Pereira MG, Vasconcelos-dos-Santos A, Chiarini LB, Slawson C, Caruso-Neves C, Romão L, Travassos LH, Carneiro K, Todeschini AR, Dias WB. Inhibition of O-GlcNAcylation Reduces Cell Viability and Autophagy and Increases Sensitivity to Chemotherapeutic Temozolomide in Glioblastoma. Cancers (Basel) 2023; 15:4740. [PMID: 37835434 PMCID: PMC10571858 DOI: 10.3390/cancers15194740] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/15/2023] [Accepted: 09/15/2023] [Indexed: 10/15/2023] Open
Abstract
Glioblastoma (GB) is the most aggressive primary malignant brain tumor and is associated with short survival. O-GlcNAcylation is an intracellular glycosylation that regulates protein function, enzymatic activity, protein stability, and subcellular localization. Aberrant O-GlcNAcylation is related to the tumorigenesis of different tumors, and mounting evidence supports O-GlcNAc transferase (OGT) as a potential therapeutic target. Here, we used two human GB cell lines alongside primary human astrocytes as a non-tumoral control to investigate the role of O-GlcNAcylation in cell proliferation, cell cycle, autophagy, and cell death. We observed that hyper O-GlcNAcylation promoted increased cellular proliferation, independent of alterations in the cell cycle, through the activation of autophagy. On the other hand, hypo O-GlcNAcylation inhibited autophagy, promoted cell death by apoptosis, and reduced cell proliferation. In addition, the decrease in O-GlcNAcylation sensitized GB cells to the chemotherapeutic temozolomide (TMZ) without affecting human astrocytes. Combined, these results indicated a role for O-GlcNAcylation in governing cell proliferation, autophagy, cell death, and TMZ response, thereby indicating possible therapeutic implications for treating GB. These findings pave the way for further research and the development of novel treatment approaches which may contribute to improved outcomes and increased survival rates for patients facing this challenging disease.
Collapse
Affiliation(s)
- Amanda V. Leonel
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (A.V.L.); (C.C.-N.); (L.H.T.); (A.R.T.)
| | - Frederico Alisson-Silva
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Ronan C. M. Santos
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (A.V.L.); (C.C.-N.); (L.H.T.); (A.R.T.)
| | - Rodrigo P. Silva-Aguiar
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (A.V.L.); (C.C.-N.); (L.H.T.); (A.R.T.)
| | - Julia C. Gomes
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (A.V.L.); (C.C.-N.); (L.H.T.); (A.R.T.)
| | - Gabriel M. C. Longo
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-590, RJ, Brazil
| | - Bruna M. Faria
- Instituto de Ciências Biomédicas (ICB), Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil (L.R.); (K.C.)
| | - Mariana S. Siqueira
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (A.V.L.); (C.C.-N.); (L.H.T.); (A.R.T.)
| | - Miria G. Pereira
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (A.V.L.); (C.C.-N.); (L.H.T.); (A.R.T.)
| | - Andreia Vasconcelos-dos-Santos
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (A.V.L.); (C.C.-N.); (L.H.T.); (A.R.T.)
| | - Luciana B. Chiarini
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (A.V.L.); (C.C.-N.); (L.H.T.); (A.R.T.)
| | - Chad Slawson
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Celso Caruso-Neves
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (A.V.L.); (C.C.-N.); (L.H.T.); (A.R.T.)
| | - Luciana Romão
- Instituto de Ciências Biomédicas (ICB), Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil (L.R.); (K.C.)
| | - Leonardo H. Travassos
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (A.V.L.); (C.C.-N.); (L.H.T.); (A.R.T.)
| | - Katia Carneiro
- Instituto de Ciências Biomédicas (ICB), Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil (L.R.); (K.C.)
| | - Adriane R. Todeschini
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (A.V.L.); (C.C.-N.); (L.H.T.); (A.R.T.)
| | - Wagner B. Dias
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (A.V.L.); (C.C.-N.); (L.H.T.); (A.R.T.)
| |
Collapse
|
15
|
Si HR, Sun SS, Liu YK, Qiu LY, Tang B, Liu F, Fu Q, Xu CD, Wan PJ. Roles of GFAT and PFK genes in energy metabolism of brown planthopper, Nilaparvata lugens. Front Physiol 2023; 14:1213654. [PMID: 37415905 PMCID: PMC10320585 DOI: 10.3389/fphys.2023.1213654] [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: 04/28/2023] [Accepted: 06/13/2023] [Indexed: 07/08/2023] Open
Abstract
Glutamine:fructose-6-phosphate aminotransferases (GFATs) and phosphofructokinase (PFKs) are the principal rate-limiting enzymes involved in hexosamine biosynthesis pathway (HBP) and glycolysis pathway, respectively. In this study, the NlGFAT and NlPFK were knocked down through RNA interference (RNAi) in Nilaparvata lugens, the notorious brown planthopper (BPH), and the changes in energy metabolism were determined. Knockdown of either NlGFAT or NlPFK substantially reduced gene expression related to trehalose, glucose, and glycogen metabolism pathways. Moreover, trehalose content rose significantly at 72 h after dsGFAT injection, and glycogen content increased significantly at 48 h after injection. Glucose content remained unchanged throughout the experiment. Conversely, dsPFK injection did not significantly alter trehalose, but caused an extreme increase in glucose and glycogen content at 72 h after injection. The Knockdown of NlGFAT or NlPFK significantly downregulated the genes in the glycolytic pathway, as well as caused a considerable and significant decrease in pyruvate kinase (PK) activity after 48 h and 72 h of inhibition. After dsGFAT injection, most of genes in TCA cycle pathway were upregulated, but after dsNlPFK injection, they were downregulated. Correspondingly, ATP content substantially increased at 48 h after NlGFAT knockdown but decreased to an extreme extent by 72 h. In contrast, ATP content decreased significantly after NlPFK was knocked down and returned. The results have suggested the knockdown of either NlGFAT or NlPFK resulted in metabolism disorders in BPHs, highlighting the difference in the impact of those two enzyme genes on energy metabolism. Given their influence on BPHs energy metabolism, developing enzyme inhibitors or activators may provide a biological control for BPHs.
Collapse
Affiliation(s)
- Hui-Ru Si
- State Key Laboratory of Rice Biology and Breeding, China National Rice Research Institute, Hangzhou, Zhejiang, China
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Si-Si Sun
- Guizhou Institute of Mountainous Environment and Climate, Guiyang, China
| | - Yong-Kang Liu
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Ling-Yu Qiu
- State Key Laboratory of Rice Biology and Breeding, China National Rice Research Institute, Hangzhou, Zhejiang, China
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Bin Tang
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Fang Liu
- State Key Laboratory of Rice Biology and Breeding, China National Rice Research Institute, Hangzhou, Zhejiang, China
| | - Qiang Fu
- State Key Laboratory of Rice Biology and Breeding, China National Rice Research Institute, Hangzhou, Zhejiang, China
| | - Cai-Di Xu
- Jing Hengyi School of Education, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Pin-Jun Wan
- State Key Laboratory of Rice Biology and Breeding, China National Rice Research Institute, Hangzhou, Zhejiang, China
| |
Collapse
|
16
|
Paneque A, Fortus H, Zheng J, Werlen G, Jacinto E. The Hexosamine Biosynthesis Pathway: Regulation and Function. Genes (Basel) 2023; 14:genes14040933. [PMID: 37107691 PMCID: PMC10138107 DOI: 10.3390/genes14040933] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/13/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
The hexosamine biosynthesis pathway (HBP) produces uridine diphosphate-N-acetyl glucosamine, UDP-GlcNAc, which is a key metabolite that is used for N- or O-linked glycosylation, a co- or post-translational modification, respectively, that modulates protein activity and expression. The production of hexosamines can occur via de novo or salvage mechanisms that are catalyzed by metabolic enzymes. Nutrients including glutamine, glucose, acetyl-CoA, and UTP are utilized by the HBP. Together with availability of these nutrients, signaling molecules that respond to environmental signals, such as mTOR, AMPK, and stress-regulated transcription factors, modulate the HBP. This review discusses the regulation of GFAT, the key enzyme of the de novo HBP, as well as other metabolic enzymes that catalyze the reactions to produce UDP-GlcNAc. We also examine the contribution of the salvage mechanisms in the HBP and how dietary supplementation of the salvage metabolites glucosamine and N-acetylglucosamine could reprogram metabolism and have therapeutic potential. We elaborate on how UDP-GlcNAc is utilized for N-glycosylation of membrane and secretory proteins and how the HBP is reprogrammed during nutrient fluctuations to maintain proteostasis. We also consider how O-GlcNAcylation is coupled to nutrient availability and how this modification modulates cell signaling. We summarize how deregulation of protein N-glycosylation and O-GlcNAcylation can lead to diseases including cancer, diabetes, immunodeficiencies, and congenital disorders of glycosylation. We review the current pharmacological strategies to inhibit GFAT and other enzymes involved in the HBP or glycosylation and how engineered prodrugs could have better therapeutic efficacy for the treatment of diseases related to HBP deregulation.
Collapse
Affiliation(s)
- Alysta Paneque
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Harvey Fortus
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Julia Zheng
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Guy Werlen
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Estela Jacinto
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| |
Collapse
|
17
|
Epigenetic regulation of Neuregulin 1 promotes breast cancer progression associated to hyperglycemia. Nat Commun 2023; 14:439. [PMID: 36707514 PMCID: PMC9883495 DOI: 10.1038/s41467-023-36179-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 01/18/2023] [Indexed: 01/28/2023] Open
Abstract
Hyperglycemia is a risk factor for breast cancer-related morbidity and mortality. Hyperglycemia induces Neuregulin 1 (Nrg1) overexpression in breast cancer, which subsequently promotes tumor progression. However, molecular mechanisms underlying hyperglycemia-induced Nrg1 overexpression remain poorly understood. Here, we show that hyperglycemia causes active histone modifications at the Nrg1 enhancer, forming enhanceosome complexes where recombination signal binding protein for immunoglobulin kappa J region (RBPJ), E1A binding protein p300 (P300), and SET domain containing 1 A (SETD1A) are recruited to upregulate Nrg1 expression. Deletions in RBPJ-binding sites causes hyperglycemia-controlled Nrg1 levels to be downregulated, resulting in decreased tumor growth in vitro and in vivo. Mice with modest-temporary hyperglycemia, induced by low-dose short-exposure streptozotocin, display accelerated tumor growth and lapatinib resistance, whereas combining lapatinib with N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S42 phenylglycine t-butyl ester (DAPT) ameliorates tumor growth under these modest hyperglycemic conditions by inhibiting NOTCH and EGFR superfamilies. NOTCH activity is correlated with NRG1 levels, and high NRG1 levels predicts poor outcomes, particularly in HER2-positive breast cancer patients. Our findings highlight the hyperglycemia-linked epigenetic modulation of NRG1 as a potential therapeutic strategy for treating breast cancer patients with diabetes.
Collapse
|
18
|
de-Souza-Ferreira M, Ferreira ÉE, de-Freitas-Junior JCM. Aberrant N-glycosylation in cancer: MGAT5 and β1,6-GlcNAc branched N-glycans as critical regulators of tumor development and progression. Cell Oncol 2023; 46:481-501. [PMID: 36689079 DOI: 10.1007/s13402-023-00770-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2023] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Changes in protein glycosylation are widely observed in tumor cells. N-glycan branching through adding β1,6-linked N-acetylglucosamine (β1,6-GlcNAc) to an α1,6-linked mannose, which is catalyzed by the N-acetylglucosaminyltransferase V (MGAT5 or GnT-V), is one of the most frequently observed tumor-associated glycan structure formed. Increased levels of this branching structure play a pro-tumoral role in various ways, for example, through the stabilization of growth factor receptors, the destabilization of intercellular adhesion, or the acquisition of a migratory phenotype. CONCLUSION In this review, we provide an updated and comprehensive summary of the physiological and pathophysiological roles of MGAT5 and β1,6-GlcNAc branched N-glycans, including their regulatory mechanisms. Specific emphasis is given to the role of MGAT5 and β1,6-GlcNAc branched N-glycans in cellular mechanisms that contribute to the development and progression of solid tumors. We also provide insight into possible future clinical implications, such as the use of MGAT5 as a prognostic biomarker.
Collapse
Affiliation(s)
- Michelle de-Souza-Ferreira
- Cellular and Molecular Oncobiology Program, Cancer Glycobiology Group, Brazilian National Cancer Institute (INCA), 37 André Cavalcanti Street, Rio de Janeiro, RJ, 20231-050, Brazil
| | - Érika Elias Ferreira
- Cellular and Molecular Oncobiology Program, Cancer Glycobiology Group, Brazilian National Cancer Institute (INCA), 37 André Cavalcanti Street, Rio de Janeiro, RJ, 20231-050, Brazil
| | - Julio Cesar Madureira de-Freitas-Junior
- Cellular and Molecular Oncobiology Program, Cancer Glycobiology Group, Brazilian National Cancer Institute (INCA), 37 André Cavalcanti Street, Rio de Janeiro, RJ, 20231-050, Brazil.
| |
Collapse
|
19
|
Dysregulation of hexosamine biosynthetic pathway wiring metabolic signaling circuits in cancer. Biochim Biophys Acta Gen Subj 2023; 1867:130250. [PMID: 36228878 DOI: 10.1016/j.bbagen.2022.130250] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/28/2022] [Accepted: 09/29/2022] [Indexed: 11/13/2022]
Abstract
Metabolite sensing, a fundamental biological process, plays a key role in metabolic signaling circuit rewiring. Hexosamine biosynthetic pathway (HBP) is a glucose metabolic pathway essential for the synthesis of uridine diphosphate N-acetylglucosamine (UDP-GlcNAc), which senses key nutrients and integrally maintains cellular homeostasis. UDP-GlcNAc dynamically regulates protein N-glycosylation and O-linked-N-acetylglucosamine modification (O-GlcNAcylation). Dysregulated HBP flux leads to abnormal protein glycosylation, and contributes to cancer development and progression by affecting protein function and cellular signaling. Furthermore, O-GlcNAcylation regulates cellular signaling pathways, and its alteration is linked to various cancer characteristics. Additionally, recent findings have suggested a close association between HBP stimulation and cancer stemness; an elevated HBP flux promotes cancer cell conversion to cancer stem cells and enhances chemotherapy resistance via downstream signal activation. In this review, we highlight the prominent roles of HBP in metabolic signaling and summarize the recent advances in HBP and its downstream signaling, relevant to cancer.
Collapse
|
20
|
Harvey DJ. Analysis of carbohydrates and glycoconjugates by matrix-assisted laser desorption/ionization mass spectrometry: An update for 2017-2018. MASS SPECTROMETRY REVIEWS 2023; 42:227-431. [PMID: 34719822 DOI: 10.1002/mas.21721] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 07/26/2021] [Accepted: 07/26/2021] [Indexed: 06/13/2023]
Abstract
This review is the tenth update of the original article published in 1999 on the application of matrix-assisted laser desorption/ionization mass spectrometry (MALDI) mass spectrometry to the analysis of carbohydrates and glycoconjugates and brings coverage of the literature to the end of 2018. Also included are papers that describe methods appropriate to glycan and glycoprotein analysis by MALDI, such as sample preparation techniques, even though the ionization method is not MALDI. Topics covered in the first part of the review include general aspects such as theory of the MALDI process, new methods, matrices, derivatization, MALDI imaging, fragmentation and the use of arrays. The second part of the review is devoted to applications to various structural types such as oligo- and poly-saccharides, glycoproteins, glycolipids, glycosides, and biopharmaceuticals. Most of the applications are presented in tabular form. The third part of the review covers medical and industrial applications of the technique, studies of enzyme reactions, and applications to chemical synthesis. The reported work shows increasing use of combined new techniques such as ion mobility and highlights the impact that MALDI imaging is having across a range of diciplines. MALDI is still an ideal technique for carbohydrate analysis and advancements in the technique and the range of applications continue steady progress.
Collapse
Affiliation(s)
- David J Harvey
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, UK
| |
Collapse
|
21
|
Foreman M, Patel A, Sheth S, Reddy A, Lucke-Wold B. Diabetes Mellitus Management in the Context of Cranial Tumors. BOHR INTERNATIONAL JOURNAL OF NEUROLOGY AND NEUROSCIENCE 2022; 1:29-39. [PMID: 36700856 PMCID: PMC9872258 DOI: 10.54646/bijnn.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The study of the relationship between cancer and diabetes mellitus (DM) has been under investigation for many decades. Particularly in the field of neurology and neurosurgery, increasing emphasis has been put on the examination of comorbid DM in patients with cranial tumors. Namely, as the most common and invasive type of malignant adult brain tumor, glioblastoma (GBS) has been the focus of said research. Several mechanisms have been described in the attempt to elucidate the underlying association between DM and GBS, with the metabolic phenomenon known as the Warburg effect and its consequential downstream effects serving as the resounding culprits in recent literature. Since the effect seen in cancers like GBS exploits an upregulated form of aerobic glycolysis, the role of a sequela of DM, known as hyperglycemia, will be investigated. In particular, in the treatment of GBS, surgical resection and subsequent chemotherapy and/or radiotherapy are used in conjunction with corticosteroid therapy, the latter of which has been linked to hyperglycemia. Unsurprisingly, comorbid DM patients are significantly susceptible to this disposition. Further, this fact is reflected in recent literature that demonstrates the impact of hyperglycemia on cancer advancement and patient outcomes in several preclinical and clinical studies. Thus, this review will aim to underline the significance of diabetes and glycemic control via standard-of-care treatments such as metformin administration, as well as to describe emerging treatments such as the signaling modulation of insulin-like growth factor and the employment of the ketogenic diet.
Collapse
Affiliation(s)
- Marco Foreman
- Department of Neurosurgery, University of Florida, Gainesville, Florida, United States
| | - Aashay Patel
- Department of Neurosurgery, University of Florida, Gainesville, Florida, United States
| | - Sohum Sheth
- Department of Neurosurgery, University of Florida, Gainesville, Florida, United States
| | - Akshay Reddy
- Department of Neurosurgery, University of Florida, Gainesville, Florida, United States
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, Florida, United States
| |
Collapse
|
22
|
Stefaniak J, Nowak MG, Wojciechowski M, Milewski S, Skwarecki AS. Inhibitors of glucosamine-6-phosphate synthase as potential antimicrobials or antidiabetics - synthesis and properties. J Enzyme Inhib Med Chem 2022; 37:1928-1956. [PMID: 35801410 PMCID: PMC9272926 DOI: 10.1080/14756366.2022.2096018] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Glucosamine-6-phosphate synthase (GlcN-6-P synthase) is known as a promising target for antimicrobial agents and antidiabetics. Several compounds of natural or synthetic origin have been identified as inhibitors of this enzyme. This set comprises highly selective l-glutamine, amino sugar phosphate or transition state intermediate cis-enolamine analogues. Relatively low antimicrobial activity of these inhibitors, poorly penetrating microbial cell membranes, has been improved using the pro-drug approach. On the other hand, a number of heterocyclic and polycyclic compounds demonstrating antimicrobial activity have been presented as putative inhibitors of the enzyme, based on the results of molecular docking to GlcN-6-P synthase matrix. The most active compounds of this group could be considered promising leads for development of novel antimicrobial drugs or antidiabetics, provided their selective toxicity is confirmed.
Collapse
Affiliation(s)
- Joanna Stefaniak
- Department of Organic Chemistry and BioTechMed Center, Gdańsk University of Technology, Gdańsk, Poland
| | - Michał G Nowak
- Department of Organic Chemistry and BioTechMed Center, Gdańsk University of Technology, Gdańsk, Poland
| | - Marek Wojciechowski
- Department of Pharmaceutical Technology and Biochemistry and BioTechMed Center, Gdańsk University of Technology, Gdańsk, Poland
| | - Sławomir Milewski
- Department of Pharmaceutical Technology and Biochemistry and BioTechMed Center, Gdańsk University of Technology, Gdańsk, Poland
| | - Andrzej S Skwarecki
- Department of Pharmaceutical Technology and Biochemistry and BioTechMed Center, Gdańsk University of Technology, Gdańsk, Poland
| |
Collapse
|
23
|
Silva-Aguiar RP, Peruchetti DB, Pinheiro AAS, Caruso-Neves C, Dias WB. O-GlcNAcylation in Renal (Patho)Physiology. Int J Mol Sci 2022; 23:ijms231911260. [PMID: 36232558 PMCID: PMC9569498 DOI: 10.3390/ijms231911260] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 12/29/2022] Open
Abstract
Kidneys maintain internal milieu homeostasis through a well-regulated manipulation of body fluid composition. This task is performed by the correlation between structure and function in the nephron. Kidney diseases are chronic conditions impacting healthcare programs globally, and despite efforts, therapeutic options for its treatment are limited. The development of chronic degenerative diseases is associated with changes in protein O-GlcNAcylation, a post-translation modification involved in the regulation of diverse cell function. O-GlcNAcylation is regulated by the enzymatic balance between O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA) which add and remove GlcNAc residues on target proteins, respectively. Furthermore, the hexosamine biosynthetic pathway provides the substrate for protein O-GlcNAcylation. Beyond its physiological role, several reports indicate the participation of protein O-GlcNAcylation in cardiovascular, neurodegenerative, and metabolic diseases. In this review, we discuss the impact of protein O-GlcNAcylation on physiological renal function, disease conditions, and possible future directions in the field.
Collapse
Affiliation(s)
- Rodrigo P. Silva-Aguiar
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
| | - Diogo B. Peruchetti
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
| | - Ana Acacia S. Pinheiro
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
- Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro 21045-900, Brazil
| | - Celso Caruso-Neves
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
- Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro 21045-900, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro 21941-902, Brazil
| | - Wagner B. Dias
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
- Correspondence:
| |
Collapse
|
24
|
Cui Y, Feng H, Liu J, Wu J, Zhu R, Huang R, Yan J. Identification of hexosamine biosynthesis pathway as a novel prognostic signature and its correlation with immune infiltration in bladder cancer. Front Mol Biosci 2022; 9:1009168. [PMID: 36158580 PMCID: PMC9493074 DOI: 10.3389/fmolb.2022.1009168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 08/16/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Urinary bladder cancer (UBC) is one of the common urological malignancies, lacking reliable biomarkers to predict clinical outcomes in UBC patients. Thus, it is needed to identify the novel diagnostic/prognostic biomarkers to stratify the high-risk UBC patients. As a shunt pathway of glycolysis, the hexosamine biosynthesis pathway (HBP) has been implicated in carcinogenesis. However, its prognostic value in UBC remains unclear. Methods: The RNA sequencing and mRNA microarray datasets were downloaded from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus databases. The expression levels of five HBP genes were analyzed in normal and UBC samples, and their associations with stage, grade and survival were plotted. The performance of HBP risk group was evaluated by receiver-operating characteristics (ROC) curve. The HBP signature was generated by Gene Set Variation Analysis (GSVA) and its association with clinicopathological parameters and survival were analyzed. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were carried out to examine the potential biological functions of HBP using DAVID online tool. The infiltration estimation fraction of immune cells was performed using CIBERSORT-ABS algorithm. Gene set enrichment analysis (GSEA) was used to explore the potential function of HBP in tumor immunoregulation. Results: Four HBP genes were upregulated in UBCs compared to normal tissues in TCGA-BLCA dataset. The upregulation of all five HBP genes was significantly associated with tumor grade and stage of UBC in three independent UBC datasets. The expression of HBP genes predicted poor clinical outcomes in UBC patients in both TCGA-BLCA and GSE13507 datasets. The high-risk group based on HBP genes showed a poor prognosis. Furthermore, HBP signature was positively associated with tumor grade and stage in TCGA-BLCA dataset and with tumor grade, stage, distal metastasis and poor survival in GSE13507 dataset. Interestingly, high-HBP signature group exhibited a high infiltration of immune cells, particularly the macrophage population. Conclusion: We identified that HBP was a promising prognostic biomarker in UBC patients and strongly associated with immune infiltration.
Collapse
Affiliation(s)
- Yangyan Cui
- Model Animal Research Center, Nanjing University, Nanjing, China
| | - Hanyi Feng
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jiakuan Liu
- Department of Laboratory Animal Science, Fudan University, Shanghai, China
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Jiajun Wu
- Department of Laboratory Animal Science, Fudan University, Shanghai, China
| | - Rujian Zhu
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
- *Correspondence: Rujian Zhu, ; Ruimin Huang, ; Jun Yan,
| | - Ruimin Huang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- *Correspondence: Rujian Zhu, ; Ruimin Huang, ; Jun Yan,
| | - Jun Yan
- Department of Laboratory Animal Science, Fudan University, Shanghai, China
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
- MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, China
- *Correspondence: Rujian Zhu, ; Ruimin Huang, ; Jun Yan,
| |
Collapse
|
25
|
Liu X, Chiu JC. Nutrient-sensitive protein O-GlcNAcylation shapes daily biological rhythms. Open Biol 2022; 12:220215. [PMID: 36099933 PMCID: PMC9470261 DOI: 10.1098/rsob.220215] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 08/17/2022] [Indexed: 11/12/2022] Open
Abstract
O-linked-N-acetylglucosaminylation (O-GlcNAcylation) is a nutrient-sensitive protein modification that alters the structure and function of a wide range of proteins involved in diverse cellular processes. Similar to phosphorylation, another protein modification that targets serine and threonine residues, O-GlcNAcylation occupancy on cellular proteins exhibits daily rhythmicity and has been shown to play critical roles in regulating daily rhythms in biology by modifying circadian clock proteins and downstream effectors. We recently reported that daily rhythm in global O-GlcNAcylation observed in Drosophila tissues is regulated via the integration of circadian and metabolic signals. Significantly, mistimed feeding, which disrupts coordination of these signals, is sufficient to dampen daily O-GlcNAcylation rhythm and is predicted to negatively impact animal biological rhythms and health span. In this review, we provide an overview of published and potential mechanisms by which metabolic and circadian signals regulate hexosamine biosynthetic pathway metabolites and enzymes, as well as O-GlcNAc processing enzymes to shape daily O-GlcNAcylation rhythms. We also discuss the significance of functional interactions between O-GlcNAcylation and other post-translational modifications in regulating biological rhythms. Finally, we highlight organ/tissue-specific cellular processes and molecular pathways that could be modulated by rhythmic O-GlcNAcylation to regulate time-of-day-specific biology.
Collapse
Affiliation(s)
- Xianhui Liu
- Department of Entomology and Nematology, College of Agricultural and Environmental Sciences, University of California Davis, Davis, CA, USA
- Department of Pharmacology, School of Medicine, University of California Davis, Davis, CA, USA
| | - Joanna C. Chiu
- Department of Entomology and Nematology, College of Agricultural and Environmental Sciences, University of California Davis, Davis, CA, USA
| |
Collapse
|
26
|
Smith ALM, Whitehall JC, Greaves LC. Mitochondrial
DNA
mutations in aging and cancer. Mol Oncol 2022; 16:3276-3294. [PMID: 35842901 PMCID: PMC9490137 DOI: 10.1002/1878-0261.13291] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/18/2022] [Accepted: 07/15/2022] [Indexed: 11/09/2022] Open
Abstract
Advancing age is a major risk factor for malignant transformation and the development of cancer. As such, over 50% of neoplasms occur in individuals over the age of 70. The pathologies of both ageing and cancer have been characterized by respective groups of molecular hallmarks, and while some features are divergent between the two pathologies, several are shared. Perturbed mitochondrial function is one such common hallmark, and this observation therefore suggests that mitochondrial alterations may be of significance in age‐related cancer development. There is now considerable evidence documenting the accumulation of somatic mitochondrial DNA (mtDNA) mutations in ageing human postmitotic and replicative tissues. Similarly, mutations of the mitochondrial genome have been reported in human cancers for decades. The plethora of functions in which mitochondria partake, such as oxidative phosphorylation, redox balance, apoptosis and numerous biosynthetic pathways, manifests a variety of ways in which alterations in mtDNA may contribute to tumour growth. However, the specific mechanisms by which mtDNA mutations contribute to tumour progression remain elusive and often contradictory. This review aims to consolidate current knowledge and describe future direction within the field.
Collapse
Affiliation(s)
- Anna LM Smith
- Wellcome Centre for Mitochondrial Research, Biosciences Institute Newcastle University Newcastle Upon Tyne NE2 4HH UK
| | - Julia C Whitehall
- Wellcome Centre for Mitochondrial Research, Biosciences Institute Newcastle University Newcastle Upon Tyne NE2 4HH UK
| | - Laura C Greaves
- Wellcome Centre for Mitochondrial Research, Biosciences Institute Newcastle University Newcastle Upon Tyne NE2 4HH UK
| |
Collapse
|
27
|
El-Sayed NNE, Almaneai NM, Ben Bacha A, El-Ashrey MK, Al-Zaben MI, Almarhoon ZM. Biological Evaluation, Molecular Docking Analyses, and ADME Profiling of Certain New Quinazolinones as Anti-colorectal Agents. ACS OMEGA 2022; 7:18443-18458. [PMID: 35694504 PMCID: PMC9178606 DOI: 10.1021/acsomega.2c00812] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 04/29/2022] [Indexed: 06/15/2023]
Abstract
Colorectal carcinogenesis is a complex process, which is linked to dysregulation of human secretory phospholipases A2 (hsPLA2-G-IIA, hsPLA2-G-V, and hsPLA2-G-X), proteases (cathepsin-B, collagenase, thrombin, elastase, and trypsin), carbohydrate hydrolyzing enzymes (α-amylase and α-glucosidase), and free radical generating enzyme (xanthine oxidoreductase (XOR)). Therefore, some new quinazolinones were synthesized and evaluated as inhibitors against this array of enzymes as well as cytotoxic agents on LoVo and HCT-116 cells of colorectal cancer. Compounds 3g, 10, 8, 3c, and 1c exhibited promising cytotoxic effects with IC50 values ranging from 206.07 to 459.79 μM. Nine compounds showed promising enzymatic inhibitory effects, 3b, 3d, 3f, 5, 1a, and 12 (α-amylase), 8 (thrombin, elastase and trypsin), 10 (hsPLA2-G-IIA and hsPLA2-G-V), and 3f (α-glucosidase and XOR). Therefore, the most active inhibitors, were subjected to validated molecular docking studies to identify their affinities and binding modes. The expected physicochemical and pharmacokinetic features of the active candidates, 1a, 1c, 3b, 3c, 3d, 3f, 3g, 5, 8, 10, and 12 were predicted using bioavailability radar charts and boiled-egg graphical representations along with the Lipinski rule of five filter. Collectively, these studies showed the significance of derivatives 1c, 3b, 3c, 3d, 8, 10, and 12 as lead scaffolds for further optimization to develop enzymes inhibitors and anti-colorectal agents.
Collapse
Affiliation(s)
- Nahed N. E. El-Sayed
- National
Organization for Drug Control and Research, Egyptian Drug Authority, 51 Wezaret El-Zerra Street, Giza 35521, Egypt
| | - Norah M. Almaneai
- Department
of Chemistry, College of Science, King Saud
University, P. O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Abir Ben Bacha
- Biochemistry
Department, College of Science, King Saud
University, P. O. Box 22452, Riyadh 11495, Saudi Arabia
- Laboratory
of Plant Biotechnology Applied to Crop Improvement, Faculty of Science
of Sfax, University of Sfax, Sfax 3038, Tunisia
| | - Mohamed K. El-Ashrey
- Pharmaceutical
Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr Elini Street, Cairo 11562, Egypt
- Medicinal
Chemistry Department, Faculty of Pharmacy, King Salman International University, Ras-Sedr, South Sinai, Egypt
| | - Maha I. Al-Zaben
- Department
of Chemistry, College of Science, King Saud
University, P. O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Zainab M. Almarhoon
- Department
of Chemistry, College of Science, King Saud
University, P. O. Box 2455, Riyadh 11451, Saudi Arabia
| |
Collapse
|
28
|
Metformin Enhancement of Therapeutic Effects of 5-Fluorouracil and Oxaliplatin in Colon Cancer Cells and Nude Mice. Biomedicines 2022; 10:biomedicines10050955. [PMID: 35625692 PMCID: PMC9138369 DOI: 10.3390/biomedicines10050955] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/16/2022] [Accepted: 04/19/2022] [Indexed: 02/06/2023] Open
Abstract
Studies have demonstrated that metformin has antitumor effects in addition to therapeutic effects on hyperglycemia; however, few studies have explored the effects of metformin in chemotherapy. Therefore, we hypothesized that the administration of metformin would enhance the therapeutic effects of 5-fluorouracil and oxaliplatin (FuOx) to inhibit the growth of colorectal cancer (CRC) cells in vitro and in vivo. The results of our in vitro experiments demonstrated that metformin significantly increased the effects of FuOx with respect to cell proliferation (p < 0.05), colony formation (p < 0.05), and migration (p < 0.01) and induced cell cycle arrest in the G0/G1 phase in HT29 cells and the S phase in SW480 and SW620 cells (p < 0.05). Flow cytometry analysis revealed that metformin combined with FuOx induced late apoptosis (p < 0.05) by mediating mitochondria-related Mcl-1 and Bim protein expression. Furthermore, in vivo, metformin combined with FuOx more notably reduced tumor volume than FuOx or metformin alone did in BALB/c mice (p < 0.05). These findings demonstrate that metformin may act as an adjunctive agent to enhance the chemosensitivity of CRC cells to FuOx. However, further clinical trials are warranted to validate the clinical implications of the findings.
Collapse
|
29
|
Loponte HF, Oliveira IA, Rodrigues BC, Nunes-da-Fonseca R, Mohana-Borges R, Alisson-Silva F, Dias WB, Todeschini AR. Hyperglycemia alters N-glycans on colon cancer cells through increased production of activated monosaccharides. Glycoconj J 2022; 39:663-675. [PMID: 35380345 DOI: 10.1007/s10719-022-10057-9] [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: 12/23/2021] [Revised: 02/26/2022] [Accepted: 03/09/2022] [Indexed: 12/01/2022]
Abstract
Diabetes Mellitus (DM) is both, correlated and a known risk factor for colorectal cancer (CRC). Besides favoring the incidence of CRC, DM also accelerates its progression, worsening its prognosis. Previously, hyperglycemia, the DM hallmark, has been shown to lead to aberrant glycosylation of CRC cells, heightening their malignancy both in vivo and in vitro. Here we use mass spectrometry to elucidate the composition and putative structures of N-glycans expressed by MC38 cultured in normoglycemic (LG) and hyperglycemic-like conditions (HG). N-glycans, 67, were identified in MC38 cells cultured in LG and HG. The cells grown in HG showed a greater abundance of N-glycans when compared to LNG cells, without changes in the proportion of sialylated, fucosylated and mannosylated N-glycans. Among the identified N-glycans, 16 were differentially expressed, mostly mannosylated and fucosylated, with a minority of them being sialylated. Metabolomics analysis indicates that the alterations observed in the N-glycosylation may be mostly due to increase of the activated monosaccharides pool, through an increased glucose entrance into the cells. The alterations found here corroborate data from the literature regarding the progression of CRC, advocating for development or repositioning of effective treatments against CRC in diabetic patients.
Collapse
Affiliation(s)
- H F Loponte
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro, Brazil.,Instituto de Microbiologia Paulo de Goes, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro, Brazil
| | - I A Oliveira
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro, Brazil
| | - B C Rodrigues
- Instituto de Biodiversidade e Sustentabilidade, Universidade Federal do Rio de Janeiro, 27965‑550, Macaé, Brazil
| | - R Nunes-da-Fonseca
- Instituto de Biodiversidade e Sustentabilidade, Universidade Federal do Rio de Janeiro, 27965‑550, Macaé, Brazil
| | - R Mohana-Borges
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro, Brazil
| | - F Alisson-Silva
- Instituto de Microbiologia Paulo de Goes, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro, Brazil
| | - W B Dias
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro, Brazil
| | - A R Todeschini
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro, Brazil.
| |
Collapse
|
30
|
Liu YY, Liu HY, Yu TJ, Lu Q, Zhang FL, Liu GY, Shao ZM, Li DQ. O-GlcNAcylation of MORC2 at threonine 556 by OGT couples TGF-β signaling to breast cancer progression. Cell Death Differ 2022; 29:861-873. [PMID: 34974534 PMCID: PMC8991186 DOI: 10.1038/s41418-021-00901-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 11/03/2021] [Accepted: 11/10/2021] [Indexed: 12/11/2022] Open
Abstract
MORC family CW-type zinc finger 2 (MORC2) is a newly identified chromatin-remodeling enzyme involved in DNA damage response and gene transcription, and its dysregulation has been linked with Charcot-Marie-Tooth disease, neurodevelopmental disorder, and cancer. Despite its functional importance, how MORC2 is regulated remains enigmatic. Here, we report that MORC2 is O-GlcNAcylated by O-GlcNAc transferase (OGT) at threonine 556. Mutation of this site or pharmacological inhibition of OGT impairs MORC2-mediated breast cancer cell migration and invasion in vitro and lung colonization in vivo. Moreover, transforming growth factor-β1 (TGF-β1) induces MORC2 O-GlcNAcylation through enhancing the stability of glutamine-fructose-6-phosphate aminotransferase (GFAT), the rate-limiting enzyme for producing the sugar donor for OGT. O-GlcNAcylated MORC2 is required for transcriptional activation of TGF-β1 target genes connective tissue growth factor (CTGF) and snail family transcriptional repressor 1 (SNAIL). In support of these observations, knockdown of GFAT, SNAIL or CTGF compromises TGF-β1-induced, MORC2 O-GlcNAcylation-mediated breast cancer cell migration and invasion. Clinically, high expression of OGT, MORC2, SNAIL, and CTGF in breast tumors is associated with poor patient prognosis. Collectively, these findings uncover a previously unrecognized mechanistic role for MORC2 O-GlcNAcylation in breast cancer progression and provide evidence for targeting MORC2-dependent breast cancer through blocking its O-GlcNAcylation.
Collapse
Affiliation(s)
- Ying-Ying Liu
- grid.8547.e0000 0001 0125 2443Fudan University Shanghai Cancer Center and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032 China ,grid.8547.e0000 0001 0125 2443Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, 200032 China ,grid.8547.e0000 0001 0125 2443Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China ,grid.8547.e0000 0001 0125 2443Department of Breast Surgery, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Hong-Yi Liu
- grid.8547.e0000 0001 0125 2443Fudan University Shanghai Cancer Center and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032 China
| | - Tian-Jian Yu
- grid.8547.e0000 0001 0125 2443Fudan University Shanghai Cancer Center and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032 China ,grid.8547.e0000 0001 0125 2443Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, 200032 China ,grid.8547.e0000 0001 0125 2443Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China ,grid.8547.e0000 0001 0125 2443Department of Breast Surgery, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Qin Lu
- grid.8547.e0000 0001 0125 2443Fudan University Shanghai Cancer Center and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032 China
| | - Fang-Lin Zhang
- grid.8547.e0000 0001 0125 2443Fudan University Shanghai Cancer Center and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032 China ,grid.8547.e0000 0001 0125 2443Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, 200032 China ,grid.8547.e0000 0001 0125 2443Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Guang-Yu Liu
- Department of Breast Surgery, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Zhi-Ming Shao
- Fudan University Shanghai Cancer Center and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China. .,Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Department of Breast Surgery, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Shanghai Key Laboratory of Breast Cancer, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Da-Qiang Li
- Fudan University Shanghai Cancer Center and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China. .,Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Department of Breast Surgery, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Shanghai Key Laboratory of Breast Cancer, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Shanghai Key Laboratory of Radiation Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
31
|
Zhang Y, Cheng J, Li J, He J, Li X, Xu F. The GLP-1R Agonist Exendin-4 Attenuates Hyperglycemia-Induced Chemoresistance in Human Endometrial Cancer Cells Through ROS-Mediated Mitochondrial Pathway. Front Oncol 2022; 11:793530. [PMID: 34988025 PMCID: PMC8721044 DOI: 10.3389/fonc.2021.793530] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/01/2021] [Indexed: 12/23/2022] Open
Abstract
This study aimed to assess the effects of the antidiabetic drug Exendin-4 (Exe-4), a GLP-1 receptor agonist, on the response of human endometrial cancer cells to chemotherapy under high glucose (HG) conditions. Cell viability was detected using a cell counting kit (CCK)-8. Cell apoptosis and reactive oxygen species (ROS) levels were measured by flow cytometry. Gene expression was evaluated by real-time PCR and immunoblotting. The chemotherapeutic drug cisplatin (DDP) dose-dependently inhibited both human endometrial adenocarcinoma Ishikawa and HEC1B cells, a response reversed by HG. Meanwhile, Exe-4 attenuated hyperglycemia’s effect by elevating intracellular lactate dehydrogenase (LDH) and ROS production. Similarly, DDP-induced elevation of intracellular rhodamine123 was attenuated by HG, and Exe-4 reversed HG’s impact. The chemoresistance genes multidrug resistance-associated protein 1 (MRP1) and P-glycoprotein (Pgp) were upregulated. At the same time, topoisomerase II (TOPO II) was downregulated under HG conditions, suggesting HG-induced chemoresistance. Exe-4 did not significantly influence the above genes. DDP downregulated Bcl-2 and Bcl-XL and upregulated Bax, cytosolic cytochrome c, and PARP under normal glucose (NG) versus HG conditions, and Exe-4 attenuated these effects. Upstream of Bax/Bcl, acetylated P53 was upregulated by DDP and downregulated by HG, whose effect was reversed by Exe-4. DPP treatment significantly induced apoptosis and cell cycle arrest in the S phase under NG, and HG reduced these effects. Prolonged exposure to HG induces DDP chemoresistance in human endometrial cancer cells but is alleviated by Exe-4.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Gynecology, Third Affiliated Hospital, Sun-Yet Sen University, Guangzhou, China
| | - Juan Cheng
- Department of Gynecology, Third Affiliated Hospital, Sun-Yet Sen University, Guangzhou, China
| | - Jing Li
- Department of Gynecology, Third Affiliated Hospital, Sun-Yet Sen University, Guangzhou, China
| | - Junxian He
- Department of Gynecology, Third Affiliated Hospital, Sun-Yet Sen University, Guangzhou, China
| | - Xiaomao Li
- Department of Gynecology, Third Affiliated Hospital, Sun-Yet Sen University, Guangzhou, China
| | - Fen Xu
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Diabetology, Guangzhou, China
| |
Collapse
|
32
|
Writing and erasing O-GlcNAc from target proteins in cells. Biochem Soc Trans 2021; 49:2891-2901. [PMID: 34783346 DOI: 10.1042/bst20210865] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/25/2021] [Accepted: 10/27/2021] [Indexed: 12/15/2022]
Abstract
O-linked N-acetylglucosamine (O-GlcNAc) is a widespread reversible modification on nucleocytoplasmic proteins that plays an important role in many biochemical processes and is highly relevant to numerous human diseases. The O-GlcNAc modification has diverse functional impacts on individual proteins and glycosites, and methods for editing this modification on substrates are essential to decipher these functions. Herein, we review recent progress in developing methods for O-GlcNAc regulation, with a focus on methods for editing O-GlcNAc with protein- and site-selectivity in cells. The applications, advantages, and limitations of currently available strategies for writing and erasing O-GlcNAc and future directions are also discussed. These emerging approaches to manipulate O-GlcNAc on a target protein in cells will greatly accelerate the development of functional studies and enable therapeutic interventions in the O-GlcNAc field.
Collapse
|
33
|
Evans CA, Corfe BM. Colorectal keratins: Integrating nutrition, metabolism and colorectal health. Semin Cell Dev Biol 2021; 128:103-111. [PMID: 34481710 DOI: 10.1016/j.semcdb.2021.08.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 08/09/2021] [Accepted: 08/18/2021] [Indexed: 01/12/2023]
Abstract
The colon mucosa is lined with crypts of circa 300 cells, forming a continuous barrier whose roles include absorption of water, recovery of metabolic energy sources (notably short chain fatty acids), secretion of a protective mucus barrier, and physiological signalling. There is high turnover and replenishment of cells in the mucosa, disruption of this may lead to bowel pathologies including cancer and inflammatory bowel disease. Keratins have been implicated in the processes of cell death, epithelial integrity, response to inflammation and as a result are often described as guardians of the colonic epithelium. Keratin proteins carry extensive post-translational modifications, the cofactors for kinases, acetyl transferases and other modification-regulating enzymes are themselves products of metabolism. A cluster of studies has begun to reveal a bidirectional relationship between keratin form and function and metabolism. In this paper we hypothesise a mechanistic interaction between keratins and metabolism is governed through regulation of post-translational modifications and may contribute significantly to the normal functioning of the colon, placing keratins at the centre of a nutrition-metabolism-health triangle.
Collapse
Affiliation(s)
- Caroline A Evans
- ChELSI Institute, Department of Chemical and Biological Engineering, University of Sheffield, Mappin St, S1 3JD Sheffield, United Kingdom
| | - Bernard M Corfe
- Population Health Sciences Institute, Human Nutrition Research Centre, Faculty of Medical Sciences, Newcastle University, Newcastle NE2 4HH, United Kingdom.
| |
Collapse
|
34
|
Ganesh GV, Mohanram RK. Metabolic reprogramming and immune regulation in viral diseases. Rev Med Virol 2021; 32:e2268. [PMID: 34176174 DOI: 10.1002/rmv.2268] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 06/02/2021] [Accepted: 06/10/2021] [Indexed: 12/11/2022]
Abstract
The recent outbreak and transmission of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) worldwide and the ensuing coronavirus disease 2019 (COVID-19) pandemic has left us scrambling for ways to contain the disease and develop vaccines that are safe and effective. Equally important, understanding the impact of the virus on the host system in convalescent patients, healthy otherwise or with co-morbidities, is expected to aid in developing effective strategies in the management of patients afflicted with the disease. Viruses possess the uncanny ability to redirect host metabolism to serve their needs and also limit host immune response to ensure their survival. An ever-increasingly powerful approach uses metabolomics to uncover diverse molecular signatures that influence a wide array of host signalling networks in different viral infections. This would also help integrate experimental findings from individual studies to yield robust evidence. In addition, unravelling the molecular mechanisms harnessed by both viruses and tumours in their host metabolism will help broaden the repertoire of therapeutic tools available to combat viral disease.
Collapse
Affiliation(s)
- Goutham V Ganesh
- Life Science Division, SRM Research Institute and Department of Biotechnology, School of Bioengineering, SRM Institute of Science & Technology, Kattankulathur, Tamil Nadu, India
| | - Ramkumar K Mohanram
- Life Science Division, SRM Research Institute and Department of Biotechnology, School of Bioengineering, SRM Institute of Science & Technology, Kattankulathur, Tamil Nadu, India
| |
Collapse
|
35
|
Nikmaneshi MR, Firoozabadi B, Mozafari A. Chemo-mechanistic multi-scale model of a three-dimensional tumor microenvironment to quantify the chemotherapy response of cancer. Biotechnol Bioeng 2021; 118:3871-3887. [PMID: 34133020 DOI: 10.1002/bit.27863] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 06/02/2021] [Accepted: 06/10/2021] [Indexed: 02/03/2023]
Abstract
Exploring efficient chemotherapy would benefit from a deeper understanding of the tumor microenvironment (TME) and its role in tumor progression. As in vivo experimental methods are unable to isolate or control individual factors of the TME, and in vitro models often cannot include all the contributing factors, some questions are best addressed with mathematical models of systems biology. In this study, we establish a multi-scale mathematical model of the TME to simulate three-dimensional tumor growth and angiogenesis and then implement the model for an array of chemotherapy approaches to elucidate the effect of TME conditions and drug scheduling on controlling tumor progression. The hyperglycemic condition as the most common disorder for cancer patients is considered to evaluate its impact on cancer response to chemotherapy. We show that combining antiangiogenic and anticancer drugs improves the outcome of treatment and can decrease accumulation of the drug in normal tissue and enhance drug delivery to the tumor. Our results demonstrate that although both concurrent and neoadjuvant combination therapies can increase intratumoral drug exposure and therapeutic accuracy, neoadjuvant therapy surpasses this, especially against hyperglycemia. Our model provides mechanistic explanations for clinical observations of tumor progression and response to treatment and establishes a computational framework for exploring better treatment strategies.
Collapse
Affiliation(s)
| | - Bahar Firoozabadi
- Department of Mechanical Engineering, Sharif University of Technology, Tehran, Iran
| | - Aliasghar Mozafari
- Department of Mechanical Engineering, Sharif University of Technology, Tehran, Iran
| |
Collapse
|
36
|
Topel H, Bağırsakçı E, Yılmaz Y, Güneş A, Bağcı G, Çömez D, Kahraman E, Korhan P, Atabey N. High glucose induced c-Met activation promotes aggressive phenotype and regulates expression of glucose metabolism genes in HCC cells. Sci Rep 2021; 11:11376. [PMID: 34059694 PMCID: PMC8166976 DOI: 10.1038/s41598-021-89765-5] [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: 11/27/2020] [Accepted: 04/26/2021] [Indexed: 02/08/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is strongly associated with metabolic dysregulations/deregulations and hyperglycemia is a common metabolic disturbance in metabolic diseases. Hyperglycemia is defined to promote epithelial to mesenchymal transition (EMT) of cancer cells in various cancers but its molecular contribution to HCC progression and aggressiveness is relatively unclear. In this study, we analyzed the molecular mechanisms behind the hyperglycemia-induced EMT in HCC cell lines. Here, we report that high glucose promotes EMT through activating c-Met receptor tyrosine kinase via promoting its ligand-independent homodimerization. c-Met activation is critical for high glucose induced acquisition of mesenchymal phenotype, survival under high glucose stress and reprogramming of cellular metabolism by modulating glucose metabolism gene expression to promote aggressiveness in HCC cells. The crucial role of c-Met in high glucose induced EMT and aggressiveness may be the potential link between metabolic syndrome-related hepatocarcinogenesis and/or HCC progression. Considering c-Met inhibition in hyperglycemic patients would be an important complementary strategy for therapy that favors sensitization of HCC cells to therapeutics.
Collapse
Affiliation(s)
- Hande Topel
- Izmir Biomedicine and Genome Center (IBG), Balcova, 35340, Izmir, Turkey.,Department of Medical Biology and Genetics, Graduate School of Health Sciences, Dokuz Eylul University, Balcova, 35340, Izmir, Turkey
| | - Ezgi Bağırsakçı
- Izmir Biomedicine and Genome Center (IBG), Balcova, 35340, Izmir, Turkey.,Department of Molecular Biology and Genetics, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Balcova, 35340, Izmir, Turkey
| | - Yeliz Yılmaz
- Izmir Biomedicine and Genome Center (IBG), Balcova, 35340, Izmir, Turkey.,Department of Medical Biology and Genetics, Graduate School of Health Sciences, Dokuz Eylul University, Balcova, 35340, Izmir, Turkey
| | - Ayşim Güneş
- Izmir Biomedicine and Genome Center (IBG), Balcova, 35340, Izmir, Turkey
| | - Gülsün Bağcı
- Izmir Biomedicine and Genome Center (IBG), Balcova, 35340, Izmir, Turkey.,Department of Molecular Biology and Genetics, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Balcova, 35340, Izmir, Turkey
| | - Dehan Çömez
- Izmir Biomedicine and Genome Center (IBG), Balcova, 35340, Izmir, Turkey.,Department of Molecular Biology and Genetics, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Balcova, 35340, Izmir, Turkey
| | - Erkan Kahraman
- Izmir Biomedicine and Genome Center (IBG), Balcova, 35340, Izmir, Turkey.,Department of Medical Biology and Genetics, Graduate School of Health Sciences, Dokuz Eylul University, Balcova, 35340, Izmir, Turkey
| | - Peyda Korhan
- Izmir Biomedicine and Genome Center (IBG), Balcova, 35340, Izmir, Turkey
| | - Neşe Atabey
- Izmir Biomedicine and Genome Center (IBG), Balcova, 35340, Izmir, Turkey.
| |
Collapse
|
37
|
Alsheikh HAM, Metge BJ, Ha CM, Hinshaw DC, Mota MSV, Kammerud SC, Lama-Sherpa T, Sharafeldin N, Wende AR, Samant RS, Shevde LA. Normalizing glucose levels reconfigures the mammary tumor immune and metabolic microenvironment and decreases metastatic seeding. Cancer Lett 2021; 517:24-34. [PMID: 34052331 DOI: 10.1016/j.canlet.2021.05.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/09/2021] [Accepted: 05/21/2021] [Indexed: 12/29/2022]
Abstract
Obesity and diabetes cumulatively create a distinct systemic metabolic pathophysiological syndrome that predisposes patients to several diseases including breast cancer. Moreover, diabetic and obese women with breast cancer show a significant increase in mortality compared to non-obese and/or non-diabetic women. We hypothesized that these metabolic conditions incite an aggressive tumor phenotype by way of impacting tumor cell-autonomous and tumor cell non-autonomous events. In this study, we established a type 2 diabetic mouse model of triple-negative mammary carcinoma and investigated the effect of a glucose lowering therapy, metformin, on the overall tumor characteristics and immune/metabolic microenvironment. Diabetic mice exhibited larger mammary tumors that had increased adiposity with high levels of O-GlcNAc protein post-translational modification. These tumors also presented with a distinct stromal profile characterized by altered collagen architecture, increased infiltration by tumor-permissive M2 macrophages, and early metastatic seeding compared to non-diabetic/lean mice. Metformin treatment of the diabetic/obese mice effectively normalized glucose levels, reconfigured the mammary tumor milieu, and decreased metastatic seeding. Our results highlight the impact of two metabolic complications of obesity and diabetes on tumor cell attributes and showcase metformin's ability to revert tumor cell and stromal changes induced by an obese and diabetic host environment.
Collapse
Affiliation(s)
| | - Brandon J Metge
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Chae-Myeong Ha
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dominique C Hinshaw
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mateus S V Mota
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sarah C Kammerud
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Tshering Lama-Sherpa
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Noha Sharafeldin
- Division of Hematology & Oncology, Dept of Medicine, UAB School of Medicine, UAB, USA; Institute for Cancer Outcomes and Survivorship, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Adam R Wende
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rajeev S Samant
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; Birmingham Veterans Affairs, Birmingham, AL, USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lalita A Shevde
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
38
|
Supabphol S, Seubwai W, Wongkham S, Saengboonmee C. High glucose: an emerging association between diabetes mellitus and cancer progression. J Mol Med (Berl) 2021; 99:1175-1193. [PMID: 34036430 DOI: 10.1007/s00109-021-02096-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 04/16/2021] [Accepted: 05/18/2021] [Indexed: 12/11/2022]
Abstract
The association of cancer and diabetes mellitus (DM) has been studied for decades. Hyperglycemia and the imbalance of hormones are factors that contribute to the molecular link between DM and carcinogenesis and cancer progression. Hyperglycemia alone or in combination with hyperinsulinemia are key factors that promote cancer aggressiveness. Many preclinical studies suggest that high glucose induces abnormal energy metabolism and aggressive cancer via several mechanisms. As evidenced by clinical studies, hyperglycemia is associated with poor clinical outcomes in patients who have comorbid DM. The prognoses of cancer patients with DM are improved when their plasma glucose levels are controlled. This suggests that high glucose level maybe be involved in the molecular mechanism that causes the link between DM and cancer and may also be useful for prognosis of cancer progression. This review comprehensively summarizes the evidence from recent pre-clinical and clinical studies of the impact of hyperglycemia on cancer advancement as well as the underlying molecular mechanism for this impact. Awareness among clinicians of the association between hyperglycemia or DM and cancer progression may improve cancer treatment outcome in patients who have DM.
Collapse
Affiliation(s)
- Suangson Supabphol
- The Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Wunchana Seubwai
- Department of Forensic Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand.,Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand.,Center for Translational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Sopit Wongkham
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand.,Center for Translational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand.,Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Charupong Saengboonmee
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand. .,Center for Translational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand. .,Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand.
| |
Collapse
|
39
|
O-GlcNAcylation and O-GlcNAc Cycling Regulate Gene Transcription: Emerging Roles in Cancer. Cancers (Basel) 2021; 13:cancers13071666. [PMID: 33916244 PMCID: PMC8037238 DOI: 10.3390/cancers13071666] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 03/26/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary O-linked β-N-acetylglucosamine (O-GlcNAc) is a post-translational modification (PTM) linking nutrient flux through the hexosamine biosynthetic pathway (HBP) to gene transcription. Mounting experimental and clinical data implicates aberrant O-GlcNAcylation in the development and progression of cancer. Herein, we discuss how alteration of O-GlcNAc-regulated transcriptional mechanisms leads to atypical gene expression in cancer. We discuss the challenges associated with studying O-GlcNAc function and present several new approaches for studies of O-GlcNAc-regulated transcription. Abstract O-linked β-N-acetylglucosamine (O-GlcNAc) is a single sugar post-translational modification (PTM) of intracellular proteins linking nutrient flux through the Hexosamine Biosynthetic Pathway (HBP) to the control of cis-regulatory elements in the genome. Aberrant O-GlcNAcylation is associated with the development, progression, and alterations in gene expression in cancer. O-GlcNAc cycling is defined as the addition and subsequent removal of the modification by O-GlcNAc Transferase (OGT) and O-GlcNAcase (OGA) provides a novel method for cells to regulate various aspects of gene expression, including RNA polymerase function, epigenetic dynamics, and transcription factor activity. We will focus on the complex relationship between phosphorylation and O-GlcNAcylation in the regulation of the RNA Polymerase II (RNAP II) pre-initiation complex and the regulation of the carboxyl-terminal domain of RNAP II via the synchronous actions of OGT, OGA, and kinases. Additionally, we discuss how O-GlcNAcylation of TATA-box binding protein (TBP) alters cellular metabolism. Next, in a non-exhaustive manner, we will discuss the current literature on how O-GlcNAcylation drives gene transcription in cancer through changes in transcription factor or chromatin remodeling complex functions. We conclude with a discussion of the challenges associated with studying O-GlcNAcylation and present several new approaches for studying O-GlcNAc regulated transcription that will advance our understanding of the role of O-GlcNAc in cancer.
Collapse
|
40
|
FGF21 facilitates autophagy in prostate cancer cells by inhibiting the PI3K-Akt-mTOR signaling pathway. Cell Death Dis 2021; 12:303. [PMID: 33753729 PMCID: PMC7985321 DOI: 10.1038/s41419-021-03588-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 02/27/2021] [Accepted: 03/03/2021] [Indexed: 12/11/2022]
Abstract
Fibroblast growth factor 21 (FGF21) plays an important role in regulating glucose and lipid metabolism, but its role in cancer is less well-studied. We aimed to investigate the action of FGF21 in the development of prostate cancer (PCa). Herein, we found that FGF21 expression was markedly downregulated in PCa tissues and cell lines. FGF21 inhibited the proliferation and clone formation of LNCaP cells (a PCa cell line) and promoted apoptosis. FGF21 also inhibited PCa cell migration and invasiveness. The Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses revealed that FGF21 was related to autophagy and the phosphatidylinositol 3-kinase–Akt kinase–mammalian target of rapamycin (PI3K–Akt–mTOR) pathway. Mechanistically, FGF21 promoted autophagy in LNCaP cells by inhibiting the PI3K–Akt–mTOR–70S6K pathway. In addition, FGF21 inhibited PCa tumorigenesis in vivo in nude mice. Altogether, our findings show that FGF21 inhibits PCa cell proliferation and promoted apoptosis in PCa cells through facilitated autophagy. Therefore, FGF21 might be a potential novel target in PCa therapy.
Collapse
|
41
|
Ma J, Wu C, Hart GW. Analytical and Biochemical Perspectives of Protein O-GlcNAcylation. Chem Rev 2021; 121:1513-1581. [DOI: 10.1021/acs.chemrev.0c00884] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Junfeng Ma
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Georgetown University, Washington D.C. 20057, United States
| | - Ci Wu
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Georgetown University, Washington D.C. 20057, United States
| | - Gerald W. Hart
- Department of Biochemistry and Molecular Biology, Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia 30602, United States
| |
Collapse
|
42
|
Caon I, Parnigoni A, Viola M, Karousou E, Passi A, Vigetti D. Cell Energy Metabolism and Hyaluronan Synthesis. J Histochem Cytochem 2021; 69:35-47. [PMID: 32623953 PMCID: PMC7780193 DOI: 10.1369/0022155420929772] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/01/2020] [Indexed: 02/06/2023] Open
Abstract
Hyaluronan (HA) is a linear glycosaminoglycan (GAG) of extracellular matrix (ECM) synthesized by three hyaluronan synthases (HASes) at the plasma membrane using uridine diphosphate (UDP)-glucuronic acid (UDP-GlcUA) and UDP-N-acetylglucosamine (UDP-GlcNAc) as substrates. The production of HA is mainly regulated by hyaluronan synthase 2 (HAS2), that can be controlled at different levels, from epigenetics to transcriptional and post-translational modifications. HA biosynthesis is an energy-consuming process and, along with HA catabolism, is strongly connected to the maintenance of metabolic homeostasis. The cytoplasmic pool of UDP-sugars is critical for HA synthesis. UDP-GlcNAc is an important nutrient sensor and serves as donor substrate for the O-GlcNAcylation of many cytosolic proteins, including HAS2. This post-translational modification stabilizes HAS2 in the membrane and increases HA production. Conversely, HAS2 can be phosphorylated by AMP activated protein kinase (AMPK), a master metabolic regulator activated by low ATP/AMP ratios, which inhibits HA secretion. Similarly, HAS2 expression and the deposition of HA within the pericellular coat are inhibited by sirtuin 1 (SIRT1), another important energetic sensor, confirming the tight connection between nutrients availability and HA metabolism.
Collapse
Affiliation(s)
- Ilaria Caon
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Arianna Parnigoni
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Manuela Viola
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Evgenia Karousou
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Davide Vigetti
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| |
Collapse
|
43
|
Clifford RE, Gerrard AD, Fok M, Vimalachandran D. Metformin as a radiosensitiser for pelvic malignancy: A systematic review of the literature. Eur J Surg Oncol 2020; 47:1252-1257. [PMID: 33358075 DOI: 10.1016/j.ejso.2020.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/25/2020] [Accepted: 12/14/2020] [Indexed: 10/22/2022] Open
Abstract
BACKGROUND The treatment of pelvic malignancies has continued to improve over recent years, with neoadjuvant radiotherapy often considered the gold standard to downstage disease. Radiosensitisers are routinely employed in an attempt to improve response of cancers to radiotherapy. Previous preclinical evidence has suggested a role for metformin, a commonly used drug for type 2 diabetes. METHOD A literature search was performed for published full text articles using the PubMed, Cochrane and Scopus databases using the search criteria string 'Metformin' AND ('Radiosensitivity' OR 'radiosensitising' OR 'radiosensitising'). Additional papers were detected by scanning the references of relevant papers. Data were extracted from each study by two authors onto a dedicated proforma. The review was registered on the PROSPERO database (ID: CRD42020199066). RESULTS A total of 242 papers were identified, 11 of which were included in this review; an additional 5 papers were obtained from reference searches. Metformin has been demonstrated to reduce cell-viability post-radiotherapy in both rectal and prostate cancer cell lines, with an enhanced effect in tumours with a p53 mutation and increased apoptosis post-radiotherapy for cervical cancer. Clinical trials demonstrate improved tumour and nodal downstaging and pCR rates for rectal cancer using metformin as a radiosensitiser. CONCLUSION With an increasing understanding of the underlying mechanism of the effects on metformin prospective studies are required to assess the effect of routine use on cancer related outcomes. Progressive future studies may be better served by the use of predictive biomarker guided treatment to enable identification of the appropriate cohort to target.
Collapse
Affiliation(s)
- R E Clifford
- Institute of Cancer Medicine, University of Liverpool, UK.
| | - A D Gerrard
- The Countess of Chester Hospital NHS Foundation Trust, Chester, UK
| | - M Fok
- Institute of Cancer Medicine, University of Liverpool, UK
| | - D Vimalachandran
- The Countess of Chester Hospital NHS Foundation Trust, Chester, UK
| |
Collapse
|
44
|
Oliveira IA, Allonso D, Fernandes TVA, Lucena DMS, Ventura GT, Dias WB, Mohana-Borges RS, Pascutti PG, Todeschini AR. Enzymatic and structural properties of human glutamine:fructose-6-phosphate amidotransferase 2 (hGFAT2). J Biol Chem 2020; 296:100180. [PMID: 33303629 PMCID: PMC7948480 DOI: 10.1074/jbc.ra120.015189] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 11/24/2022] Open
Abstract
Glycoconjugates play a central role in several cellular processes, and alteration in their composition is associated with numerous human pathologies. Substrates for cellular glycosylation are synthesized in the hexosamine biosynthetic pathway, which is controlled by the glutamine:fructose-6-phosphate amidotransfera-se (GFAT). Human isoform 2 GFAT (hGFAT2) has been implicated in diabetes and cancer; however, there is no information about structural and enzymatic properties of this enzyme. Here, we report a successful expression and purification of a catalytically active recombinant hGFAT2 (rhGFAT2) in Escherichia coli cells fused or not to a HisTag at the C-terminal end. Our enzyme kinetics data suggest that hGFAT2 does not follow the expected ordered bi–bi mechanism, and performs the glucosamine-6-phosphate synthesis much more slowly than previously reported for other GFATs. In addition, hGFAT2 is able to isomerize fructose-6-phosphate into glucose-6-phosphate even in the presence of equimolar amounts of glutamine, which results in unproductive glutamine hydrolysis. Structural analysis of a three-dimensional model of rhGFAT2, corroborated by circular dichroism data, indicated the presence of a partially structured loop in the glutaminase domain, whose sequence is present in eukaryotic enzymes but absent in the E. coli homolog. Molecular dynamics simulations suggest that this loop is the most flexible portion of the protein and plays a key role on conformational states of hGFAT2. Thus, our study provides the first comprehensive set of data on the structure, kinetics, and mechanics of hGFAT2, which will certainly contribute to further studies on the (patho)physiology of hGFAT2.
Collapse
Affiliation(s)
- Isadora A Oliveira
- Laboratório de Glicobiologia Estrutural e Funcional, Instituto de Biofísica Carlos Chagas Filho (IBCCF), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil.
| | - Diego Allonso
- Laboratório de Glicobiologia Estrutural e Funcional, Instituto de Biofísica Carlos Chagas Filho (IBCCF), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil; Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, UFRJ, Rio de Janeiro, RJ, Brazil
| | - Tácio V A Fernandes
- Laboratório de Modelagem e Dinâmica Molecular, IBCCF, UFRJ, Rio de Janeiro, RJ, Brazil; Laboratório de Macromoléculas, Diretoria de Metrologia Aplicada às Ciências da Vida, Instituto Nacional de Metrologia, Qualidade e Tecnologia (INMETRO), Duque de Caxias, RJ, Brazil
| | - Daniela M S Lucena
- Laboratório de Glicobiologia Estrutural e Funcional, Instituto de Biofísica Carlos Chagas Filho (IBCCF), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Gustavo T Ventura
- Laboratório de Glicobiologia Estrutural e Funcional, Instituto de Biofísica Carlos Chagas Filho (IBCCF), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Wagner Barbosa Dias
- Laboratório de Glicobiologia Estrutural e Funcional, Instituto de Biofísica Carlos Chagas Filho (IBCCF), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | | | - Pedro G Pascutti
- Laboratório de Modelagem e Dinâmica Molecular, IBCCF, UFRJ, Rio de Janeiro, RJ, Brazil
| | - Adriane R Todeschini
- Laboratório de Glicobiologia Estrutural e Funcional, Instituto de Biofísica Carlos Chagas Filho (IBCCF), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
45
|
Montemurro N, Perrini P, Rapone B. Clinical Risk and Overall Survival in Patients with Diabetes Mellitus, Hyperglycemia and Glioblastoma Multiforme. A Review of the Current Literature. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E8501. [PMID: 33212778 PMCID: PMC7698156 DOI: 10.3390/ijerph17228501] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 11/11/2020] [Accepted: 11/15/2020] [Indexed: 12/15/2022]
Abstract
The relationship between type 2 diabetes mellitus (DM2) and hyperglycemia with cancer patients remains controversial also in the setting of patients with glioblastoma multiforme (GBM), the most common and aggressive form of astrocytoma with a short overall survival (OS) and poor prognosis. A systematic search of two databases was performed for studies published up to 19 August 2020, reporting the OS of patients with DM2 or high blood sugar level and GBM and the clinical risk of diabetic patients for development of GBM. According to PRISMA guidelines, we included a total of 20 papers reporting clinical data of patients with GBM and diabetes and/or hyperglycemia. The aim of this review was to investigate the effect of DM2, hyperglycemia and metformin on OS of patients with GBM. In addition, we evaluated the effect of these factors on the risk of development of GBM. This review supports accumulating evidence that hyperglycemia, rather than DM2, and elevated BMI are independent risk factors for poor outcome and shorter OS in patients with GBM. GBM patients with normal weight compared to obese, and diabetic patients on metformin compared to other therapies, seems to have a longer OS. Further studies are needed to understand better these associations.
Collapse
Affiliation(s)
- Nicola Montemurro
- Department of Neurosurgery, Azienda Ospedaliera Universitaria Pisana (AOUP), 56126 Pisa, Italy;
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy
| | - Paolo Perrini
- Department of Neurosurgery, Azienda Ospedaliera Universitaria Pisana (AOUP), 56126 Pisa, Italy;
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy
| | - Biagio Rapone
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, “Aldo Moro” University of Bari, 70121 Bari, Italy;
| |
Collapse
|
46
|
Koobotse MO, Schmidt D, Holly JMP, Perks CM. Glucose Concentration in Cell Culture Medium Influences the BRCA1-Mediated Regulation of the Lipogenic Action of IGF-I in Breast Cancer Cells. Int J Mol Sci 2020; 21:E8674. [PMID: 33212987 PMCID: PMC7698585 DOI: 10.3390/ijms21228674] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/11/2020] [Accepted: 11/13/2020] [Indexed: 01/09/2023] Open
Abstract
Hyperglycaemia is a common metabolic alteration associated with breast cancer risk and progression. We have previously reported that BRCA1 restrains metabolic activity and proliferative response to IGF-I anabolic actions in breast cancer cells cultured in high glucose. Here, we evaluated the impact of normal physiological glucose on these tumour suppressive roles of BRCA1. Human breast cancer cells cultured in normal physiological and high glucose were treated with IGF-I (0-500 ng/mL). Cellular responses were evaluated using immunoblotting, co-immunoprecipitation, and cell viability assay. As we previously reported, IGF-I induced ACCA dephosphorylation by reducing the association between BRCA1 and phosphorylated ACCA in high glucose, and upregulated FASN abundance downstream of ACCA. However, these effects were not observed in normal glucose. Normal physiological glucose conditions completely blocked IGF-I-induced ACCA dephosphorylation and FASN upregulation. Co-immunoprecipitation studies showed that normal physiological glucose blocked ACCA dephosphorylation by increasing the association between BRCA1 and phosphorylated ACCA. Compared to high glucose, the proliferative response of breast cancer cells to IGF-I was reduced in normal glucose, whereas no difference was observed in normal mammary epithelial cells. Considering these results collectively, we conclude that normal physiological glucose promotes the novel function of BRCA1 as a metabolic restraint of IGF-I actions. These data suggest that maintaining normal glucose levels may improve BRCA1 function in breast cancer and slow down cancer progression.
Collapse
Affiliation(s)
- Moses O. Koobotse
- IGFs & Metabolic Endocrinology Group, Bristol Medical School, Translational Health Sciences, University of Bristol, Bristol BS10 5NB, UK; (M.O.K.); (D.S.); (J.M.P.H.)
- Faculty of Health Sciences, School of Allied Health Professions, University of Botswana, Gaborone, Plot 4775, Botswana
| | - Dayane Schmidt
- IGFs & Metabolic Endocrinology Group, Bristol Medical School, Translational Health Sciences, University of Bristol, Bristol BS10 5NB, UK; (M.O.K.); (D.S.); (J.M.P.H.)
| | - Jeff M. P. Holly
- IGFs & Metabolic Endocrinology Group, Bristol Medical School, Translational Health Sciences, University of Bristol, Bristol BS10 5NB, UK; (M.O.K.); (D.S.); (J.M.P.H.)
| | - Claire M. Perks
- IGFs & Metabolic Endocrinology Group, Bristol Medical School, Translational Health Sciences, University of Bristol, Bristol BS10 5NB, UK; (M.O.K.); (D.S.); (J.M.P.H.)
| |
Collapse
|
47
|
O-GlcNAcylation Links Nutrition to the Epigenetic Downregulation of UNC5A during Colon Carcinogenesis. Cancers (Basel) 2020; 12:cancers12113168. [PMID: 33126652 PMCID: PMC7693889 DOI: 10.3390/cancers12113168] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/22/2020] [Accepted: 10/25/2020] [Indexed: 12/23/2022] Open
Abstract
Simple Summary Nutritional disorders represent major risk factors for colorectal cancer according to mechanisms of action that are still insufficiently known. The aim of our study was to investigate the putative involvement of nutrition in the epigenetic downregulation of the tumor suppressor genes of the UNC5 (Uncoordinated 5) family during colonic carcinogenesis and to understand its molecular relays. Herein, we provided evidence that the consumption of a High Carbohydrate Diet worsens colon carcinogenesis in mice and is correlated with the downregulation of several members of the UNC5 family whose UNC5A (Uncoordinated 5A). Mechanistically, we identified the nutritional sensor O-GlcNAcylation as one of the molecular relays that regulate the recruitment of the PRC2 complex onto the UNC5A promoter to repress its transcription. Abstract While it is now accepted that nutrition can influence the epigenetic modifications occurring in colorectal cancer (CRC), the underlying mechanisms are not fully understood. Among the tumor suppressor genes frequently epigenetically downregulated in CRC, the four related genes of the UNC5 family: UNC5A, UNC5B, UNC5C and UNC5D encode dependence receptors that regulate the apoptosis/survival balance. Herein, in a mouse model of CRC, we found that the expression of UNC5A, UNC5B and UNC5C was diminished in tumors but only in mice subjected to a High Carbohydrate Diet (HCD) thus linking nutrition to their repression in CRC. O-GlcNAcylation is a nutritional sensor which has enhanced levels in CRC and regulates many cellular processes amongst epigenetics. We then investigated the putative involvement of O-GlcNAcylation in the epigenetic downregulation of the UNC5 family members. By a combination of pharmacological inhibition and RNA interference approaches coupled to RT-qPCR (Reverse Transcription-quantitative Polymerase Chain Reaction) analyses, promoter luciferase assay and CUT&RUN (Cleavage Under Target & Release Using Nuclease) experiments, we demonstrated that the O-GlcNAcylated form of the histone methyl transferase EZH2 (Enhancer of Zeste Homolog 2) represses the transcription of UNC5A in human colon cancer cells. Collectively, our data support the hypothesis that O-GlcNAcylation could represent one link between nutrition and epigenetic downregulation of key tumor suppressor genes governing colon carcinogenesis including UNC5A.
Collapse
|
48
|
Zhang Y, Chen C, Yu T, Chen T. Proteomic Analysis of Protein Ubiquitination Events in Human Primary and Metastatic Colon Adenocarcinoma Tissues. Front Oncol 2020; 10:1684. [PMID: 33014840 PMCID: PMC7511592 DOI: 10.3389/fonc.2020.01684] [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: 04/27/2020] [Accepted: 07/29/2020] [Indexed: 12/16/2022] Open
Abstract
Protein ubiquitination is essential for multiple physiological processes through regulating the stability or function of target proteins and has been found to play critical roles in human cancers. However, the protein ubiquitination profile of human metastatic colon adenocarcinoma tissue has not been elucidated yet. In this study, a proprietary ubiquitin branch (K-ε-GG) antibody-based label-free quantitative proteomics and bioinformatics were performed to identify the global protein ubiquitination profile between human primary (Colon) and metastatic colon adenocarcinoma (Meta) tissues. A total of 375 ubiquitination sites from 341 proteins were identified as differentially modificated (| Fold change| > 1.5, p < 0.05) in Meta group compared with the Colon group. Among them, 132 ubiquitination sites from 127 proteins were upregulated and 243 ubiquitination sites from 214 proteins were downregulated in Meta group. Fifteen ubiquitination motifs were found. Furthermore, GO and KEGG pathway analysis indicated that proteins with altered ubiquitination in Meta group were enriched in pathways highly related to cancer metastasis, such as RNA transport and cell cycle. We speculate that the altered ubiquitination of CDK1 may be a pro-metastatic factor in colon adenocarcinoma. This study provides novel scientific evidences to elucidate the biological functions of protein ubiquitination in human colon adenocarcinoma and insights into its potential mechanisms of colon cancer metastasis, which would be helpful to discover novel biomarkers and therapeutic targets for effective treatment of colon cancer.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Colorectal Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Cong Chen
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Tingting Yu
- Department of Medical Genetics, School of Basic Medical Science, Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China
| | - Tao Chen
- Department of Colorectal Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
49
|
Rodrigues Mantuano N, Stanczak MA, Oliveira IDA, Kirchhammer N, Filardy AA, Monaco G, Santos RC, Fonseca AC, Fontes M, Bastos CDS, Dias WB, Zippelius A, Todeschini AR, Läubli H. Hyperglycemia Enhances Cancer Immune Evasion by Inducing Alternative Macrophage Polarization through Increased O-GlcNAcylation. Cancer Immunol Res 2020; 8:1262-1272. [PMID: 32819969 DOI: 10.1158/2326-6066.cir-19-0904] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 04/25/2020] [Accepted: 07/21/2020] [Indexed: 11/16/2022]
Abstract
Diabetes mellitus (DM) significantly increases the risk for cancer and cancer progression. Hyperglycemia is the defining characteristic of DM and tightly correlates with a poor prognosis in patients with cancer. The hexosamine biosynthetic pathway (HBP) is emerging as a pivotal cascade linking high glucose, tumor progression, and impaired immune function. Here we show that enhanced glucose flow through the HBP drives cancer progression and immune evasion by increasing O-GlcNAcylation in tumor-associated macrophages (TAM). Increased O-GlcNAc skewed macrophage polarization to a M2-like phenotype supporting tumor progression. Finally, we found an upregulation of M2 markers on TAMs in DM2 patients with colorectal cancer compared with nondiabetic normoglycemic patients. Our results provide evidence for a new and targetable mechanism of cancer immune evasion in patients with hyperglycemia, advocating for strict control of hyperglycemia in patients with cancer.
Collapse
Affiliation(s)
- Natália Rodrigues Mantuano
- Cancer Immunology Laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland.,Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Michal A Stanczak
- Cancer Immunology Laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Isadora de Araújo Oliveira
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Nicole Kirchhammer
- Cancer Immunology Laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Alessandra A Filardy
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gianni Monaco
- Cancer Immunology Laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Ronan Christian Santos
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Miguel Fontes
- Hospital Naval Marcílio Dias, Rio de Janeiro, Brazil
| | | | - Wagner B Dias
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alfred Zippelius
- Cancer Immunology Laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland.,Division of Oncology, Department of Internal Medicine, University Hospital Basel, Basel, Switzerland
| | - Adriane R Todeschini
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Heinz Läubli
- Cancer Immunology Laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland. .,Division of Oncology, Department of Internal Medicine, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
50
|
Narvaez CJ, Grebenc D, Balinth S, Welsh JE. Vitamin D regulation of HAS2, hyaluronan synthesis and metabolism in triple negative breast cancer cells. J Steroid Biochem Mol Biol 2020; 201:105688. [PMID: 32360595 PMCID: PMC8432753 DOI: 10.1016/j.jsbmb.2020.105688] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 03/28/2020] [Accepted: 04/26/2020] [Indexed: 01/29/2023]
Abstract
The vitamin D receptor (VDR) and its ligand 1,25(OH)2D3 (1,25D) exert anti-tumor effects, but considerable heterogeneity has been reported in different model systems. In general, cell lines derived from aggressive tumor subtypes such as Triple Negative Breast Cancer (TNBC) express low levels of VDR and are less sensitive to 1,25D than those derived from more differentiated tumor types. We have previously reported that 1,25D inhibits hyaluronic acid synthase 2 (HAS2) expression and hyaluronic acid (HA) synthesis in murine TNBC cells. Here we confirmed the inhibitory effect of 1,25D on HA synthesis in human Hs578T cells representative of the mesenchymal/stem-like (MSL) subtype of TNBC. Because HA synthesis requires the production of hexoses for incorporation into HA, we predicted that the high HA production characteristic of Hs578T cells would require sustained metabolic changes through the hexosamine biosynthetic pathway (HBP). We thus examined metabolic gene expression in Hs578T cell variants sorted for High (HAHigh) and Low (HALow) HA production, and the ability of 1,25D to reverse these adaptive changes. HAHigh populations exhibited elevated HA production, smaller size, increased proliferation and higher motility than HALow populations. Despite their more aggressive phenotype, HAHigh populations retained expression of VDR protein at levels comparable to that of parental Hs578T cells and HALow subclones. Treatment with 1,25D decreased production of HA in both HAHigh and HALow populations. We also found that multiple metabolic enzymes were aberrantly expressed in HAHigh cells, especially those involved in glutamine and glucose metabolism. Notably, Glutaminase (GLS), a known oncogene for breast cancer, was strongly upregulated in HAHigh vs. HALow cells and its expression was significantly reduced by 1,25D (100 nM, 24 h). Consistent with this finding, Seahorse extracellular flux analysis indicated that respiration in HAHigh cells was significantly more dependent on exogenous glutamine than HALow cells, however, acute 1,25D exposure did not alter metabolic flux. In contrast to GLS, the glutamate transporter SLC1A7 was significantly reduced in HAHigh cells compared to HALow cells and its expression was enhanced by 1,25D. These findings support the concept that 1,25D can reverse the metabolic gene expression changes associated with HA production in cancer cells with aggressive phenotypes.
Collapse
Affiliation(s)
- C J Narvaez
- Cancer Research Center, University at Albany, Rensselaer, NY 12144, United States.
| | - D Grebenc
- Department of Biochemistry, Queens University, Kingston, ON K7L 3N6, Canada
| | - S Balinth
- Cancer Research Center, University at Albany, Rensselaer, NY 12144, United States
| | - J E Welsh
- Cancer Research Center, University at Albany, Rensselaer, NY 12144, United States
| |
Collapse
|