1
|
Abstract
Sirtuin 7 (SIRT7) is a member of the sirtuin family and has emerged as a key player in numerous cellular processes. It exhibits various enzymatic activities and is predominantly localized in the nucleolus, playing a role in ribosomal RNA expression, DNA damage repair, stress response and chromatin compaction. Recent studies have revealed its involvement in diseases such as cancer, cardiovascular and bone diseases, and obesity. In cancer, SIRT7 has been found to be overexpressed in multiple types of cancer, including breast cancer, clear cell renal cell carcinoma, lung adenocarcinoma, prostate adenocarcinoma, hepatocellular carcinoma, and gastric cancer, among others. In general, cancer cells exploit SIRT7 to enhance cell growth and metabolism through ribosome biogenesis, adapt to stress conditions and exert epigenetic control over cancer-related genes. The aim of this review is to provide an in-depth understanding of the role of SIRT7 in cancer carcinogenesis, evolution and progression by elucidating the underlying molecular mechanisms. Emphasis is placed on unveiling the intricate molecular pathways through which SIRT7 exerts its effects on cancer cells. In addition, this review discusses the feasibility and challenges associated with the development of drugs that can modulate SIRT7 activity.
Collapse
Affiliation(s)
- Francisco Alejandro Lagunas-Rangel
- Department of Genetics and Molecular Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, San Pedro Zacatenco, Gustavo A. Madero, 07480, Mexico City, Mexico.
| |
Collapse
|
2
|
Ji JX, Hoang LN, Cochrane DR, Lum A, Senz J, Farnell D, Tessier-Cloutier B, Huntsman DG, Klein Geltink RI. The unique metabolome of clear cell ovarian carcinoma. J Pathol 2024; 264:160-173. [PMID: 39096103 DOI: 10.1002/path.6329] [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: 02/25/2024] [Revised: 04/29/2024] [Accepted: 06/08/2024] [Indexed: 08/04/2024]
Abstract
Clear cell ovarian carcinoma (CCOC) is an aggressive malignancy affecting younger women. Despite ovarian cancer subtypes having diverse molecular and clinical characteristics, the mainstay of treatment for advanced stage disease remains cytotoxic chemotherapy. Late stage CCOC is resistant to conventional chemotherapy, which means a suboptimal outcome for patients affected. Despite detailed genomic, epigenomic, transcriptomic, and proteomic characterisation, subtype-specific treatment for CCOC has shown little progress. The unique glycogen accumulation defining CCOC suggests altered metabolic pathway activity and dependency. This study presents the first metabolomic landscape of ovarian cancer subtypes, including 42 CCOC, 20 high-grade serous and 21 endometrioid ovarian carcinomas, together comprising the three most common ovarian carcinoma subtypes. We describe a distinct metabolomic landscape of CCOC compared with other ovarian cancer subtypes, including alterations in energy utilisation and cysteine metabolism. In addition, we identify CCOC-specific alterations in metabolic pathways including serine biosynthesis and ROS-associated pathways that could serve as potential therapeutic targets. Our study provides the first in-depth study into the metabolome of ovarian cancers and a rich resource to support ongoing research efforts to identify subtype-specific therapeutic targets that could improve the dismal outcome for patients with this devastating malignancy. © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Jennifer X Ji
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Lien N Hoang
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Dawn R Cochrane
- Department of Molecular Oncology, BC Cancer, Vancouver, BC, Canada
| | - Amy Lum
- Department of Molecular Oncology, BC Cancer, Vancouver, BC, Canada
| | - Janine Senz
- Department of Molecular Oncology, BC Cancer, Vancouver, BC, Canada
| | - David Farnell
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | | | - David G Huntsman
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Molecular Oncology, BC Cancer, Vancouver, BC, Canada
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, BC, Canada
| | - Ramon I Klein Geltink
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Molecular Oncology, BC Cancer, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| |
Collapse
|
3
|
Shi Z, Zeng H, Zhao B, Zeng C, Zhang F, Liu Z, Kwan HY, Su T. Sulforaphane reverses the enhanced NSCLC metastasis by regulating the miR-7-5p/c-Myc/LDHA axis in the acidic tumor microenvironment. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 133:155874. [PMID: 39079314 DOI: 10.1016/j.phymed.2024.155874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 06/13/2024] [Accepted: 07/09/2024] [Indexed: 09/01/2024]
Abstract
BACKGROUND The presence of distant metastasis at the time of initial diagnosis is a prevalent issue in non-small cell lung cancer (NSCLC), affecting around 30-40 % of the patients. Acidic tumor microenvironment (TME) provides favorable conditions that increase the invasiveness and aggressiveness of NSCLC. The activity of the glycolytic enzyme lactate dehydrogenase (LDHA) increases intracellular lactate accumulation, which creates an acidic TME. However, it is not yet known whether LDHA is involved in enhancing the metastatic potential of NSCLC and if LDHA is a druggable therapeutic target for NSCLC. PURPOSE We aimed to investigate the molecular mechanisms underlying the enhanced NSCLC metastasis in acidic TME, and to explore whether sulforaphane (SFN), an active compound in Raphani Semen, can serve as a LDHA inhibitor to inhibit NSCLC metastasis in the acidic TME. METHODS To mimic the acidic TME, NSCLC cells were cultured in acidic medium (pH 6.6), normal medium (pH 7.4) served as control. Western blotting, bioinformatic analysis, luciferase assay and rescue experiments were used to explore the mechanism and investigate the anti-metastatic effect of SFN both in vitro and in vivo. RESULTS Acidic environment increases the expression of LDHA which in turn increases the production of lactic acid that contributes to the acidity of TME. Interestingly, elevated LDHA expression results from increased c-Myc expression, which transactivates LDHA. c-Myc expression is directly regulated by miR-7-5p. In vitro study shows that overexpression of miR-7-5p reverses the acidic pH-enhanced c-Myc and LDHA expressions and also abolishes the enhanced NSCLC cell migration. More importantly, SFN significantly inhibits NSCLC growth and metastasis by reducing lactate production via the miR-7-5p/c-Myc/LDHA axis. Besides, it also regulates the expressions of monocarboxylate transporter 1 (MCT1) and MCT4 that transport lactate across cell membrane. CONCLUSIONS The miR-7-5p/c-Myc/LDHA axis is involved in the enhanced NSCLC metastasis in the acidic TME. SFN, a novel LDHA inhibitor, reduces lactate production by targeting the miR-7-5p/c-Myc/LDHA axis, and hence inhibits NSCLC metastasis. Our findings not only delineate a novel mechanism, but also support the clinical translation of SFN as a novel therapeutic agent for treating metastatic NSCLC.
Collapse
Affiliation(s)
- Zhiqiang Shi
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Huiyan Zeng
- Department of Endocrinology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Bingquan Zhao
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Chen Zeng
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Fan Zhang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Zhongqiu Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau.
| | - Hiu Yee Kwan
- Centre for Cancer & Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, PR China.
| | - Tao Su
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China.
| |
Collapse
|
4
|
Wang X, Zhang J, Xu X, Pan S, Cheng L, Dang K, Qi X, Li Y. Associations of daily eating frequency and nighttime fasting duration with biological aging in National Health and Nutrition Examination Survey (NHANES) 2003-2010 and 2015-2018. Int J Behav Nutr Phys Act 2024; 21:104. [PMID: 39300516 DOI: 10.1186/s12966-024-01654-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Information on the influences of daily eating frequency (DEF) and nighttime fasting duration (NFD) on biological aging is minimal. Our study investigated the potential associations of DEF and NFD with accelerated aging. METHODS Out of 24212 participants in NHANES 2003-2010 and 2015-2018, 4 predicted age metrics [homeostatic dysregulation (HD), Klemera-Doubal method (KDM), phenoAge (PA), and allostatic load (AL)] were computed based on 12 blood chemistry parameters. Utilizing 24-h dietary recall, DEF was measured by the frequency of eating occurrences, while NFD was determined by assessing the timing of the initial and final meals throughout the day. Weighted multivariate linear regression models and restricted cubic spline (RCS) were utilized to examine the associations. RESULTS Compared to DEF of ≤ 3.0 times, subjects with DEF ≥ 4.6 times demonstrated lower KDM residual [β: -0.57, 95% confidence-interval (CI): (-0.97, -0.17)] and PA residual [β: -0.47, 95% CI: (-0.69, -0.25)]. In comparison to NFD between 10.1 and 12.0 h, individuals with NFD ≤ 10.0 h were at higher HD [β: 0.03, 95% CI: (0.01, 0.04)], KDM residual [β: 0.34, 95% CI: (0.05, 0.63)], and PA residual [β: 0.38, 95% CI: (0.18, 0.57)]. Likewise, those with NFD ≥ 14.1 h also had higher HD [β: 0.02, 95% CI: (0.01, 0.04)] and KDM residual [β: 0.33, 95% CI: (0.03, 0.62)]. The results were confirmed by the dose-response relationships of DEF and NFD with predicted age metrics. Lactate dehydrogenase (LDH) and globulin (Glo) were acknowledged as implicated in and mediating the relationships. CONCLUSIONS DEF below 3.0 times and NFD less than 10.0 or more than 14.1 h were independently associated with higher predicted age metrics.
Collapse
Affiliation(s)
- Xuanyang Wang
- Department of Nutrition and Food Hygiene, School of Public Health, the National Key Discipline, Harbin Medical University, 157 Baojian Road, Harbin, 150081, P. R. China
| | - Jia Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, the National Key Discipline, Harbin Medical University, 157 Baojian Road, Harbin, 150081, P. R. China
| | - Xiaoqing Xu
- Department of Nutrition and Food Hygiene, School of Public Health, the National Key Discipline, Harbin Medical University, 157 Baojian Road, Harbin, 150081, P. R. China
| | - Sijia Pan
- Department of Nutrition and Food Hygiene, School of Public Health, the National Key Discipline, Harbin Medical University, 157 Baojian Road, Harbin, 150081, P. R. China
| | - Licheng Cheng
- Department of Nutrition and Food Hygiene, School of Public Health, the National Key Discipline, Harbin Medical University, 157 Baojian Road, Harbin, 150081, P. R. China
| | - Keke Dang
- Department of Nutrition and Food Hygiene, School of Public Health, the National Key Discipline, Harbin Medical University, 157 Baojian Road, Harbin, 150081, P. R. China
| | - Xiang Qi
- Department of Nutrition and Food Hygiene, School of Public Health, the National Key Discipline, Harbin Medical University, 157 Baojian Road, Harbin, 150081, P. R. China
| | - Ying Li
- Department of Nutrition and Food Hygiene, School of Public Health, the National Key Discipline, Harbin Medical University, 157 Baojian Road, Harbin, 150081, P. R. China.
| |
Collapse
|
5
|
Sharma H, Mondal S, Urquiza U, Esparza C, Bartlett S, Santa-Pinter L, Hill H, White M, Sharma P, Luckett-Chastain L, Cooper A, Rasel M, Gao P, Battaile KP, Shukla SK, Lovell S, Ihnat MA. Synthesis and biological characterization of an orally bioavailable lactate dehydrogenase-A inhibitor against pancreatic cancer. Eur J Med Chem 2024; 275:116598. [PMID: 38925013 DOI: 10.1016/j.ejmech.2024.116598] [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: 04/29/2024] [Revised: 06/09/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024]
Abstract
Lactate dehydrogenase-A (LDHA) is the major isoform of lactate dehydrogenases (LDH) that is overexpressed and linked to poor survival in pancreatic ductal adenocarcinoma (PDAC). Despite some progress, current LDH inhibitors have poor structural and physicochemical properties or exhibit unfavorable pharmacokinetics that have hampered their development. The present study reports the synthesis and biological evaluation of a novel class of LDHA inhibitors comprising a succinic acid monoamide motif. Compounds 6 and 21 are structurally related analogs that demonstrated potent inhibition of LDHA with IC50s of 46 nM and 72 nM, respectively. We solved cocrystal structures of compound 21-bound to LDHA that showed that the compound binds to a distinct allosteric site between the two subunits of the LDHA tetramer. Inhibition of LDHA correlated with reduced lactate production and reduction of glycolysis in MIA PaCa-2 pancreatic cancer cells. The lead compounds inhibit the proliferation of human pancreatic cancer cell lines and patient-derived 3D organoids and exhibit a synergistic cytotoxic effect with the OXPHOS inhibitor phenformin. Unlike current LDHA inhibitors, 6 and 21 have appropriate pharmacokinetics and ligand efficiency metrics, exhibit up to 73% oral bioavailability, and a cumulative half-life greater than 4 h in mice.
Collapse
Affiliation(s)
- Horrick Sharma
- Department of Pharmaceutical Sciences, College of Pharmacy, Southwestern Oklahoma State University, Weatherford, OK, USA.
| | - Somrita Mondal
- Department of Pharmaceutical Sciences, College of Pharmacy, Southwestern Oklahoma State University, Weatherford, OK, USA
| | - Uzziah Urquiza
- Department of Biological & Biomedical Sciences, Southwestern Oklahoma State University, Weatherford, OK, USA
| | - Colter Esparza
- Department of Biological & Biomedical Sciences, Southwestern Oklahoma State University, Weatherford, OK, USA
| | - Seth Bartlett
- Department of Pharmaceutical Sciences, College of Pharmacy, Southwestern Oklahoma State University, Weatherford, OK, USA
| | - Landon Santa-Pinter
- Department of Pharmaceutical Sciences, College of Pharmacy, Southwestern Oklahoma State University, Weatherford, OK, USA
| | - Hanna Hill
- Department of Biological & Biomedical Sciences, Southwestern Oklahoma State University, Weatherford, OK, USA
| | - Madalyn White
- Department of Biological & Biomedical Sciences, Southwestern Oklahoma State University, Weatherford, OK, USA
| | - Pragya Sharma
- Department of Biological & Biomedical Sciences, Southwestern Oklahoma State University, Weatherford, OK, USA
| | - Lerin Luckett-Chastain
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Anne Cooper
- Protein Structure and X-ray Crystallography Laboratory, The University of Kansas, Lawrence, KS, USA
| | - Mohammad Rasel
- Protein Structure and X-ray Crystallography Laboratory, The University of Kansas, Lawrence, KS, USA
| | - Philip Gao
- Protein Production Group, The University of Kansas, Lawrence, KS, USA
| | | | - Surendra K Shukla
- Department of Oncology Science, OU College of Medicine, Oklahoma City, USA
| | - Scott Lovell
- Protein Structure and X-ray Crystallography Laboratory, The University of Kansas, Lawrence, KS, USA
| | - Michael A Ihnat
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, USA
| |
Collapse
|
6
|
Peng X, He Z, Yuan D, Liu Z, Rong P. Lactic acid: The culprit behind the immunosuppressive microenvironment in hepatocellular carcinoma. Biochim Biophys Acta Rev Cancer 2024; 1879:189164. [PMID: 39096976 DOI: 10.1016/j.bbcan.2024.189164] [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: 01/08/2024] [Revised: 07/25/2024] [Accepted: 07/28/2024] [Indexed: 08/05/2024]
Abstract
As a solid tumor with high glycolytic activity, hepatocellular carcinoma (HCC) produces excess lactic acid and increases extracellular acidity, thus forming a unique immunosuppressive microenvironment. L-lactate dehydrogenase (LDH) and monocarboxylate transporters (MCTs) play a very important role in glycolysis. LDH is the key enzyme for lactic acid (LA) production, and MCT is responsible for the cellular import and export of LA. The synergistic effect of the two promotes the formation of an extracellular acidic microenvironment. In the acidic microenvironment of HCC, LA can not only promote the proliferation, survival, transport and angiogenesis of tumor cells but also have a strong impact on immune cells, ultimately leading to an inhibitory immune microenvironment. This article reviews the role of LA in HCC, especially its effect on immune cells, summarizes the progress of LDH and MCT-related drugs, and highlights the potential of immunotherapy targeting lactate combined with HCC.
Collapse
Affiliation(s)
- Xiaopei Peng
- Department of Radiology, the Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China; Molecular Imaging Research Center, Central South University, Changsha, Hunan 410013, China
| | - Zhenhu He
- Department of Radiology, the Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China; Molecular Imaging Research Center, Central South University, Changsha, Hunan 410013, China
| | - Dandan Yuan
- Department of Radiology, the Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China; Molecular Imaging Research Center, Central South University, Changsha, Hunan 410013, China
| | - Zhenguo Liu
- Department of Infectious Disease, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Pengfei Rong
- Department of Radiology, the Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China; Molecular Imaging Research Center, Central South University, Changsha, Hunan 410013, China.
| |
Collapse
|
7
|
Xu W, Huo J, Hu Q, Xu J, Chen G, Mo J, Zhou T, Jiang J. Association between lactate dehydrogenase to albumin ratio and acute kidney injury in patients with sepsis: a retrospective cohort study. Clin Exp Nephrol 2024; 28:882-893. [PMID: 38584195 DOI: 10.1007/s10157-024-02500-y] [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: 12/13/2023] [Accepted: 03/27/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND Serum lactate dehydrogenase to albumin ratio (LAR) is associated with poor outcomes in malignancy and pneumonia. However, there are few studies suggesting that LAR is associated with the occurrence of acute kidney injury (AKI) in patients with sepsis, which was investigated in this study. METHODS We conducted a retrospective cohort study based on the Medical Information Mart for Intensive Care (MIMIC)-IV database. The primary outcome was the occurrence of AKI within 2 days and 7 days. Multivariable logistic regression models were used to calculate odds ratios to validate the association between LAR and AKI, in-hospital mortality, RRT use, and recovery of renal function, respectively. RESULTS A total of 4010 participants were included in this study. The median age of the participants was 63.5 years and the median LAR was 10.5. After adjusting for confounding variables, patients in the highest LAR quartile had a higher risk of AKI than those in the lowest LAR quartile within 2 days and 7 days, with odds ratios of 1.37 (95% confidence interval [CI]: 1.23-1.52) and 1.95 (95% CI: 1.72-2.22), respectively. The adjusted odds of AKI within 2 and 7 days were 1.16 (95% CI: 1.12-1.20) and 1.29 (95% CI: 1.24-1.35) for each 1 unit increase in LAR(log2), respectively. CONCLUSION This study demonstrated that elevated LAR was associated with poor prognosis in patients with sepsis. The risk of AKI and in-hospital mortality increased, the need for RRT increased, and the chance of recovery of renal function decreased with the increase of LAR.
Collapse
Affiliation(s)
- Weigan Xu
- Department of Emergency, First People's Hospital of Foshan, No.18 Lingnan Avenue North, Chancheng District, Foshan, 528000, Guangdong Province, China.
| | - Jianyang Huo
- Department of Emergency, First People's Hospital of Foshan, No.18 Lingnan Avenue North, Chancheng District, Foshan, 528000, Guangdong Province, China
| | - Qiaohua Hu
- Department of Emergency, First People's Hospital of Foshan, No.18 Lingnan Avenue North, Chancheng District, Foshan, 528000, Guangdong Province, China
| | - Jingtao Xu
- Department of Emergency, First People's Hospital of Foshan, No.18 Lingnan Avenue North, Chancheng District, Foshan, 528000, Guangdong Province, China
| | - Guojun Chen
- Department of Emergency, First People's Hospital of Foshan, No.18 Lingnan Avenue North, Chancheng District, Foshan, 528000, Guangdong Province, China
| | - Jierong Mo
- Department of Emergency, First People's Hospital of Foshan, No.18 Lingnan Avenue North, Chancheng District, Foshan, 528000, Guangdong Province, China
| | - Tianen Zhou
- Department of Emergency, First People's Hospital of Foshan, No.18 Lingnan Avenue North, Chancheng District, Foshan, 528000, Guangdong Province, China
| | - Jun Jiang
- First People's Hospital of Foshan, Foshan, Guangdong Province, China
| |
Collapse
|
8
|
Feng X, Ren J, Zhang X, Kong D, Yin L, Zhou Q, Wang S, Li A, Guo Y, Wang Y, Feng X, Wang X, Niu J, Jiang Y, Zheng C. Lactate dehydrogenase A is implicated in the pathogenesis of B-cell lymphoma through regulation of the FER signaling pathway. Biofactors 2024; 50:1024-1038. [PMID: 38516823 DOI: 10.1002/biof.2053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 02/22/2024] [Indexed: 03/23/2024]
Abstract
Lactate dehydrogenase A (LDHA) is highly expressed in various tumors. However, the role of LDHA in the pathogenesis of B-cell lymphoma remains unclear. Analysis of data from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) databases revealed an elevated LDHA expression in diffuse large B-cell lymphoma (DLBC) tissues compared with normal tissues. Similarly, our results demonstrated a significant increase in LDHA expression in tumor tissues from the patients with B-cell lymphoma compared with those with lymphadenitis. To further elucidate potential roles of LDHA in B-cell lymphoma pathogenesis, we silenced LDHA in the Raji cells (a B-cell lymphoma cell line) using shRNA techniques. Silencing LDHA led to reduced mitochondrial membrane integrity, adenosine triphosphate (ATP) production, glycolytic activity, cell viability and invasion. Notably, LDHA knockdown substantially suppressed in vivo growth of Raji cells and extended survival in mice bearing lymphoma (Raji cells). Moreover, proteomic analysis identified feline sarcoma-related protein (FER) as a differential protein positively associated with LDHA expression. Treatment with E260, a FER inhibitor, significantly reduced the metabolism, proliferation and invasion of Raji cells. In summary, our findings highlight that LDHA plays multiple roles in B-cell lymphoma pathogenesis via FER pathways, establishing LDHA/FER may as a potential therapeutic target.
Collapse
MESH Headings
- Humans
- Animals
- Signal Transduction
- Mice
- Cell Line, Tumor
- Lymphoma, B-Cell/genetics
- Lymphoma, B-Cell/pathology
- Lymphoma, B-Cell/metabolism
- Gene Expression Regulation, Neoplastic
- Cell Proliferation/genetics
- Lactate Dehydrogenase 5/metabolism
- Lactate Dehydrogenase 5/genetics
- L-Lactate Dehydrogenase/genetics
- L-Lactate Dehydrogenase/metabolism
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Female
Collapse
Affiliation(s)
- Xiumei Feng
- Department of Hematology, The Second Hospital of Shandong University, Jinan, China
- Department of Hematology, Fourth People's Hospital of Jinan City, Jinan, China
| | - Jing Ren
- Department of Hematology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Xunqi Zhang
- Department of Hematology, The Second Hospital of Shandong University, Jinan, China
| | - Dexiao Kong
- Department of Hematology, The Second Hospital of Shandong University, Jinan, China
| | - Linlin Yin
- Department of Hematology, Fourth People's Hospital of Jinan City, Jinan, China
| | - Qian Zhou
- Hematology Department, Linyi Central Hospital, Yishui, China
| | - Shunye Wang
- Department of Hematology, The Second Hospital of Shandong University, Jinan, China
| | - Ai Li
- Department of Hematology, The Second Hospital of Shandong University, Jinan, China
| | - Yanan Guo
- Department of Hematology, The Second Hospital of Shandong University, Jinan, China
| | - Yongjing Wang
- Department of Hematology, The Second Hospital of Shandong University, Jinan, China
| | - Xiaoli Feng
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - Xiaoyun Wang
- Department of Nursing, The Second Hospital of Shandong University, Jinan, China
| | - Jianhua Niu
- Department of Hematology, Fourth People's Hospital of Jinan City, Jinan, China
| | - Yang Jiang
- Department of Hematology, The Second Hospital of Shandong University, Jinan, China
| | - Chengyun Zheng
- Department of Hematology, The Second Hospital of Shandong University, Jinan, China
| |
Collapse
|
9
|
Liu F, Wang Q, Wang Z, Zhang S, Ni Q, Chang H. ETV4 promotes the progression of cholangiocarcinoma by regulating glycolysis via the TGF-β signaling. Transl Oncol 2024; 47:102035. [PMID: 38878613 PMCID: PMC11225894 DOI: 10.1016/j.tranon.2024.102035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/29/2024] [Accepted: 06/11/2024] [Indexed: 07/08/2024] Open
Abstract
BACKGROUND Considerable studies show that ETS variant 4 (ETV4) plays an important roles in multitudinous tumor. This study investigated its function in cholangiocarcinoma (CCA) progression and revealed the underlying mechanisms. METHODS The expression of ETV4 in CCA was evaluated using TCGA database and the single-cell analysis based on GSE189903 dataset. ETV4 expression in CCA human specimens was detected by reverse transcription-quantitative PCR, immunohistochemistry, and western blot. Cell Counting Kit-8, EdU, colony formation, wound healing, and Transwell assays were used to analyze the effects of ETV4. Extracellular acidification rate, oxygen consumption rate, glucose uptake, and lactate production were used to measure glycolysis in CAA cells. Western blot was performed to explore glycolysis-related proteins. Tumor growth was evaluated in mice xenograft tumors. RESULTS ETV4 was up-regulated in CCA epithelial cells. The high-expression of ETV4 was associated with poor prognosis of patients with CCA. ETV4 overexpression enhanced the proliferation, migration, invasion, and glycolysis of CCA cells; ETV4 silencing led to the contrary effects. Mechanistically, ETV4 activates TGF-β/Smad2/3 signaling pathway. In mice xenograft mode, ETV4 silencing inhibits the tumor growth, the expression of glycolysis-related proteins and TGF-β/Smad2/3 pathway proteins. CONCLUSIONS ETV4 functions as an essential factor in the roles of TGF-β1 in CCA cells, and may be a promising target for TGF-β1-mediated CCA progression.
Collapse
Affiliation(s)
- Fangfeng Liu
- Department of Hepatobiliary surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| | - Qianchang Wang
- Department of Hepatobiliary surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Zhengjian Wang
- Department of Hepatobiliary surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Shizhe Zhang
- Department of Hepatobiliary surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Qingqiang Ni
- Department of Hepatobiliary surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Hong Chang
- Department of Hepatobiliary surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| |
Collapse
|
10
|
Arak H, Elboga U, Cayirli YB, Aytekin A. Prognostic significance of 68 Ga-FAPI PET/CT in patients with bone metastases in various cancers. Ann Nucl Med 2024; 38:630-638. [PMID: 38684594 DOI: 10.1007/s12149-024-01935-3] [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: 02/19/2024] [Accepted: 04/18/2024] [Indexed: 05/02/2024]
Abstract
OBJECTIVE This study aimed to compare 18FDGPET/CT in patients who develop bone metastases due to various cancers and to investigate the prognostic significance of the 68FAPI-PET/CT SUVmax value for survival. METHODS Patients with bone metastases who underwent both 68 Ga-FAPI PET/CT and 18FDGPET/CT within a 1 week period were included in this retrospective study. The effect of the SUVmax value of bone lesions on overall survival was analyzed. RESULTS A total of 75 eligible patients with 139 bone lesions were included in this study. The median age of the patients was 55 (30-83) and 48(64%) patients were newly diagnosed. The primary lesion median 68 Ga-FAPI PET/CT SUVmax value was higher than the median 18FDGPET/CT SUVmax (10.75 versus 6.7). Bone lesions 68 Ga-FAPI PET/CT SUVmax median (IQR) were 7.8 (4.6-13.2), and 18FDGPET/CT SUVmax of bone lesions were 5.9 (3.8-8.2). More bone lesions were detected on 68 Ga-FAPI PET/CT than on 18FDGPET/CT(median IQR 4 [1-9] versus 2 [1-6] (p = 0.014). The extra lesions observed on 68 Ga-FAPI PET/CT were mostly sclerotic bone lesions (p = 0.001).68 Ga-FAPI PET/CT SUVmax was significantly higher in vertebra and thorax lesions (p = 0.011 and p = 0.018, respectively). While the bone lesion 68 Ga-FAPI PET/CT SUVmax affected the OS, the 18FDGPET/CT SUVmax value did not affect the OS (p < 0.001 and p = 0.079, respectively). In ROC analysis, a cut-off-off value of 68 Ga-FAPI PET/CT SUVmax > 7.7 was found for OS (AUC: 0.619). The median OS in the group above the cut-off value was worse than that in the group below the cut-off value (32 versus 45) months (p = 0.002). In the multivariate analysis for OS, the 68 Ga-FAPI PET/CT SUVmax of bone lesions was an important parameter, as well as cancer subtype, ALP level, and disease occurrence. CONCLUSIONS 68 Ga-FAPI PET/CT detected more bone lesions and higher SUVmax values than 18FDGPET/CT in various cancers. The prognostic value of the SUVmax value of 68 Ga-FAPI PET/CT bone lesions was observed regardless of disease subtype.
Collapse
Affiliation(s)
- Hacı Arak
- Department of Medical Oncology, Gaziantep University Şahinbey Training and Research Hospital, TR-27310, Gaziantep, Turkey.
| | - Umut Elboga
- Department of Nuclear Medicine, Gaziantep University Şahinbey Training and Research Hospital, Şahinbey, Gaziantep, Turkey
| | - Yusuf Burak Cayirli
- Department of Nuclear Medicine, Gaziantep University Şahinbey Training and Research Hospital, Şahinbey, Gaziantep, Turkey
| | - Aydın Aytekin
- Department of Medical Oncology, Gaziantep University Şahinbey Training and Research Hospital, TR-27310, Gaziantep, Turkey
| |
Collapse
|
11
|
Yu Y, Jiang Y, Glandorff C, Sun M. Exploring the mystery of tumor metabolism: Warburg effect and mitochondrial metabolism fighting side by side. Cell Signal 2024; 120:111239. [PMID: 38815642 DOI: 10.1016/j.cellsig.2024.111239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 05/17/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
The metabolic reconfiguration of tumor cells constitutes a pivotal aspect of tumor proliferation and advancement. This study delves into two primary facets of tumor metabolism: the Warburg effect and mitochondrial metabolism, elucidating their contributions to tumor dominance. The Warburg effect facilitates efficient energy acquisition by tumor cells through aerobic glycolysis and lactic acid fermentation, offering metabolic advantages conducive to growth and proliferation. Simultaneously, mitochondrial metabolism, serving as the linchpin of sustained tumor vitality, orchestrates the tricarboxylic acid cycle and electron transport chain, furnishing a steadfast and dependable wellspring of biosynthesis for tumor cells. Regarding targeted therapy, this discourse examines extant strategies targeting tumor glycolysis and mitochondrial metabolism, underscoring their potential efficacy in modulating tumor metabolism while envisaging future research trajectories and treatment paradigms in the realm of tumor metabolism. By means of a thorough exploration of tumor metabolism, this study aspires to furnish crucial insights into the regulation of tumor metabolic processes, thereby furnishing valuable guidance for the development of novel therapeutic modalities. This comprehensive deliberation is poised to catalyze advancements in tumor metabolism research and offer novel perspectives and pathways for the formulation of cancer treatment strategies in the times ahead.
Collapse
Affiliation(s)
- Yongxin Yu
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yulang Jiang
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Christian Glandorff
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; University Clinic of Hamburg at the HanseMerkur Center of TCM, Hamburg, Germany
| | - Mingyu Sun
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
12
|
Wang DH, Ye LH, Ning JY, Zhang XK, Lv TT, Li ZJ, Wang ZY. Single-cell sequencing and multiple machine learning algorithms to identify key T-cell differentiation gene for progression of NAFLD cirrhosis to hepatocellular carcinoma. Front Mol Biosci 2024; 11:1301099. [PMID: 38993839 PMCID: PMC11237165 DOI: 10.3389/fmolb.2024.1301099] [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: 09/24/2023] [Accepted: 05/20/2024] [Indexed: 07/13/2024] Open
Abstract
Introduction: Hepatocellular carcinoma (HCC), which is closely associated with chronicinflammation, is the most common liver cancer and primarily involves dysregulated immune responses in the precancerous microenvironment. Currently, most studies have been limited to HCC incidence. However, the immunopathogenic mechanisms underlying precancerous lesions remain unknown. Methods: We obtained single-cell sequencing data (GSE136103) from two nonalcoholic fatty liver disease (NAFLD) cirrhosis samples and five healthy samples. Using pseudo-time analysis, we systematically identified five different T-cell differentiation states. Ten machine-learning algorithms were used in 81 combinations to integrate the frameworks and establish the best T-cell differentiation-related prognostic signature in a multi-cohort bulk transcriptome analysis. Results: LDHA was considered a core gene, and the results were validated using multiple external datasets. In addition, we validated LDHA expression using immunohistochemistry and flow cytometry. Conclusion: LDHA is a crucial marker gene in T cells for the progression of NAFLD cirrhosis to HCC.
Collapse
Affiliation(s)
- De-hua Wang
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Division of Liver Disease, The Fifth Hospital of Shijiazhuang, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Li-hong Ye
- Department of Pathology, The Fifth Hospital of Shijiazhuang, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jing-yuan Ning
- Department of Immunology, Immunology Department of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiao-kuan Zhang
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ting-ting Lv
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zi-jie Li
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zhi-yu Wang
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
13
|
Lu Y, Wang Y, Zhang L, Ma Z, Yu K, Shu Y, Zou X, Yang J, Liu X, Wang C, Du Y, Li Q. KAT7 enhances the proliferation and metastasis of head and neck squamous carcinoma by promoting the acetylation level of LDHA. Cancer Lett 2024; 590:216869. [PMID: 38593918 DOI: 10.1016/j.canlet.2024.216869] [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: 12/13/2023] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 04/11/2024]
Abstract
Lysine acetyltransferase 7 (KAT7), a histone acetyltransferase, has recently been identified as an oncoprotein and has been implicated in the development of various malignancies. However, its specific role in head and neck squamous carcinoma (HNSCC) has not been fully elucidated. Our study revealed that high expression of KAT7 in HNSCC patients is associated with poor survival prognosis and silencing KAT7 inhibits the Warburg effect, leading to reduced proliferation, invasion, and metastatic potential of HNSCC. Further investigation uncovered a link between the high expression of KAT7 in HNSCC and tumor-specific glycolytic metabolism. Notably, KAT7 positively regulates Lactate dehydrogenase A (LDHA), a key enzyme in metabolism, to promote lactate production and create a conducive environment for tumor proliferation and metastasis. Additionally, KAT7 enhances LDHA activity and upregulates LDHA protein expression by acetylating the lysine 118 site of LDHA. Treatment with WM3835, a KAT7 inhibitor, effectively suppressed the growth of subcutaneously implanted HNSCC cells in mice. In conclusion, our findings suggest that KAT7 exerts pro-cancer effects in HNSCC by acetylating LDHA and may serve as a potential therapeutic target. Inhibiting KAT7 or LDHA expression holds promise as a therapeutic strategy to suppress the growth and progression of HNSCC.
Collapse
Affiliation(s)
- Ying Lu
- School of Stomatology, Southern Medical University, Guang Zhou, 510515, China; Department of Stomatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Yong Wang
- Department of Nuclear Medicine, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, 100071, China
| | - Leilei Zhang
- Department of Stomatology, 920th Hospital of the Joint Logistics Support Force, PLA, Kunming, 650032, China
| | - Zhaofeng Ma
- Department of Stomatology, Beijing Shunyi District Hospital, Beijing, 101300, China
| | - Kaitao Yu
- Department of Stomatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Yao Shu
- Department of Stomatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Xuan Zou
- Department of Stomatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Jinjin Yang
- Department of Stomatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Xin Liu
- Department of Stomatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Chenglong Wang
- Department of Stomatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China.
| | - Yimeng Du
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, 100071, China.
| | - Qihong Li
- School of Stomatology, Southern Medical University, Guang Zhou, 510515, China; Department of Stomatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China.
| |
Collapse
|
14
|
Lin J, Zhong W, Lyu Z, Peng J, Rong Y, Zeng K, Lai J, Wu D, Wang J, Li Y, Zheng J, Zhang J, Pan Z. Circular RNA circTATDN3 promotes the Warburg effect and proliferation in colorectal cancer. Cancer Lett 2024; 589:216825. [PMID: 38548218 DOI: 10.1016/j.canlet.2024.216825] [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: 01/31/2024] [Revised: 03/12/2024] [Accepted: 03/18/2024] [Indexed: 04/07/2024]
Abstract
As one of the key metabolic enzymes in the glycolytic pathway, lactate dehydrogenase A (LDHA) might be linked to tumor proliferation by driving the Warburg effect. Circular RNAs (circRNAs) are widely implicated in tumor progression. Here, we report that circTATDN3, a circular RNA that interacts with LDHA, plays a critical role in proliferation and energy metabolism in CRC. We found that circTATDN3 expression was increased in CRC cells and tumor tissues and that high circTATDN3 expression was positively associated with poor postoperative prognosis in CRC patients. Additionally, circTATDN3 promoted the proliferation of CRC cells in vivo and vitro. Mechanistically, circTATDN3 was shown to function as an adaptor molecule that enhances the binding of LDHA to FGFR1, leading to increased LDHA phosphorylation and consequently promoting the Warburg effect. Moreover, circTATDN3 increased the expression of LDHA by sponging miR-511-5p, which synergistically promoted CRC progression and the Warburg effect. In conclusion, circTATDN3 may be a target for the treatment of CRC.
Collapse
Affiliation(s)
- Jiatong Lin
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, China; School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, 510006, China
| | - Wenhui Zhong
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Zejian Lyu
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, China
| | - Jingwen Peng
- Department of Rehabilitation Medicine, Sun Yat-sen Memorial Hospital, SunYat-sen University, Guangzhou, Guandong, China
| | - Yi Rong
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou City, Guangdong Province, China; State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangzhou, China
| | - Kejing Zeng
- Department of Endocrinology, Department of Diabetes and Obesity Reversal Research Center Guangdong Second Provincial General Hospital, 466 Xingang Middle Road, Guangzhou, Guangdong, 510317, China
| | - Jianguo Lai
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 510080, Guangzhou, China
| | - Deqing Wu
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, China
| | - Junjiang Wang
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, China
| | - Yong Li
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, China.
| | - Jun Zheng
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou, 510630, China.
| | - Jianwei Zhang
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
| | - Zihao Pan
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, China.
| |
Collapse
|
15
|
Chen J, Chen C, Zhang Z, Zeng F, Zhang S. Exploring the Key Amino Acid Residues Surrounding the Active Center of Lactate Dehydrogenase A for the Development of Ideal Inhibitors. Molecules 2024; 29:2029. [PMID: 38731521 PMCID: PMC11085338 DOI: 10.3390/molecules29092029] [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: 02/27/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Lactate dehydrogenase A (LDHA) primarily catalyzes the conversion between lactic acid and pyruvate, serving as a key enzyme in the aerobic glycolysis pathway of sugar in tumor cells. LDHA plays a crucial role in the occurrence, development, progression, invasion, metastasis, angiogenesis, and immune escape of tumors. Consequently, LDHA not only serves as a biomarker for tumor diagnosis and prognosis but also represents an ideal target for tumor therapy. Although LDHA inhibitors show great therapeutic potential, their development has proven to be challenging. In the development of LDHA inhibitors, the key active sites of LDHA are emphasized. Nevertheless, there is a relative lack of research on the amino acid residues around the active center of LDHA. Therefore, in this study, we investigated the amino acid residues around the active center of LDHA. Through structure comparison analysis, five key amino acid residues (Ala30, Met41, Lys131, Gln233, and Ala259) were identified. Subsequently, the effects of these five residues on the enzymatic properties of LDHA were investigated using site-directed mutagenesis. The results revealed that the catalytic activities of the five mutants varied to different degrees in both the reaction from lactic acid to pyruvate and pyruvate to lactic acid. Notably, the catalytic activities of LDHAM41G and LDHAK131I were improved, particularly in the case of LDHAK131I. The results of the molecular dynamics analysis of LDHAK131I explained the reasons for this phenomenon. Additionally, the optimum temperature of LDHAM41G and LDHAQ233M increased from 35 °C to 40 °C, whereas in the reverse reaction, the optimum temperature of LDHAM41G and LDHAK131I decreased from 70 °C to 60 °C. These findings indicate that Ala30, Met41, Lys131, Gln233, and Ala259 exert diverse effects on the catalytic activity and optimum temperature of LHDA. Therefore, these amino acid residues, in addition to the key catalytic site of the active center, play a crucial role. Considering these residues in the design and screening of LDHA inhibitors may lead to the development of more effective inhibitors.
Collapse
Affiliation(s)
- Jie Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China; (J.C.); (C.C.); (Z.Z.)
| | - Chen Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China; (J.C.); (C.C.); (Z.Z.)
| | - Zhengfu Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China; (J.C.); (C.C.); (Z.Z.)
| | - Fancai Zeng
- Key Laboratory of Southwest China Wildlife Resources Conservation, China West Normal University, Ministry of Education, Nanchong 637009, China
| | - Shujun Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China; (J.C.); (C.C.); (Z.Z.)
| |
Collapse
|
16
|
Vannas C, Escobar M, Österlund T, Andersson D, Mouhanna P, Soomägi A, Molin C, Wennergren D, Fagman H, Ståhlberg A. Treatment Monitoring of a Patient with Synchronous Metastatic Angiosarcoma and Breast Cancer Using ctDNA. Int J Mol Sci 2024; 25:4023. [PMID: 38612833 PMCID: PMC11012383 DOI: 10.3390/ijms25074023] [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: 03/03/2024] [Revised: 03/29/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
Angiosarcoma is a rare and aggressive type of soft-tissue sarcoma with high propensity to metastasize. For patients with metastatic angiosarcoma, prognosis is dismal and treatment options are limited. To improve the outcomes, identifying patients with poor treatment response at an earlier stage is imperative, enabling alternative therapy. Consequently, there is a need for improved methods and biomarkers for treatment monitoring. Quantification of circulating tumor-DNA (ctDNA) is a promising approach for patient-specific monitoring of treatment response. In this case report, we demonstrate that quantification of ctDNA using SiMSen-Seq was successfully utilized to monitor a patient with metastatic angiosarcoma. By quantifying ctDNA levels using 25 patient-specific mutations in blood plasma throughout surgery and palliative chemotherapy, we predicted the outcome and monitored the clinical response to treatment. This was accomplished despite the additional complexity of the patient having a synchronous breast cancer. The levels of ctDNA showed a superior correlation to the clinical outcome compared with the radiological evaluations. Our data propose a promising approach for personalized biomarker analysis to monitor treatment in angiosarcomas, with potential applicability to other cancers and for patients with synchronous malignancies.
Collapse
Affiliation(s)
- Christoffer Vannas
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden; (M.E.); (T.Ö.); (D.A.); (P.M.); (A.S.); (H.F.)
- Department of Oncology, Sahlgrenska University Hospital, Region Västra Götaland, 41345 Gothenburg, Sweden;
| | - Mandy Escobar
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden; (M.E.); (T.Ö.); (D.A.); (P.M.); (A.S.); (H.F.)
| | - Tobias Österlund
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden; (M.E.); (T.Ö.); (D.A.); (P.M.); (A.S.); (H.F.)
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Region Västra Götaland, 41345 Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Daniel Andersson
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden; (M.E.); (T.Ö.); (D.A.); (P.M.); (A.S.); (H.F.)
| | - Pia Mouhanna
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden; (M.E.); (T.Ö.); (D.A.); (P.M.); (A.S.); (H.F.)
- Department of Oncology, Ryhov County Hospital, 55185 Jönköping, Sweden
| | - Amanda Soomägi
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden; (M.E.); (T.Ö.); (D.A.); (P.M.); (A.S.); (H.F.)
| | - Claes Molin
- Department of Oncology, Sahlgrenska University Hospital, Region Västra Götaland, 41345 Gothenburg, Sweden;
| | - David Wennergren
- Department of Orthopaedics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden;
| | - Henrik Fagman
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden; (M.E.); (T.Ö.); (D.A.); (P.M.); (A.S.); (H.F.)
- Department of Clinical Pathology, Sahlgrenska University Hospital, Region Västra Götaland, 41345 Gothenburg, Sweden
| | - Anders Ståhlberg
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden; (M.E.); (T.Ö.); (D.A.); (P.M.); (A.S.); (H.F.)
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Region Västra Götaland, 41345 Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, 40530 Gothenburg, Sweden
| |
Collapse
|
17
|
Calbiague-Garcia V, Chen Y, Cádiz B, Tapia F, Paquet-Durand F, Schmachtenberg O. Extracellular lactate as an alternative energy source for retinal bipolar cells. J Biol Chem 2024; 300:106794. [PMID: 38403245 PMCID: PMC10966802 DOI: 10.1016/j.jbc.2024.106794] [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: 12/22/2023] [Revised: 02/11/2024] [Accepted: 02/16/2024] [Indexed: 02/27/2024] Open
Abstract
Retinal bipolar and amacrine cells receive visual information from photoreceptors and participate in the first steps of image processing in the retina. Several studies have suggested the operation of aerobic glycolysis and a lactate shuttle system in the retina due to the high production of this metabolite under aerobic conditions. However, whether bipolar cells form part of this metabolic circuit remains unclear. Here, we show that the monocarboxylate transporter 2 is expressed and functional in inner retinal neurons. Additionally, we used genetically encoded FRET nanosensors to demonstrate the ability of inner retinal neurons to consume extracellular lactate as an alternative to glucose. In rod bipolar cells, lactate consumption allowed cells to maintain the homeostasis of ions and electrical responses. We also found that lactate synthesis and transporter inhibition caused functional alterations and an increased rate of cell death. Overall, our data shed light on a notable but still poorly understood aspect of retinal metabolism.
Collapse
Affiliation(s)
- Victor Calbiague-Garcia
- PhD Program in Neuroscience, Universidad de Valparaíso, Valparaíso, Chile; CINV, Instituto de Biología, Universidad de Valparaíso, Valparaíso, Chile.
| | - Yiyi Chen
- Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | - Bárbara Cádiz
- CINV, Instituto de Biología, Universidad de Valparaíso, Valparaíso, Chile
| | - Felipe Tapia
- CINV, Instituto de Biología, Universidad de Valparaíso, Valparaíso, Chile
| | | | | |
Collapse
|
18
|
Yang Y, Song B, Guo M, Gao J, Jiang L, Li Q, Liu Y, Cao J. p53-dependent HIF-1α /autophagy mediated glycolysis to support Cr(VI)-induced cell growth and cell migration. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 272:116076. [PMID: 38335577 DOI: 10.1016/j.ecoenv.2024.116076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 01/23/2024] [Accepted: 02/03/2024] [Indexed: 02/12/2024]
Abstract
Cr(VI) is known to be seriously toxic and carcinogenic. Hypoxia-inducible factor-1α (HIF-1α) is a crucial regulator to promote tumor development. In this study, we found that Cr(VI) significantly increased the expression of HIF-1α in A549 cells and in lung of BALB/c mice but not in HELF cells. Treatment with Lificiguat (YC-1), HIF-1α inhibitor, or CoCl2, HIF-1α inducer, could alter Cr(VI)-induced autophagy, glycolysis, and cell growth in A549 cells but not in HELF cells, validating the involvement of HIF-1α in these effects of Cr(VI) in A549 cells. Co-treatments of pcATG4B with YC-1, or siATG4B with CoCl2 demonstrated the role of HIF-1α / autophagy axis in inducing glycolysis and cell growth in A549 cells. In HELF cells, however, only autophagy but not HIF-1α played a role in inducing glycolysis. The protein level of p53 was significantly lower in A549 cells than in HELF cells. RITA, a p53 inducer, attenuated Cr(VI)-induced HIF-1α and LC3-II in A549 cells, suggesting that p53 might be the mechanism underlying the different effects of Cr(VI) on HIF-1α in A549 and HELF cells. Thus, p53-dependent HIF-1α / autophagy-mediated glycolysis plays a role in facilitating Cr(VI)-induced carcinogenesis.
Collapse
Affiliation(s)
- Yanqiu Yang
- Department of Occupational and Environmental Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian 116044, China
| | - Bin Song
- Department of Gynecology and Obstetrics, First Affiliated Hospital (Army Medical University), Chongqing 400038, China
| | - Minna Guo
- Department of Occupational and Environmental Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian 116044, China
| | - Jianfeng Gao
- Department of Surgery, the Second Hospital of Dalian Medical University, Dalian, China
| | - Liping Jiang
- Department of Occupational and Environmental Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian 116044, China
| | - Qiujuan Li
- Department of Occupational and Environmental Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian 116044, China
| | - Yong Liu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China.
| | - Jun Cao
- Department of Occupational and Environmental Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian 116044, China.
| |
Collapse
|
19
|
Abdallah S, Abdel-Halim KY, Alm-Eldeen A. Anticancer potency of Egyptian venom snakes on MCF-7 and HepG2 carcinoma cells. Environ Anal Health Toxicol 2024; 39:e2024001-0. [PMID: 38631393 PMCID: PMC11079407 DOI: 10.5620/eaht.2024001] [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: 09/11/2023] [Accepted: 12/13/2023] [Indexed: 04/19/2024] Open
Abstract
Breast and hepatic cancers are the leading incidences in the globe occurring of the human sufferings from various cancers. Snake venoms have been reported to provide effective therapeutic agents. The current study investigates the anticancer potency of Egyptian venoms snakes on two cells: breast cancer cells (MCF-7) and hepato-cancer cells (HepG2) (In vitro assay). The examined venoms were more potent on MCF-7 than HepG2 cells. Their inhibition % on MCF-7 ranged from 71.47 to 99.02% with medium inhibition concentrations (IC50s): 3.48, 3.60, 3.70, 4.33, and 4.49 μg/ml for venoms: Echis pyramid (E.H), Cerastes vipera (C.V), Naja haje (N.H), Echis coloratus (E.C), and Cerastes cerastes (C.C), respectively. The values of IC50s on HepG2 were 4.32, 17.77, 59.72, 63.75, and 217.90 μg/ml for toxins: E.C, E.P, C.V, C.C, and N.H, respectively. Some biomarkers were conducted to investigate the apoptotic effects of toxins into the cells. Increasing profiles of lactate dehydrogenase (LDH) activity and levels of glutathione content (GSH) and malodialdhyde (MDA) as well as repairment of DNA indicated such these actions. So, more reliable investigations on these venoms were needed to provide intelligent therapeutic agent for cancer treatment.
Collapse
Affiliation(s)
- Sherif Abdallah
- Department of Zoology, Faculty of Science, Tanta University, Tanta, Egypt
| | - Khaled Y. Abdel-Halim
- Mammalian & Aquatic Toxicology Department, Central Agricultural Pesticides Laboratory (CAPL), Agricultural Research Center (ARC), Giza, Egypt
| | - Abeer Alm-Eldeen
- Department of Zoology, Faculty of Science, Tanta University, Tanta, Egypt
| |
Collapse
|
20
|
Luo Y, Yang Y, Ye M, Zuo J. Targeting metabolic reprogramming promotes the efficacy of transarterial chemoembolization in the rabbit VX2 liver tumor model. Oncol Lett 2024; 27:111. [PMID: 38304171 PMCID: PMC10831404 DOI: 10.3892/ol.2024.14244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 11/30/2023] [Indexed: 02/03/2024] Open
Abstract
Transarterial chemoembolization (TACE) may prolong the survival of patients with hepatocellular carcinoma (HCC); however, its efficacy is limited due to the high rate of incomplete embolization. Hypoxia after embolization can cause a series of changes in the tumor microenvironment, including lactate dehydrogenase A (LDHA) upregulation. Therefore, the current study assessed the antitumor effect and the underlying mechanism of the LDHA inhibitor, sodium oxamate (Ox), combined with TACE, using the rabbit VX2 liver tumor model. VX2 liver tumor models were created in the left liver lobe of rabbits, and after 14 days of treatments, the rabbits were sacrificed for the collection of the tumor tissues and blood samples. The antitumor effects of Ox, and the combination of Ox and TACE, and changes in the tumor microenvironment after treatments were assessed by histopathological evaluation, and the safety of the treatments was analyzed by measuring changes in the serum levels of alanine aminotransferase, aspartate aminotransferase, blood urea nitrogen and creatinine. The results demonstrated that the combination of Ox and TACE notably improved antitumor effects compared with in the other groups, as it significantly inhibited tumor growth. Additionally, treatment with Ox + TACE downregulated vascular endothelial growth factor and matrix metalloproteinase-9, and enhanced the infiltration of CD3+ and CD8+ T cells into tumor tissues, thus suggesting that Ox + TACE may have a synergistic effect on increasing the infiltration of immune cells in the tumor microenvironment. With a well-tolerated and manageable impairment of hepatorenal function, targeting metabolic reprogramming could promote the efficacy of TACE, thus providing novel avenues for the future clinical management of patients with advanced HCC.
Collapse
Affiliation(s)
- Yi Luo
- Department of Interventional Radiology, The Second Hospital of Wuhan Iron and Steel (Group) Corp., Wuhan, Hubei 430022, P.R. China
| | - Yong Yang
- Department of Oncology, The Second Hospital of Wuhan Iron and Steel (Group) Corp., Wuhan, Hubei 430022, P.R. China
| | - Meize Ye
- Department of Interventional Radiology, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| | - Jing Zuo
- Department of Oncology, The Second Hospital of Wuhan Iron and Steel (Group) Corp., Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
21
|
Kim HS, Cho JY. Exosome proteomes reveal glycolysis-related enzyme enrichment in primary canine mammary gland tumor compared to metastases. Proteome Sci 2024; 22:4. [PMID: 38419074 PMCID: PMC10900604 DOI: 10.1186/s12953-023-00226-5] [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: 09/10/2023] [Accepted: 12/20/2023] [Indexed: 03/02/2024] Open
Abstract
OBJECTIVE Numerous evidence has highlighted the differences between primary tumors and metastases. Nonetheless, the differences in exosomal proteins derived from primary tumor and metastases remain elusive. Here, we aimed to identify differentially expressed exosomal proteins from primary canine mammary gland tumor and metastases to understand how they shape their own tumor microenvironment. METHODS We clearly distinguished primary canine mammary gland tumors (CHMp) from metastases (CHMm) and profiled the proteins within their secreted exosomes using LC-MS/MS. Moreover, the abundance of glycolysis enzymes (GPI, LDHA) in CHMp exosome was verified with Western blotting, To broaden the scope, we extended to human colorectal cancer-derived exosomes (SW480 vs. SW620) for comparison. RESULTS We identified significant differences in 87 and 65 proteins derived from CHMp and CHMm, respectively. Notably, glycolysis enzymes (GPI, LDHA, LDHB, TPI1, and ALDOA) showed specific enrichment in exosomes from the primary tumor. CONCLUSION We observed significant differences in the cellular proteome between primary tumors and metastases, and intriguingly, we identified a parallel heterogeneity the protein composition of exosomes. Specifically, we reported that glycolysis enzymes were significantly enriched in CHMp exosomes compared to CHMm exosomes. We further demonstrated that this quantitative difference in glycolysis enzymes persisted across primary and metastases, extending to human colorectal cancer-derived exosomes (SW480 vs. SW620). Our findings of the specific enrichment of glycolysis enzymes in primary tumor-derived exosomes contribute to a better understanding of tumor microenvironment modulation and heterogeneity between primary tumors and metastases.
Collapse
Affiliation(s)
- Hui-Su Kim
- Department of Biochemistry, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 FOUR Future Veterinary Medicine Leading Education and Research Center, Seoul National University, Gwanak-ro1, Gwanak-Gu, Seoul, 08826, Republic of Korea
- Comparative Medicine Disease Research Center (CDRC), Science Research Center (SRC), Seoul National University, Seoul, 08826, Republic of Korea
| | - Je-Yoel Cho
- Department of Biochemistry, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 FOUR Future Veterinary Medicine Leading Education and Research Center, Seoul National University, Gwanak-ro1, Gwanak-Gu, Seoul, 08826, Republic of Korea.
- Comparative Medicine Disease Research Center (CDRC), Science Research Center (SRC), Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
22
|
Hou Z, Wu C, Tang J, Liu S, Li L. CLSPN actives Wnt/β-catenin signaling to facilitate glycolysis and cell proliferation in oral squamous cell carcinoma. Exp Cell Res 2024; 435:113935. [PMID: 38237848 DOI: 10.1016/j.yexcr.2024.113935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/01/2024] [Accepted: 01/15/2024] [Indexed: 02/04/2024]
Abstract
OBJECTIVE Oral squamous cell carcinoma (OSCC) is a common malignancy with a poor prognosis. This study aimed to determine the influence and underlying mechanisms of CLSPN on OSCC. METHODS CLSPN expression was tested using quantitative real-time polymerase chain reaction, immunohistochemistry, and western blotting. Flow cytometry, cell counting kit, and colony formation assays were performed to determine OSCC cell apoptosis, viability, and proliferation, respectively. In OSCC cells, the extracellular acidification rate (ECAR), oxygen consumption rate (OCR), glucose uptake, and lactate production were determined using the corresponding kits. Changes in the protein levels of HK2, PKM2, LDHA, Wnt3a, and β-catenin were assessed using western blotting. RESULTS CLSPN expression was increased in OSCC tissues. Overexpression of CLSPN in HSC-2 cells promoted cell proliferation, increased the levels of ECAR, glucose uptake, and lactate production, and increased the protein levels of HK2, PKM2, LDHA, Wnt3a, and β-catenin, but inhibited OCR levels and apoptosis. The knockdown of CLSPN in CAL27 cells resulted in the opposite results. Moreover, the effects of CLSPN overexpression on glycolysis and OSCC cell proliferation were reversed by Wnt3a knockdown. In vivo, knockdown of CLSPN restrained tumor growth, glycolysis, and the activation of Wnt/β-catenin signaling. CONCLUSION CLSPN promoted glycolysis and OSCC cell proliferation, and reduced apoptosis, which was achieved by the activation of Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Zeyu Hou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Chenzhou Wu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jinru Tang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Shaohua Liu
- Department of Oral and Maxillofacial Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China.
| | - Longjiang Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
23
|
Gödicke S, Kresbach C, Ehlert M, Obrecht D, Altendorf L, Hack K, von Hoff K, Carén H, Melcher V, Kerl K, Englinger B, Filbin M, Pajtler KW, Gojo J, Pietsch T, Rutkowski S, Schüller U. Clinically relevant molecular hallmarks of PFA ependymomas display intratumoral heterogeneity and correlate with tumor morphology. Acta Neuropathol 2024; 147:23. [PMID: 38265527 PMCID: PMC10808473 DOI: 10.1007/s00401-023-02682-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/05/2023] [Accepted: 12/30/2023] [Indexed: 01/25/2024]
Abstract
Posterior fossa type A (PF-EPN-A, PFA) ependymoma are aggressive tumors that mainly affect children and have a poor prognosis. Histopathology shows significant intratumoral heterogeneity, ranging from loose tissue to often sharply demarcated, extremely cell-dense tumor areas. To determine molecular differences in morphologically different areas and to understand their clinical significance, we analyzed 113 PF-EPN-A samples, including 40 corresponding relapse samples. Cell-dense areas ranged from 0 to 100% of the tumor area and displayed a higher proportion of proliferating tumor cells (p < 0.01). Clinically, cell density was associated with poor progression-free and overall survival (pPFS = 0.0026, pOS < 0.01). Molecularly, tumor areas with low and high cell density showed diverging DNA methylation profiles regarding their similarity to distinct previously discovered PF-EPN-A subtypes in 9/21 cases. Prognostically relevant chromosomal changes at 1q and 6q showed spatial heterogeneity within single tumors and were significantly enriched in cell-dense tumor areas as shown by single-cell RNA (scRNA)-sequencing as well as copy number profiling and fluorescence in situ hybridization (FISH) analyses of different tumor areas. Finally, spatial transcriptomics revealed cell-dense areas of different tumors to be more similar than various different areas of the same tumor. High-density areas distinctly overexpressed genes encoding histone proteins, WNT5A, TGFB1, or IGF2. Relapsing tumors displayed a higher proportion of cell-dense areas (p = 0.036), a change in PF-EPN-A methylation subtypes (13/32 patients), and novel chromosome 1q gains and 6q losses (12/32 cases) compared to corresponding primary tumors. Our data suggest that PF-EPN-A ependymomas habor a previously unrecognized intratumoral heterogeneity with clinical implications, which has to be accounted for when selecting diagnostic material, inter alia, by histological evaluation of the proportion of cell-dense areas.
Collapse
Affiliation(s)
- Swenja Gödicke
- Department of Pediatric Hematolgoy and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Research Institute Children's Cancer Center, Hamburg-Eppendorf, Hamburg, Germany
| | - Catena Kresbach
- Department of Pediatric Hematolgoy and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Research Institute Children's Cancer Center, Hamburg-Eppendorf, Hamburg, Germany
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Max Ehlert
- Department of Pediatric Hematolgoy and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Research Institute Children's Cancer Center, Hamburg-Eppendorf, Hamburg, Germany
| | - Denise Obrecht
- Department of Pediatric Hematolgoy and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lea Altendorf
- Department of Pediatric Hematolgoy and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Research Institute Children's Cancer Center, Hamburg-Eppendorf, Hamburg, Germany
| | - Karoline Hack
- Department of Pediatric Hematolgoy and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Research Institute Children's Cancer Center, Hamburg-Eppendorf, Hamburg, Germany
| | - Katja von Hoff
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, Berlin Institute of Health, Berlin, Germany
- Department of Pediatrics and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Helena Carén
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Viktoria Melcher
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, 48149, Münster, Germany
| | - Kornelius Kerl
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, 48149, Münster, Germany
| | - Bernhard Englinger
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
- Center for Cancer Research and Comprehensive Cancer Center, Medical University Vienna, 1090, Vienna, Austria
| | - Mariella Filbin
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Kristian W Pajtler
- Hopp Children's Cancer Center Heidelberg (KiTZ), 69120, Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), 69120, Heidelberg, Germany
- Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Johannes Gojo
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
| | - Torsten Pietsch
- Institute of Neuropathology, DGNN Brain Tumor Reference Center, University of Bonn Medical Center, Bonn, Germany
| | - Stefan Rutkowski
- Department of Pediatric Hematolgoy and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulrich Schüller
- Department of Pediatric Hematolgoy and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Research Institute Children's Cancer Center, Hamburg-Eppendorf, Hamburg, Germany.
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.
| |
Collapse
|
24
|
Gupta P, Singh S, Rai N, Verma A, Tiwari H, Kamble SC, Gautam HK, Gautam V. Unveiling the cytotoxic and anti-proliferative potential of green-synthesized silver nanoparticles mediated by Colletotrichum gloeosporioides. RSC Adv 2024; 14:4074-4088. [PMID: 38292267 PMCID: PMC10825743 DOI: 10.1039/d3ra06145k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 12/19/2023] [Indexed: 02/01/2024] Open
Abstract
Fungal endophytes are a putative source of bioactive metabolites that have found significant applications in nanomedicine due to their metabolic versatility. In the present study, an aqueous extract of the fungal endophyte, Colletotrichum gloeosporioides associated with a medicinal plant Oroxylum indicum, has been used for the fabrication of green silver nanoparticles (CgAgNPs) and further evaluated their cytotoxic and anti-proliferative activity. Bioanalytical techniques including UV-Vis spectral analysis revealed a sharp band at 435 nm and functional molecules from the aqueous extract involved in the synthesis of CgAgNPs were evidenced through FTIR. Further, the crystalline nature of CgAgNPs was determined through XRD analysis and microscopy techniques including AFM, TEM and FESEM demonstrated the spherical shape of CgAgNPs exhibiting a crystalline hexagonal lattice and the size was found to be in the range of 9-29 nm. The significant cytotoxic potential of CgAgNPs was observed against breast cancer cells, MDA-MB-231 and MCF-7 with IC50 values of 18.398 ± 0.376 and 38.587 ± 1.828 μg mL-1, respectively. The biochemical study revealed that the treatment of MDA-MB-231 and MCF-7 cells with CgAgNPs reduces glucose uptake, suppresses cell proliferation, and enhances LDH release, indicating reduced cell viability and progression. Moreover, our research revealed differential expression of genes associated with apoptosis, cell cycle inhibition and metastasis suppression, evidencing anti-proliferative activity of CgAgNPs. The main objective of the present study is to harness anti-breast cancer activity of novel biogenic nanoparticles synthesized using the aqueous extract of O. indicum associated C. gloeosporioides and study the underlying mechanistic pathway exerted by these mycogenic nanoparticles.
Collapse
Affiliation(s)
- Priyamvada Gupta
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University Varanasi-221005 India +918860182113
| | - Swati Singh
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University Varanasi-221005 India +918860182113
| | - Nilesh Rai
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University Varanasi-221005 India +918860182113
| | - Ashish Verma
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University Varanasi-221005 India +918860182113
| | - Harshita Tiwari
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University Varanasi-221005 India +918860182113
| | - Swapnil C Kamble
- Department of Technology, Savitribai Phule Pune University Ganeshkhind Pune 411007 India
| | - Hemant Kumar Gautam
- Department of Immunology and Infectious Disease Biology, CSIR-Institute of Genomics and Integrative Biology Sukhdev Vihar New Delhi 110025 India
| | - Vibhav Gautam
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University Varanasi-221005 India +918860182113
| |
Collapse
|
25
|
Javani Jouni F, Rastegar-Pouyani N, Najjar N, Nasirpour M, Payez A, Kashi G, Zafari J. Evaluation of the effects of photodynamic therapy consisted of the blue laser and zinc oxide QDs on MDA-MB-231 cancer cells by inhibiting cancer markers and inducing apoptosis. Lasers Med Sci 2024; 39:28. [PMID: 38216721 DOI: 10.1007/s10103-024-03977-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/02/2024] [Indexed: 01/14/2024]
Abstract
The increasing number of cancer patients has cast attention on developing new anti-cancer modalities. Photodynamic therapy is a safe anti-cancer approach, which encompasses (1) local administration of a photosensitizer and (2) light irradiation. Zinc oxide (ZnO) quantum dots (QDs) are photosensitizers that can be utilized for this purpose. In the present study, to better appreciate the likely more efficient cytotoxic effect of the combination of ZnO QDs and the visible 470-nm blue light in comparison to the QDs alone, several assays were to be conducted upon breast cancer MDA-MB 231 cells. MTT assay showed that in certain groups the combination displayed higher cytotoxic effects compared to those following QD treatment alone. LDH leakage and lipid peroxidation rates by the combination were significantly higher than treatment with either the blue laser or QDs. Although the combination managed to meaningfully reduce the number of colonies and CAT activity compared to QD treatment, there were no palpable differences between them. Lastly, the combination was able to increase the apoptotic genes, including BAX, TP53, caspase 3, and caspase 9 compared to QD, while, in the case of Bcl-2, an anti-apoptotic gene, none of the groups managed to make any tangible differences on its expression levels. Our findings propose that there may be synergistic effects between the blue laser and QD that can possibly be adopted in anti-cancer therapy in the future. However, further investigations regarding this matter are of the essence.
Collapse
Affiliation(s)
- Fatemeh Javani Jouni
- Department of Biochemistry and Biophysics, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Nima Rastegar-Pouyani
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Nabaa Najjar
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Nasirpour
- Department of Medical Genetics, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Atefeh Payez
- Department of Biology Education, Farhangian University, P.O. Box, Tehran, 14665-889, Iran
| | - Giti Kashi
- Department of Environmental Health Engineering, Faculty of Health and Medical Engineering, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Water Purification Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Jaber Zafari
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
26
|
Deng Y, Zhu G, Mi X, Jing X. Prognostic implication of a novel lactate score correlating with immunotherapeutic responses in pan-cancer. Aging (Albany NY) 2024; 16:820-843. [PMID: 38198170 PMCID: PMC10817381 DOI: 10.18632/aging.205423] [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: 07/18/2023] [Accepted: 12/01/2023] [Indexed: 01/11/2024]
Abstract
A thorough assessment of lactate-related genes (LRGs) in different types of human cancers is currently lacking. To elucidate the molecular landscape of LRGs, we conducted a comprehensive analysis using genomic, mRNA, and microRNA expression profiles and developed a lactate score model using the least absolute shrinkage and selection operator (LASSO) algorithm. We found that our lactate score could be a prognostic marker instead of LDHA for several cancer patients who possess high-frequency variants in LRGs. The lactate score also demonstrated an association with CD8+ T cells infiltration in multiple cancer types. Furthermore, our findings indicate that the lactate score holds promise as a potential biomarker for immunotherapy in patients with bladder cancer (BLCA) and skin cutaneous melanoma (SKCM). Among the seventeen genes of the lactate score model, PDP1 showed the strongest positive correlation with lactate score and the potential as a standalone biomarker for prognosis. In general, our study has yielded crucial insights into the potential application of the lactate score as a predictive biomarker for both survival outcomes and the response to immunotherapy. By recognizing the prognostic significance of lactate metabolism, we open avenues for further investigations aimed at harnessing the therapeutic potential of lactate.
Collapse
Affiliation(s)
- Ying Deng
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Disease of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Guoqiang Zhu
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Xiao Mi
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Xianyang, China
| | - Xiaoyu Jing
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Disease of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| |
Collapse
|
27
|
Ngai D, Schilperoort M, Tabas I. Efferocytosis-induced lactate enables the proliferation of pro-resolving macrophages to mediate tissue repair. Nat Metab 2023; 5:2206-2219. [PMID: 38012414 PMCID: PMC10782856 DOI: 10.1038/s42255-023-00921-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 10/05/2023] [Indexed: 11/29/2023]
Abstract
The clearance of apoptotic cells by macrophages (efferocytosis) prevents necrosis and inflammation and activates pro-resolving pathways, including continual efferocytosis. A key resolution process in vivo is efferocytosis-induced macrophage proliferation (EIMP), in which apoptotic cell-derived nucleotides trigger Myc-mediated proliferation of pro-resolving macrophages. Here we show that EIMP requires a second input that is integrated with cellular metabolism, notably efferocytosis-induced lactate production. Lactate signalling via GPR132 promotes Myc protein stabilization and subsequent macrophage proliferation. This mechanism is validated in vivo using a mouse model of dexamethasone-induced thymocyte apoptosis, which elevates apoptotic cell burden and requires efferocytosis to prevent inflammation and necrosis. Thus, EIMP, a key process in tissue resolution, requires inputs from two independent processes: a signalling pathway induced by apoptotic cell-derived nucleotides and a cellular metabolism pathway involving lactate production. These findings illustrate how seemingly distinct pathways in efferocytosing macrophages are integrated to carry out a key process in tissue resolution.
Collapse
Affiliation(s)
- David Ngai
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| | - Maaike Schilperoort
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Ira Tabas
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
- Departments of Physiology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
28
|
Paul SK, Dutta Chowdhury K, Dey SR, Paul A, Haldar R. Exploring the possibility of drug repurposing for cancer therapy targeting human lactate dehydrogenase A: a computational approach. J Biomol Struct Dyn 2023; 41:9967-9976. [PMID: 36576127 DOI: 10.1080/07391102.2022.2158134] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 11/12/2022] [Indexed: 12/29/2022]
Abstract
Human lactate dehydrogenase A (LDHA) is an anaerobic glycolytic enzyme involved in the inter-conversion of pyruvate to lactate. The level of LDHA in various types of cancer cells is found to be elevated and the dependence of cancer cells on anaerobic glycolysis is viewed as the reason for this elevation. Moreover, inhibition of LDHA activity has been shown to be effective in impairing the growth of tumors, making the LDHA as a potential target for cancer therapy. In this computational study, we have performed a pharmacophore based screening of approved drugs followed by a molecular docking based screening to find a few potential LDHA inhibitors. Molecular dynamics simulations have also been performed to examine the stability of the LDHA-drug complexes as obtained from the docking study. The result of the study showed that darunavir, moxalactam and eprosartan can bind to the active site of LDHA with high affinity in comparison to two known synthetic inhibitors of LDHA. The results of the molecular dynamics simulation showed that these drugs can bind stably with the enzyme through hydrogen bond and hydrophobic interactions. Hence, it is concluded that darunavir, moxalactam and eprosartan may be considered as potential inhibitors of LDHA and can be used for cancer therapy after proper validation of their effectiveness through in vitro, in vivo and clinical trials.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Sanjay Kumar Paul
- Department of Zoology, Rammohan College, Kolkata, West Bengal, India
| | | | - Santi Ranjan Dey
- Department of Zoology, Rammohan College, Kolkata, West Bengal, India
| | - Ayantika Paul
- Department of Physiology, University of Calcutta, Kolkata, West Bengal, India
| | - Rajen Haldar
- Department of Physiology, University of Calcutta, Kolkata, West Bengal, India
| |
Collapse
|
29
|
Han JH, Lee EJ, Park W, Ha KT, Chung HS. Natural compounds as lactate dehydrogenase inhibitors: potential therapeutics for lactate dehydrogenase inhibitors-related diseases. Front Pharmacol 2023; 14:1275000. [PMID: 37915411 PMCID: PMC10616500 DOI: 10.3389/fphar.2023.1275000] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 09/27/2023] [Indexed: 11/03/2023] Open
Abstract
Lactate dehydrogenase (LDH) is a crucial enzyme involved in energy metabolism and present in various cells throughout the body. Its diverse physiological functions encompass glycolysis, and its abnormal activity is associated with numerous diseases. Targeting LDH has emerged as a vital approach in drug discovery, leading to the identification of LDH inhibitors among natural compounds, such as polyphenols, alkaloids, and terpenoids. These compounds demonstrate therapeutic potential against LDH-related diseases, including anti-cancer effects. However, challenges concerning limited bioavailability, poor solubility, and potential toxicity must be addressed. Combining natural compounds with LDH inhibitors has led to promising outcomes in preclinical studies. This review highlights the promise of natural compounds as LDH inhibitors for treating cancer, cardiovascular, and neurodegenerative diseases.
Collapse
Affiliation(s)
- Jung Ho Han
- Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu, Republic of Korea
| | - Eun-Ji Lee
- Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu, Republic of Korea
| | - Wonyoung Park
- Korean Convergence Medical Science Major, KIOM Campus, University of Science and Technology (UST), Daegu, Republic of Korea
| | - Ki-Tae Ha
- Korean Convergence Medical Science Major, KIOM Campus, University of Science and Technology (UST), Daegu, Republic of Korea
| | - Hwan-Suck Chung
- Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu, Republic of Korea
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Republic of Korea
| |
Collapse
|
30
|
Ji P, Yang K, Xu Q, Qin G, Zhu Q, Qian Y, Yao W. Mechanisms and Application of Gas-Based Anticancer Therapies. Pharmaceuticals (Basel) 2023; 16:1394. [PMID: 37895865 PMCID: PMC10609769 DOI: 10.3390/ph16101394] [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: 08/15/2023] [Revised: 09/20/2023] [Accepted: 09/27/2023] [Indexed: 10/29/2023] Open
Abstract
Cancer is still one of the major factors threatening public health, with morbidity and mortality rates at the forefront of the world. Clinical drawbacks, such as high toxicity and side effects of drug therapy, and easy recurrence after surgery affect its therapeutic effect. Gas signaling molecules are essential in maintaining biological homeostasis and physiological functions as specific chemical substances for biological information transfer. In recent years, the physiological regulatory functions of gas molecules in the cancer process have been gradually revealed and have shown broad application prospects in tumor therapy. In this paper, standard gas therapies are classified and introduced. Taking H2, CO2, NO, CO, H2S, and SO2 gases as examples, the research progress and application of gas therapies in malignant tumors are mainly introduced in terms of biological characteristics, anticancer mechanisms, and treatment strategies. Finally, the problems and prospects for developing gases as anticancer drugs are outlined.
Collapse
Affiliation(s)
- Peng Ji
- College of Pharmacy and Chemistry & Chemical Engineering, Jiangsu Provincial Key Laboratory of Chiral Pharmaceutical Chemicals Biologically Manufacturing, Taizhou University, Taizhou 225300, China
| | - Kexin Yang
- College of Pharmacy and Chemistry & Chemical Engineering, Jiangsu Provincial Key Laboratory of Chiral Pharmaceutical Chemicals Biologically Manufacturing, Taizhou University, Taizhou 225300, China
| | - Qingqing Xu
- College of Pharmacy and Chemistry & Chemical Engineering, Jiangsu Provincial Key Laboratory of Chiral Pharmaceutical Chemicals Biologically Manufacturing, Taizhou University, Taizhou 225300, China
| | - Guilin Qin
- College of Pharmacy and Chemistry & Chemical Engineering, Jiangsu Provincial Key Laboratory of Chiral Pharmaceutical Chemicals Biologically Manufacturing, Taizhou University, Taizhou 225300, China
| | - Qianyu Zhu
- College of Pharmacy and Chemistry & Chemical Engineering, Jiangsu Provincial Key Laboratory of Chiral Pharmaceutical Chemicals Biologically Manufacturing, Taizhou University, Taizhou 225300, China
| | - Ying Qian
- College of Pharmacy and Chemistry & Chemical Engineering, Jiangsu Provincial Key Laboratory of Chiral Pharmaceutical Chemicals Biologically Manufacturing, Taizhou University, Taizhou 225300, China
| | - Wenshui Yao
- Department of Anesthesiology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, China
| |
Collapse
|
31
|
Batheja S, Sahoo RK, Tarannum S, Vaiphei KK, Jha S, Alexander A, Goyal AK, Gupta U. Hepatocellular carcinoma: Preclinical and clinical applications of nanotechnology with the potential role of carbohydrate receptors. Biochim Biophys Acta Gen Subj 2023; 1867:130443. [PMID: 37573973 DOI: 10.1016/j.bbagen.2023.130443] [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: 02/15/2023] [Revised: 08/04/2023] [Accepted: 08/09/2023] [Indexed: 08/15/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common types of liver cancer; accounts for 75-85% of cases. The treatment and management of HCC involve different sanative options like surgery, chemotherapy, immunotherapy, etc. Recently, various advancements have been introduced for the diagnosis and targeting of hepatic tumor cells. Among these, biomarkers are considered the primary source for the diagnosis and differentiation of tumor cells. With the advancement in the field of nanotechnology, different types of nanocarriers have been witnessed in tumor targeting. Nanocarriers such as nanoparticles, liposomes, polymeric micelles, nanofibers, etc. are readily prepared for effective tumor targeting with minimal side-effects. The emergence of various approaches tends to improve the effectiveness of these nanocarriers as demonstrated in ample clinical trials. This review focuses on the significant role of carbohydrates such as mannose, galactose, fructose, etc. in the development, diagnosis, and therapy of HCC. Hence, the current focus of this review is to acknowledge various perspectives regarding the occurrence, diagnosis, treatment, and management of HCC.
Collapse
Affiliation(s)
- Sanya Batheja
- Nanopolymeric Drug Delivery Lab, Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan 305817, India
| | - Rakesh Kumar Sahoo
- Nanopolymeric Drug Delivery Lab, Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan 305817, India
| | - Sofiya Tarannum
- Nanopolymeric Drug Delivery Lab, Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan 305817, India
| | - Klaudi K Vaiphei
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sila Katamur, Changsari, Kamrup, Guwahati, Assam 781101, India
| | - Shikha Jha
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sila Katamur, Changsari, Kamrup, Guwahati, Assam 781101, India
| | - Amit Alexander
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sila Katamur, Changsari, Kamrup, Guwahati, Assam 781101, India
| | - Amit Kumar Goyal
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan 305817, India
| | - Umesh Gupta
- Nanopolymeric Drug Delivery Lab, Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan 305817, India.
| |
Collapse
|
32
|
Joo HY, Jung JK, Kim MY, Woo SR, Jeong JM, Park ER, Kim YM, Park JJ, Kim J, Yun M, Shin HJ, Lee KH. NADH elevation during chronic hypoxia leads to VHL-mediated HIF-1α degradation via SIRT1 inhibition. Cell Biosci 2023; 13:182. [PMID: 37777750 PMCID: PMC10543270 DOI: 10.1186/s13578-023-01130-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 09/08/2023] [Indexed: 10/02/2023] Open
Abstract
BACKGROUND Under conditions of hypoxia, cancer cells with hypoxia inducible factor-1α (HIF-1α) from heterogeneous tumor cells show greater aggression and progression in an effort to compensate for harsh environmental conditions. Extensive study on the stability of HIF-1α under conditions of acute hypoxia in cancer progression has been conducted, however, understanding of its involvement during the chronic phase is limited. METHODS In this study, we investigated the effect of SIRT1 on HIF1 stability in a typical chronic hypoxic conditon that maintains cells for 24 h under hypoxia using Western blotting, co-IP, measurement of intracellular NAD + and NADH levels, semi-quantitative RT-PCR analysis, invasion assay, gene knockdown. RESULTS Here we demonstrated that the high concentration of pyruvate in the medium, which can be easily overlooked, has an effect on the stability of HIF-1α. We also demonstrated that NADH functions as a signal for conveyance of HIF-1α degradation via the SIRT1 and VHL signaling pathway under conditions of chronic hypoxia, which in turn leads to attenuation of hypoxically strengthened invasion and angiogenic activities. A steep increase in the level of NADH occurs during chronic hypoxia, leading to upregulation of acetylation and degradation of HIF-1α via inactivation of SIRT1. Of particular interest, p300-mediated acetylation at lysine 709 of HIF-1α is recogonized by VHL, which leads to degradation of HIF-1α via ubiquitin/proteasome machinary under conditions of chronic hypoxia. In addition, we demonstrated that NADH-elevation-induced acetylation and subsequent degradation of HIF-1α was independent of proline hydroxylation. CONCLUSIONS Our findings suggest a critical role of SIRT1 as a metabolic sensor in coordination of hypoxic status via regulation of HIF-1α stability. These results also demonstrate the involvement of VHL in degradation of HIF-1α through recognition of PHD-mediated hydroxylation in normoxia and p300-mediated HIF-1α acetylation in hypoxia.
Collapse
Affiliation(s)
- Hyun-Yoo Joo
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
- Lab. of Biochemistry, School of Life Sciences & Biotechnology, Korea University, Seoul, Korea
| | - Jin Kyu Jung
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
- Laboratory of Toxicology, College of Veterinary Medicine, Seoul National University, Seoul, Korea
- Neuro-Oncology Branch, The Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mi-Yeon Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Seon Rang Woo
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
- Department of Otolaryngology-Head and Neck Surgery, Kyung Hee University School of Medicine, Hyung Hee University Medical Center, Seoul, Republic of Korea
| | - Jae Min Jeong
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Eun-Ran Park
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Yong-Min Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Joong-Jean Park
- Department of Physiology, College of Medicine, Korea University, Seoul, Korea
| | - Joon Kim
- Lab. of Biochemistry, School of Life Sciences & Biotechnology, Korea University, Seoul, Korea
| | - Miyong Yun
- Department of Bioindustry and Bioresource Engineering, College of Life Sciences, Sejong University, Seoul, Korea.
| | - Hyun-Jin Shin
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea.
| | - Kee-Ho Lee
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea.
| |
Collapse
|
33
|
Guo W, Zhao G, Liu S, Deng T, Zhang G, Zhang B. Development of the prognostic value in lung adenocarcinoma based on anoikis-related genes and initial experimental validation. J Gene Med 2023; 25:e3534. [PMID: 37259225 DOI: 10.1002/jgm.3534] [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: 03/25/2023] [Revised: 04/21/2023] [Accepted: 05/01/2023] [Indexed: 06/02/2023] Open
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is a highly aggressive cancer in advanced stages and has the highest cancer-related death across the world. Anoikis has emerged as a specific form of apoptotic cell death that may play a vital role in the formation and development of tumors. METHODS Based on The Cancer Genome Atlas dataset, we developed a novel anoikis-related genes (ARGs) signature in LUAD and evaluated the differences between low and high-risk groups in clinical characteristics, expression patterns, immune cell infiltration, and drug sensitivity, etc. According to multivariate Cox regression analysis, the risk score was identified as a significant independent prognostic factor. The possible biological pathways of ARGs' were assessed by Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses. The immune infiltration landscape and risk score of ARGs were analyzed by ESTIMATE and CIBERSORT analysis. A nomogram grounded on six key ARGs and clinicopathological features was provided. Moreover, experiment validation of the expression patterns of six hub ARGs in lung cancer cell lines was conducted. RESULTS We identified 53 survival-related LUAD anoikis-related differentially expressed genes and finally six hub anoikis genes (LDHA, SLC2A1, SERPINB5, ITGB4, BRCA2, and PIK3R1) were selected to construct an ARG model. The risk model could efficiently cluster the patients into low- and high-risk groups which could accurately predict clinical outcomes for LUAD patients. There is evidence that the prognostic risk score is a remarkable prognostic factor in determining overall survival. Different immune statuses and drug sensitivity between low- and high-risk groups were explored according to functional analysis. On the basis of risk scores and LUAD clinicopathological features, a novel nomogram was developed. Ultimately, all six key genes except for PIK3R1 were proved to be upregulated in LUAD tissues and cell lines by bioinformatics analysis and experimental validation. CONCLUSIONS The result of the present study suggest that ARGs could be carcinogenic to LUAD and could be used as an effective stratification factor to customize therapies and forecast the survival rate in LUAD patients.
Collapse
Affiliation(s)
- Wenwei Guo
- Department of Thoracic Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Cardiothoracic Surgery, Ji 'an Central People's Hospital, Ji'an, China
| | - Guang Zhao
- Department of Thoracic Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Suping Liu
- Department of Rehabilitation Medicine, Ji'an Central People's Hospital, Ji'an, China
| | - Tao Deng
- Department of Pain, Ji 'an Central People's Hospital, Ji'an, China
| | - Guangjian Zhang
- Department of Thoracic Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Boxiang Zhang
- Department of Thoracic Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
34
|
Tian Q, Guo Y, Liu J, Pang C, Wang Q, Xie Q, Li J. CircDUS2L (circ_0039908) promotes lung adenocarcinoma progression by upregulating PGAM1 by acting as a miR-590-5p molecular sponge. J Biochem Mol Toxicol 2023; 37:e23406. [PMID: 37392398 DOI: 10.1002/jbt.23406] [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: 11/23/2022] [Revised: 04/14/2023] [Accepted: 06/08/2023] [Indexed: 07/03/2023]
Abstract
Lung adenocarcinoma (LUAD) is usually found at the metastatic stage. Circular RNA dihydrouridine synthase 2-like (DUS2L) (circDUS2L) has been discovered to be upregulated in LUAD. Nevertheless, the function of circDUS2L in LUAD has not been verified. Levels of circDUS2L, microRNA-590-5p (miR-590-5p), and phosphoglycerate mutase 1 (PGAM1) mRNA were analyzed using quantitative real-time polymerase chain reaction (RT-qPCR). Cell proliferation, apoptosis, metastasis, and invasion were assessed by 3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide (MTT), colony formation, 5-ethynyl-2'-deoxyuridine (Edu), flow cytometry, and transwell assays. Protein levels were detected by western blotting. Cell glycolysis was analyzed by measuring cell glucose consumption, lactate production, and extracellular acidification rate (ECAR). The regulatory mechanism of circDUS2L in LUAD cells was analyzed by bioinformatics analysis, dual-luciferase reporter, RNA pull-down, and RNA immunoprecipitation (RIP) assays. Xenograft assay was conducted to confirm the function of circDUS2L in vivo. CircDUS2L was highly expressed in LUAD tissues and cells. CircDUS2L silencing constrained xenograft tumor growth in vivo. CircDUS2L knockdown induced apoptosis, repressed viability, colony formation, proliferation, metastasis, invasion, and glycolysis of LUAD cells in vitro by releasing miR-590-5p via functioning as a miR-590-5p sponge. MiR-590-5p was lowly expressed in LUAD tissues and cells, and miR-590-5p mimic curbed malignant behaviors and glycolysis of LUAD cells by targeting PGAM1. PGAM1 was overexpressed in LUAD tissues and cells, and circDUS2L sponged miR-590-5p to regulate PGAM1 expression. CircDUS2L elevated PGAM1 expression through functioning as a miR-590-5p sponge, thus driving malignant behaviors and glycolysis of LUAD cells.
Collapse
Affiliation(s)
- Qing Tian
- Department of Thoracic surgery, The First Hospital of Hebei Medical University, Shijiazhuang City, Hebei Province, China
| | - Ying Guo
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang City, Hebei Province, China
| | - Jinfeng Liu
- Department of Thoracic surgery, The First Hospital of Hebei Medical University, Shijiazhuang City, Hebei Province, China
| | - Chao Pang
- Department of Pathology, The First Hospital of Hebei Medical University, Shijiazhuang City, Hebei Province, China
| | - Qiang Wang
- Department of Thoracic surgery, The First Hospital of Hebei Medical University, Shijiazhuang City, Hebei Province, China
| | - Qi Xie
- Department of Clinical Nutrition, The Fourth Hospital of Hebei Medical University, Shijiazhuang City, Hebei Province, China
| | - Jianke Li
- Department of Thoracic surgery, The First Hospital of Hebei Medical University, Shijiazhuang City, Hebei Province, China
| |
Collapse
|
35
|
Xing W, Li X, Zhou Y, Li M, Zhu M. Lactate metabolic pathway regulates tumor cell metastasis and its use as a new therapeutic target. EXPLORATION OF MEDICINE 2023:541-559. [DOI: https:/doi.org/10.37349/emed.2023.00160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/17/2023] [Indexed: 09/04/2023] Open
Abstract
Abnormal energy metabolism is one of the ten hallmarks of tumors, and tumor cell metabolism provides energy and a suitable microenvironment for tumorigenesis and metastasis. Tumor cells can consume large amounts of glucose and produce large amounts of lactate through glycolysis even in the presence of oxygen, a process called aerobic glycolysis, also known as the Warburg effect. Lactate is the end product of the aerobic glycolysis. Lactate dehydrogenase A (LDHA), which is highly expressed in cancer cells, promotes lactate production and transports lactate to the tumor microenvironment and is taken up by surrounding stromal cells under the action of monocarboxylate transporter 1/4 (MCT1/4), which in turn influences the immune response and enhances the invasion and metastasis of cancer cells. Therapeutic strategies targeting lactate metabolism have been intensively investigated, focusing on its metastasis-promoting properties and various target inhibitors; AZD3965, an MCT1 inhibitor, has entered phase I clinical trials, and the LDHA inhibitor N-hydroxyindole (NHI) has shown cancer therapeutic activity in pre-clinical studies. Interventions targeting lactate metabolism are emerging as a promising option for cancer therapy, with chemotherapy or radiotherapy combined with lactate-metabolism-targeted drugs adding to the effectiveness of cancer treatment. Based on current research, this article outlines the role of lactate metabolism in tumor metastasis and the potential value of inhibitors targeting lactate metabolism in cancer therapy.
Collapse
Affiliation(s)
- Weimei Xing
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou 571199, Hainan, China
| | - Xiaowei Li
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou 571199, Hainan, China
| | - Yuli Zhou
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou 571199, Hainan, China
| | - Mengsen Li
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou 571199, Hainan, China; Department of Medical Oncology, Second Affiliated Hospital, Hainan Medical University, Haikou 570311, Hainan, China; Institution of Tumour, First Affiliated Hospital, Hainan Medical University, Haikou 570102, Hainan, China
| | - Mingyue Zhu
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou 571199, Hainan, China
| |
Collapse
|
36
|
Shah FY, Shetty DC, Kaur G, Gupta S, Pathak A, Aggarwal V. Serum and Saliva LDH Levels in Patients with Habit, Oral Potentially Malignant Disorders (OPMDs), and Oral Squamous Cell Carcinoma (OSCC): A Spectrophotometry Study. JOURNAL OF PHARMACY AND BIOALLIED SCIENCES 2023; 15:S881-S884. [PMID: 37694076 PMCID: PMC10485454 DOI: 10.4103/jpbs.jpbs_90_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/04/2023] [Accepted: 02/05/2023] [Indexed: 09/12/2023] Open
Abstract
Background Oral potentially malignant disorders (OPMDs) are chronic lesions or conditions characterized by a potential for malignant transformation. One of the hallmarks of cancer is aerobic glycolysis which confers immortality to cancer cells, correlates with the belligerent behavior of various malignancies, and shows an increase in the expression of lactate dehydrogenase (LDH). The present study was conducted to measure and compare serum and salivary LDH levels in patients with habit, OPMDs and oral squamous cell carcinoma (OSCC) patients, and to evaluate if LDH levels can be used as a biomarker in the progression into potentially malignant disorders (PMDs), the prognosis of OSCC, and to assess if saliva is a better biomarker. Materials and Methods Thirty patients of both genders were divided into three groups. Group I had patients with habits, group II had OPMDs, and group III had OSCC with 10 subjects from each group. Saliva and serum of patients were collected according to the standard protocol and measured for LDH using spectrophotometry of 340 nm. Results Group I had patients with habits, group II had OPMDs, and group III had OSCC. Group I had 6 males and 4 females, group II had 8 males and 2 females, and group III had 7 males and 3 females. The mean serum LDH level in group I was 265.50, in group II was 194.10, and in group III was 224.22. The difference was non-significant (P > 0.05). The mean salivary LDH level in group I was 345.68, in group II was 532.72, and in group III was 1.105. The difference was significant (P < 0.05). Conclusion Salivary LDH is a non-invasive, cost-effective technique, which can be used as an effective modality for the diagnosis and prognosis of oral cancer as well as for monitoring tobacco users and OPMDs.
Collapse
Affiliation(s)
- Farnaz Yasmin Shah
- Department of Oral Pathology, Institute of Dental Studies and Technologies, Modinagar, Ghaziabad, Uttar Pradesh, India
| | - Devi Charan Shetty
- Department of Oral Pathology, I.T.S. Centre for Dental Studies and Research, Murad Nagar, Ghaziabad, Uttar Pradesh, India
| | - Geetpriya Kaur
- Department of Oral Pathology, Institute of Dental Studies and Technologies, Modinagar, Ghaziabad, Uttar Pradesh, India
| | - Sushma Gupta
- Department of Oral Pathology and Microbiology, RDCH Jaipur, Rajasthan, India
| | - Aparna Pathak
- Department of Oral Pathology, Paradise Diagnostics, New Delhi, India
| | | |
Collapse
|
37
|
Li Z, Gu Z, Wang L, Guan Y, Lyu Y, Zhang J, Wang Y, Wang X, Xiong J, Liu Y. Nuclear Translocation of LDHA Promotes the Catabolism of BCAAs to Sustain GBM Cell Proliferation through the TxN Antioxidant Pathway. Int J Mol Sci 2023; 24:ijms24119365. [PMID: 37298317 DOI: 10.3390/ijms24119365] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/15/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023] Open
Abstract
Glutamate is excitotoxic to neurons. The entry of glutamine or glutamate from the blood into the brain is limited. To overcome this, branched-chain amino acids (BCAAs) catabolism replenishes the glutamate in brain cells. Branched-chain amino acid transaminase 1 (BCAT1) activity is silenced by epigenetic methylation in IDH mutant gliomas. However, glioblastomas (GBMs) express wild type IDH. Here, we investigated how oxidative stress promotes BCAAs' metabolism to maintain intracellular redox balance and, consequently, the rapid progression of GBMs. We found that reactive oxygen species (ROS) accumulation promoted the nuclear translocation of lactate dehydrogenase A (LDHA), which triggered DOT1L (disruptor of telomeric silencing 1-like)-mediated histone H3K79 hypermethylation and enhanced BCAA catabolism in GBM cells. Glutamate derived from BCAAs catabolism participates in antioxidant thioredoxin (TxN) production. The inhibition of BCAT1 decreased the tumorigenicity of GBM cells in orthotopically transplanted nude mice, and prolonged their survival time. In GBM samples, BCAT1 expression was negatively correlated with the overall survival time (OS) of patients. These findings highlight the role of the non-canonical enzyme activity of LDHA on BCAT1 expression, which links the two major metabolic pathways in GBMs. Glutamate produced by the catabolism of BCAAs was involved in complementary antioxidant TxN synthesis to balance the redox state in tumor cells and promote the progression of GBMs.
Collapse
Affiliation(s)
- Zhujun Li
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Yixueyuan Rd. 138, Shanghai 200032, China
| | - Zhiyan Gu
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Yixueyuan Rd. 138, Shanghai 200032, China
| | - Lan Wang
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Yixueyuan Rd. 138, Shanghai 200032, China
| | - Yun Guan
- Cyberknife Center, Department of Neurosurgery, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Yingying Lyu
- Department of Neurosurgery, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
- Department of Oncology, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jialong Zhang
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Yixueyuan Rd. 138, Shanghai 200032, China
| | - Yin Wang
- Department of Pathology, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Xin Wang
- Cyberknife Center, Department of Neurosurgery, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Ji Xiong
- Department of Pathology, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Ying Liu
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Yixueyuan Rd. 138, Shanghai 200032, China
| |
Collapse
|
38
|
Malayil R, Chhichholiya Y, Vasudeva K, Singh HV, Singh T, Singh S, Munshi A. Oncogenic metabolic reprogramming in breast cancer: focus on signaling pathways and mitochondrial genes. Med Oncol 2023; 40:174. [PMID: 37170010 DOI: 10.1007/s12032-023-02037-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 04/20/2023] [Indexed: 05/13/2023]
Abstract
Oncogenic metabolic reprogramming impacts the abundance of key metabolites that regulate signaling and epigenetics. Metabolic vulnerability in the cancer cell is evident from the Warburg effect. The research on metabolism in the progression and survival of breast cancer (BC) is under focus. Oncogenic signal activation and loss of tumor suppressor are important regulators of tumor cell metabolism. Several intrinsic and extrinsic factors contribute to metabolic reprogramming. The molecular mechanisms underpinning metabolic reprogramming in BC are extensive and only partially defined. Various signaling pathways involved in the metabolism play a significant role in the modulation of BC. Notably, PI3K/AKT/mTOR pathway, lactate-ERK/STAT3 signaling, loss of the tumor suppressor Ras, Myc, oxidative stress, activation of the cellular hypoxic response and acidosis contribute to different metabolic reprogramming phenotypes linked to enhanced glycolysis. The alterations in mitochondrial genes have also been elaborated upon along with their functional implications. The outcome of these active research areas might contribute to the development of novel therapeutic interventions and the remodeling of known drugs.
Collapse
Affiliation(s)
- Rhuthuparna Malayil
- Department of Human Genetics and Molecular Medicine, Central University of Punjab Bathinda, Punjab, India
| | - Yogita Chhichholiya
- Department of Human Genetics and Molecular Medicine, Central University of Punjab Bathinda, Punjab, India
| | | | - Harsh Vikram Singh
- Department of Orthopedics, All India Institute of Medical Sciences, Bathinda, India
| | - Tashvinder Singh
- Department of Human Genetics and Molecular Medicine, Central University of Punjab Bathinda, Punjab, India
| | - Sandeep Singh
- Department of Human Genetics and Molecular Medicine, Central University of Punjab Bathinda, Punjab, India.
| | - Anjana Munshi
- Department of Human Genetics and Molecular Medicine, Central University of Punjab Bathinda, Punjab, India.
| |
Collapse
|
39
|
You M, Xie Z, Zhang N, Zhang Y, Xiao D, Liu S, Zhuang W, Li L, Tao Y. Signaling pathways in cancer metabolism: mechanisms and therapeutic targets. Signal Transduct Target Ther 2023; 8:196. [PMID: 37164974 PMCID: PMC10172373 DOI: 10.1038/s41392-023-01442-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 03/20/2023] [Accepted: 04/17/2023] [Indexed: 05/12/2023] Open
Abstract
A wide spectrum of metabolites (mainly, the three major nutrients and their derivatives) can be sensed by specific sensors, then trigger a series of signal transduction pathways and affect the expression levels of genes in epigenetics, which is called metabolite sensing. Life body regulates metabolism, immunity, and inflammation by metabolite sensing, coordinating the pathophysiology of the host to achieve balance with the external environment. Metabolic reprogramming in cancers cause different phenotypic characteristics of cancer cell from normal cell, including cell proliferation, migration, invasion, angiogenesis, etc. Metabolic disorders in cancer cells further create a microenvironment including many kinds of oncometabolites that are conducive to the growth of cancer, thus forming a vicious circle. At the same time, exogenous metabolites can also affect the biological behavior of tumors. Here, we discuss the metabolite sensing mechanisms of the three major nutrients and their derivatives, as well as their abnormalities in the development of various cancers, and discuss the potential therapeutic targets based on metabolite-sensing signaling pathways to prevent the progression of cancer.
Collapse
Affiliation(s)
- Mengshu You
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 410078, Changsha, Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 410078, Changsha, Hunan, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 410078, Changsha, Hunan, China
| | - Zhuolin Xie
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 410078, Changsha, Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 410078, Changsha, Hunan, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 410078, Changsha, Hunan, China
| | - Nan Zhang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 410078, Changsha, Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 410078, Changsha, Hunan, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 410078, Changsha, Hunan, China
| | - Yixuan Zhang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 410078, Changsha, Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 410078, Changsha, Hunan, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 410078, Changsha, Hunan, China
| | - Desheng Xiao
- Department of Pathology, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Shuang Liu
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Wei Zhuang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, People's Republic of China.
| | - Lili Li
- Cancer Epigenetics Laboratory, Department of Clinical Oncology, State Key Laboratory of Translational Oncology, Sir YK Pao Centre for Cancer and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Ma Liu Shui, Hong Kong.
| | - Yongguang Tao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 410078, Changsha, Hunan, China.
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 410078, Changsha, Hunan, China.
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 410078, Changsha, Hunan, China.
- Department of Thoracic Surgery, Hunan Key Laboratory of Early Diagnosis and Precision Therapy in Lung Cancer, Second Xiangya Hospital, Central South University, 410011, Changsha, China.
| |
Collapse
|
40
|
Zhang Y, Qin W, Zhang W, Qin Y, Zhou YL. Guidelines on lung adenocarcinoma prognosis based on immuno-glycolysis-related genes. Clin Transl Oncol 2023; 25:959-975. [PMID: 36447119 PMCID: PMC10025218 DOI: 10.1007/s12094-022-03000-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 10/29/2022] [Indexed: 12/05/2022]
Abstract
OBJECTIVES This study developed a new model for risk assessment of immuno-glycolysis-related genes for lung adenocarcinoma (LUAD) patients to predict prognosis and immunotherapy efficacy. METHODS LUAD samples and data obtained from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases are used as training and test columns, respectively. Twenty-two (22) immuno-glycolysis-related genes were screened, the patients diagnosed with LUAD were divided into two molecular subtypes by consensus clustering of these genes. The initial prognosis model was developed using the multiple regression analysis method and Receiver Operating characteristic (ROC) analysis was used to verify its predictive potential. Gene set enrichment analysis (GSEA) showed the immune activities and pathways in different risk populations, we calculated immune checkpoints, immune escape, immune phenomena (IPS), and tumor mutation burden (TMB) based on TCGA datasets. Finally, the relationship between the model and drug sensitivity was analyzed. RESULTS Fifteen (15) key differentially expressed genes (DEGs) with prognostic value were screened and a new prognostic model was constructed. Four hundred and forty-three (443) samples were grouped into two different risk cohorts based on median model risk values. It was observed that survival rates in high-risk groups were significantly low. ROC curves were used to evaluate the model's accuracy in determining the survival time and clinical outcome of LUAD patients. Cox analysis of various clinical factors proved that the risk score has great potential as an independent prognostic factor. The results of immunological analysis can reveal the immune infiltration and the activity of related functions in different pathways in the two risk groups, and immunotherapy was more effective in low-risk patients. Most chemotherapeutic agents are more sensitive to low-risk patients, making them more likely to benefit. CONCLUSION A novel prognostic model for LUAD patients was established based on IGRG, which could more accurately predict the prognosis and an effective immunotherapy approach for patients.
Collapse
Affiliation(s)
- Yuting Zhang
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, Jiangsu, China
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, Research Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Wen Qin
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, Jiangsu, China
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, Research Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Wenhui Zhang
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, Jiangsu, China
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, Research Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Yi Qin
- Nursing Department, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
| | - You Lang Zhou
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
41
|
Marrero-Rodríguez D, Taniguchi-Ponciano K, Kerbel J, Cano-Zaragoza A, Remba-Shapiro I, Silva-Román G, Vela-Patiño S, Andonegui-Elguera S, Valenzuela-Perez A, Mercado M. The hallmarks of cancer… in pituitary tumors? Rev Endocr Metab Disord 2023; 24:177-190. [PMID: 36586070 DOI: 10.1007/s11154-022-09777-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/07/2022] [Indexed: 01/01/2023]
Abstract
Over 20 years ago, Hanahan and Weinberg published a seminal review that addressed the biological processes that underly malignant transformation. This classical review, along with two revisions published in 2011 and 2022, has remain a classic of the oncology literature. Since many of the addressed biological processes may apply to non-malignant tumorigenesis, we evaluated to what extent these hallmarks pertain to the development of pituitary adenomas.Some of the biological processes analyzed in this review include genome instability generated by somatic USP8 and GNAS mutations in Cushing's diseases and acromegaly respectively; non-mutational epigenetic reprograming through changes in methylation; induction of angiogenesis through alterations of VEGF gene expression; promotion of proliferative signals mediated by EGFR; evasion of growth suppression by disrupting cyclin dependent kinase inhibitors; avoidance of immune destruction; and the promotion of inflammation mediated by alteration of gene expression of immune check points. We also elaborate further on the existence of oncogene induced senescence in pituitary tumors. We conclude that a better understanding of these processes can help us dilucidated why pituitary tumors are so resistant to malignant transformation and can potentially contribute to the development of novel anticancer treatments.
Collapse
Affiliation(s)
- Daniel Marrero-Rodríguez
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, México, D.F., 06720, Mexico City, Mexico
| | - Keiko Taniguchi-Ponciano
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, México, D.F., 06720, Mexico City, Mexico.
| | - Jacobo Kerbel
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, México, D.F., 06720, Mexico City, Mexico
| | - Amayrani Cano-Zaragoza
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, México, D.F., 06720, Mexico City, Mexico
| | - Ilan Remba-Shapiro
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, México, D.F., 06720, Mexico City, Mexico
| | - Gloria Silva-Román
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, México, D.F., 06720, Mexico City, Mexico
| | - Sandra Vela-Patiño
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, México, D.F., 06720, Mexico City, Mexico
| | - Sergio Andonegui-Elguera
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, México, D.F., 06720, Mexico City, Mexico
| | - Alejandra Valenzuela-Perez
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, México, D.F., 06720, Mexico City, Mexico
| | - Moisés Mercado
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, México, D.F., 06720, Mexico City, Mexico.
| |
Collapse
|
42
|
Wang Q, Atluri K, Tiwari AK, Babu RJ. Exploring the Application of Micellar Drug Delivery Systems in Cancer Nanomedicine. Pharmaceuticals (Basel) 2023; 16:ph16030433. [PMID: 36986532 PMCID: PMC10052155 DOI: 10.3390/ph16030433] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/07/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023] Open
Abstract
Various formulations of polymeric micelles, tiny spherical structures made of polymeric materials, are currently being investigated in preclinical and clinical settings for their potential as nanomedicines. They target specific tissues and prolong circulation in the body, making them promising cancer treatment options. This review focuses on the different types of polymeric materials available to synthesize micelles, as well as the different ways that micelles can be tailored to be responsive to different stimuli. The selection of stimuli-sensitive polymers used in micelle preparation is based on the specific conditions found in the tumor microenvironment. Additionally, clinical trends in using micelles to treat cancer are presented, including what happens to micelles after they are administered. Finally, various cancer drug delivery applications involving micelles are discussed along with their regulatory aspects and future outlooks. As part of this discussion, we will examine current research and development in this field. The challenges and barriers they may have to overcome before they can be widely adopted in clinics will also be discussed.
Collapse
Affiliation(s)
- Qi Wang
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
| | - Keerthi Atluri
- Product Development Department, Alcami Corporation, Morrisville, NC 27560, USA
| | - Amit K. Tiwari
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH 43614, USA
- Department of Cell and Cancer Biology, University of Toledo, Toledo, OH 43614, USA
| | - R. Jayachandra Babu
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
- Correspondence:
| |
Collapse
|
43
|
Transcriptome Analysis on Hepatopancreas Reveals the Metabolic Dysregulation Caused by Vibrio parahaemolyticus Infection in Litopenaeus vannamei. BIOLOGY 2023; 12:biology12030417. [PMID: 36979109 PMCID: PMC10044748 DOI: 10.3390/biology12030417] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/19/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023]
Abstract
Acute hepatopancreas necrosis disease (AHPND) has caused massive deaths of shrimp and has led to huge economic losses in aquaculture. Vibrio parahaemolyticus (VPAHPND) carrying a plasmid encoding binary toxins homologous to the photorhabdus insect-related (Pir) toxins is one of the main pathogens causing this disease. Previous studies have reported many immune-related genes of shrimp in response to this pathogenic bacteria. However, few studies have so far focused on the metabolic changes in Litopenaeus vannamei upon VPAHPND infection. In the present study, comparative transcriptomic analysis was performed on the hepatopancreas of shrimp at different times during VPAHPND infection. Functional analyses on the differentially expressed genes (DEGs) during infection showed that pathways related to glucose, energy and amino acid metabolism, as well as nucleic acid synthesis, were obviously changed in the hepatopancreas after VPAHPND infection. Additionally, three signaling pathways, which could regulate metabolic processes, including HIF-1 signaling pathway, PI3K-Akt signaling pathway and NF-KappaB signaling pathway, also changed significantly. Collectively, these data reveal a close relationship between host metabolism processes and Vibrio infection. The information will enrich our understanding of the interaction mechanism between the shrimp and Vibrio.
Collapse
|
44
|
Lactate dehydrogenase: relationship with the diagnostic GLIM criterion for cachexia in patients with advanced cancer. Br J Cancer 2023; 128:760-765. [PMID: 36517550 PMCID: PMC9977728 DOI: 10.1038/s41416-022-02099-5] [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: 08/04/2022] [Revised: 11/23/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Although suggestive of dysregulated metabolism, the relationship between serum LDH level, phenotypic/aetiologic diagnostic Global Leadership Initiative on Malnutrition (GLIM) criteria and survival in patients with advanced cancer has yet to examined. METHODS Prospectively collected data from patients with advanced cancer, undergoing anti-cancer therapy with palliative intent, across nine sites in the UK and Ireland between 2011-2016, was retrospectively analysed. LDH values were grouped as <250/250-500/>500 Units/L. Relationships were examined using χ2 test for linear-by-linear association and binary logistics regression analysis. RESULTS A total of 436 patients met the inclusion criteria. 46% (n = 200) were male and 59% (n = 259) were ≥65 years of age. The median serum LDH was 394 Units/L and 33.5% (n = 146) had an LDH > 500 Units/L. LDH was significantly associated with ECOG-PS (p < 0.001), NLR (p < 0.05), mGPS (p < 0.05) and 3-month survival (p < 0.001). LDH was significantly associated with 3-month survival independent of weight loss (p < 0.01), BMI (p < 0.05), skeletal muscle mass (p < 0.01), metastatic disease (p < 0.05), NLR (p < 0.05) and mGPS (p < 0.01). DISCUSSION LDH was associated with performance status, systemic inflammation and survival in patients with advanced cancer. LDH measurement may be considered as an aetiologic criteria and become a potential therapeutic target in the treatment of cancer cachexia.
Collapse
|
45
|
Brooks GA, Osmond AD, Arevalo JA, Duong JJ, Curl CC, Moreno-Santillan DD, Leija RG. Lactate as a myokine and exerkine: drivers and signals of physiology and metabolism. J Appl Physiol (1985) 2023; 134:529-548. [PMID: 36633863 PMCID: PMC9970662 DOI: 10.1152/japplphysiol.00497.2022] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
No longer viewed as a metabolic waste product and cause of muscle fatigue, a contemporary view incorporates the roles of lactate in metabolism, sensing and signaling in normal as well as pathophysiological conditions. Lactate exists in millimolar concentrations in muscle, blood, and other tissues and can rise more than an order of magnitude as the result of increased production and clearance limitations. Lactate exerts its powerful driver-like influence by mass action, redox change, allosteric binding, and other mechanisms described in this article. Depending on the condition, such as during rest and exercise, following carbohydrate nutrition, injury, or pathology, lactate can serve as a myokine or exerkine with autocrine-, paracrine-, and endocrine-like functions that have important basic and translational implications. For instance, lactate signaling is: involved in reproductive biology, fueling the heart, muscle adaptation, and brain executive function, growth and development, and a treatment for inflammatory conditions. Lactate also works with many other mechanisms and factors in controlling cardiac output and pulmonary ventilation during exercise. Ironically, lactate can be disruptive of normal processes such as insulin secretion when insertion of lactate transporters into pancreatic β-cell membranes is not suppressed, and in carcinogenesis when factors that suppress carcinogenesis are inhibited, whereas factors that promote carcinogenesis are upregulated. Lactate signaling is important in areas of intermediary metabolism, redox biology, mitochondrial biogenesis, neurobiology, gut physiology, appetite regulation, nutrition, and overall health and vigor. The various roles of lactate as a myokine and exerkine are reviewed.NEW & NOTEWORTHY Lactate sensing and signaling is a relatively new and rapidly changing field. As a physiological signal lactate works both independently and in concert with other signals. Lactate operates via covalent binding and canonical signaling, redox change, and lactylation of DNA. Lactate can also serve as an element of feedback loops in cardiopulmonary regulation. From conception through aging lactate is not the only a myokine or exerkine, but it certainly deserves consideration as a physiological signal.
Collapse
Affiliation(s)
- George A Brooks
- Exercise Physiology Laboratory, Department of Integrative Biology, University of California, Berkeley, California, United States
| | - Adam D Osmond
- Exercise Physiology Laboratory, Department of Integrative Biology, University of California, Berkeley, California, United States
| | - Jose A Arevalo
- Exercise Physiology Laboratory, Department of Integrative Biology, University of California, Berkeley, California, United States
| | - Justin J Duong
- Exercise Physiology Laboratory, Department of Integrative Biology, University of California, Berkeley, California, United States
| | - Casey C Curl
- Exercise Physiology Laboratory, Department of Integrative Biology, University of California, Berkeley, California, United States
| | - Diana D Moreno-Santillan
- Exercise Physiology Laboratory, Department of Integrative Biology, University of California, Berkeley, California, United States
| | - Robert G Leija
- Exercise Physiology Laboratory, Department of Integrative Biology, University of California, Berkeley, California, United States
| |
Collapse
|
46
|
Yang Q, He Y, Tian L, Zhang Z, Qiu L, Tao X, Wei H. Anti-tumor effect of infant-derived Enterococcus via the inhibition of proliferation and inflammation as well as the promotion of apoptosis. Food Funct 2023; 14:2223-2238. [PMID: 36757840 DOI: 10.1039/d2fo03045d] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Probiotic Enterococcus hirae WEHI01 and Enterococcus faecium WEFA23 from infants were previously found to effectively inhibit the development of melanoma. In this study, their immunomodulatory and antitumor mechanisms were systemically studied. In vitro assay showed that E. hirae WEHI01 and E. faecium WEFA23 achieved biphasic immune regulation, which was revealed by the activation of resting spleen lymphocytes and RAW264.7 macrophages, as well as the anti-inflammation effect when immune cells were treated with LPS. The antitumor effects of E. hirae WEHI01 and E. faecium WEFA23 in vitro and vivo were then investigated. CCK8 and the cell scratch assay showed that the conditioned media, which were co-incubated with Enterococcus and spleen lymphocytes, significantly inhibited the proliferation and migration of B16F10, HepG-2 and HT-29 cells. The results of the tumor-bearing mice model experiment showed that E. faecium WEFA23 inhibition of the growth of tumors in mice, and the anti-tumor mechanism involved three aspects, namely tumor proliferation (decreasing expressions of LDHA, VEGF, MMP2, MMP9 and HIF-1α), inhibition of the pro-inflammation state (decreasing expressions of IL-6, TGF-β and IL-17) and the promotion of apoptosis (increasing expression of Bax/Bcl-2, caspase-3 and p53). The results suggest that the two strains of Enterococcus could be promising candidates for treating melanoma with a highly inhibitory effect.
Collapse
Affiliation(s)
- Qin Yang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, 330047, P. R. China.
| | - Yao He
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, 330047, P. R. China.
| | - Linlin Tian
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, 330047, P. R. China.
| | - Zhihong Zhang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, 330047, P. R. China.
| | - Liang Qiu
- Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 330004, P. R. China
| | - Xueying Tao
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, 330047, P. R. China.
| | - Hua Wei
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, 330047, P. R. China.
| |
Collapse
|
47
|
Serum lactate dehydrogenase is associated with impaired lung function: NHANES 2011-2012. PLoS One 2023; 18:e0281203. [PMID: 36730242 PMCID: PMC9894433 DOI: 10.1371/journal.pone.0281203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 01/17/2023] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Serum lactate dehydrogenase levels reflect disease status in a variety of organs, but its role in indicating pulmonary function is not yet clear. Therefore, this study explored the correlation between pulmonary function and serum lactate dehydrogenase, and investigated thresholds for changes in pulmonary function indicators in the total population as well as in different strata of the population. METHODS Based on data from the National Health and Nutrition Examination Survey (NHANES) 2011-2012 (n = 3453), univariate and stratified analyses were performed to investigate factors associated with pulmonary function, and multiple regression analysis was used to further investigate the specific relationship with serum lactate dehydrogenase. Smoothed curve fitting, threshold effect and saturation effect analysis were used to explore the threshold level of serum lactate dehydrogenase at the onset of changes in pulmonary function indicators. RESULTS Adjusted smoothed curve fit plots showed a linear relationship between serum lactate dehydrogenase levels and forced vital capacity and forced expiratory volume in one second: for each 1 U/L increase in serum lactate dehydrogenase levels, forced vital capacity decreased by 1.24 mL (95% CI = -2.05, -0.42, P = 0.0030) and forced expiratory volume in one second by 1.11 mL (95% CI = -1.82, -0.39, P = 0.0025). CONCLUSIONS Serum lactate dehydrogenase was negatively and linearly correlated with pulmonary function indices in the total population analyzed. Based on the total population and different population stratifications, this study determined the threshold values of serum lactate dehydrogenase at the onset of decline of pulmonary function in different populations. This provides a new serological monitoring indicator for patients suffering from respiratory diseases and has implications for patients with possible clinical impairment of pulmonary function. However, our cross-sectional study was not able to determine a causal relationship between these two factors, and further research is needed.
Collapse
|
48
|
Label-Free Proteomics of Oral Mucosa Tissue to Identify Potential Biomarkers That Can Flag Predilection of Precancerous Lesions to Oral Cell Carcinoma: A Preliminary Study. DISEASE MARKERS 2023; 2023:1329061. [PMID: 36776920 PMCID: PMC9908334 DOI: 10.1155/2023/1329061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/09/2023] [Accepted: 01/11/2023] [Indexed: 02/05/2023]
Abstract
Oral squamous cell carcinomas are mostly preceded by precancerous lesions such as leukoplakia and erythroplakia. Our study is aimed at identifying potential biomarker proteins in precancerous lesions of leukoplakia and erythroplakia that can flag their transformation to oral cancer. Four biological replicate samples from clinical phenotypes of healthy control, leukoplakia, erythroplakia, and oral carcinoma were annotated based on clinical screening and histopathological evaluation of buccal mucosa tissue. Differentially expressed proteins were delineated using a label-free quantitative proteomic experiment done on an Orbitrap Fusion Tribrid mass spectrometer in three technical replicate sets of samples. Raw files were processed using MaxQuant version 2.0.1.0, and downstream analysis was done via Perseus version 1.6.15.0. Validation included functional annotation based on biological processes and pathways using the ClueGO plug-in of Cytoscape. Hierarchical clustering and principal component analysis were performed using the ClustVis tool. Across control, leukoplakia, and cancer, L-lactate dehydrogenase A chain, plectin, and WD repeat-containing protein 1 were upregulated, whereas thioredoxin 1 and spectrin alpha chain, nonerythrocytic 1 were downregulated. Across control, erythroplakia, and cancer, L-lactate dehydrogenase A chain was upregulated whereas aldehyde dehydrogenase 2, peroxiredoxin 1, heat shock 70 kDa protein 1B, and spectrin alpha chain, nonerythrocytic 1 were downregulated. We found that proteins involved in leukoplakia were associated with alteration in cytoskeletal disruption and glycolysis, while in erythroplakia, they were associated with alteration in response to oxidative stress and glycolysis across phenotypes. Hierarchical clustering subgrouped half of precancerous samples under the main branch of the control and the remaining half under carcinoma. Similarly, principal component analysis identified segregated clusters of control, precancerous lesions, and cancer, but erythroplakia phenotypes, in particular, overlapped more with the cancer cluster. Qualitative and quantitative protein signatures across control, precancer, and cancer phenotypes explain possible functional outcomes that dictate malignant transformation to oral carcinoma.
Collapse
|
49
|
Zakaria S, Elsebaey S, Allam S, Abdo W, El-Sisi A. Siah2 inhibitor and the metabolic antagonist Oxamate retard colon cancer progression and downregulate PD1 expression. Recent Pat Anticancer Drug Discov 2023; 19:PRA-EPUB-128869. [PMID: 36650629 DOI: 10.2174/1574892818666230116142606] [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: 05/25/2022] [Revised: 10/31/2022] [Accepted: 11/11/2022] [Indexed: 01/19/2023]
Abstract
BACKGROUND Solid tumors such as colon cancer are characterized by rapid and sustained cell proliferation, which ultimately results in hypoxia, induction of hypoxia-inducible factor-1α (HIF-1α), and activation of glycolysis to promote tumor survival and immune evasion. We hypothesized that a combinatorial approach of menadione (MEN) as an indirect HIF-1α inhibitor and sodium oxamate (OX) as a glycolysis inhibitor may be a promising treatment strategy for colon cancer. OBJECTIVES We investigated the potential efficacy of this combination for promoting an antitumor immune response and suppressing tumor growth in a rat model of colon cancer. METHODS Colon cancer was induced by once-weekly subcutaneous injection of 20 mg/kg dimethylhydrazine (DMH) for 16 weeks. Control rats received the vehicle and then no further treatment (negative control) or MEN plus OX for 4 weeks (drug control). Dimethylhydrazine-treated rats were then randomly allocated to four groups: DMH alone group and other groups treated with MEN, OX, and a combination of (MEN and OX) for 4 weeks. Serum samples were assayed for the tumor marker carbohydrate antigen (CA19.9), while expression levels of HIF-1α, caspase-3, PHD3, LDH, and PD1 were evaluated in colon tissue samples by immunoassay and qRT-PCR. Additionally, Ki-67 and Siah2 expression levels were examined by immunohistochemistry. RESULTS The combination of MEN plus OX demonstrated a greater inhibitory effect on the expression levels of HIF-1α, Siah2, LDH, Ki-67, and PD1, and greater enhancement of caspase-3 and PHD3 expression in colon cancer tissues than either drug alone. CONCLUSION Simultaneous targeting of hypoxia and glycolysis pathways by a combination of MEN and OX could be a promising therapy for inhibiting colon cancer cell growth and promoting antitumor immunity [1].
Collapse
Affiliation(s)
- Sherin Zakaria
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kaferelsheikh University, 33516, Kaferelsheikh, Egypt
| | - Samar Elsebaey
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kaferelsheikh University, 33516, Kaferelsheikh, Egypt
| | - Shady Allam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Menoufia University, 32511, Menoufia, Egypt
| | - Walied Abdo
- Department of Pathology, Faculty of Veterinary Medicine, Kafrelsheikh University, 33516 Kaferelsheikh, Egypt
| | - Alaa El-Sisi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, 31512, Tanta, Egypt
| |
Collapse
|
50
|
Gong R, Xing L, Yin J, Ding Y, Liu X, Bao J, Li J. Appropriate cold stimulation changes energy distribution to improve stress resistance in broilers. J Anim Sci 2023; 101:skad185. [PMID: 37279534 PMCID: PMC10276644 DOI: 10.1093/jas/skad185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 06/01/2023] [Indexed: 06/08/2023] Open
Abstract
Appropriate cold stimulation can improve stress resistance in broilers and alleviate the adverse impacts of a cold environment. To investigate the effects of intermittent mild cold stimulation (IMCS) on energy distribution in the livers of broilers, 96 healthy 1-d-old Ross-308 male broilers were randomly divided into the control group (CC) and the cold stimulation group (H5). The CC group was raised at a normal thermal temperature, i.e., 35 °C until 3 d, after which the temperature was dropped gradually by 0.5 °C/d until 20 °C at 33 d. This temperature was maintained until 49 d. The H5 group was raised at the same temperature as the CC group until 14 d (35 to 29.5 °C) and at 3 °C below the temperature of the CC group starting at 0930 hours for 5 h every other day from 15 to 35 d (26 to 17°C). The temperature was returned to 20 °C at 36 d and maintained until 49 d. At 50 d, all broilers were subjected to acute cold stress (ACS) at 10 °C for 6 and 12 h. We found that IMCS had positive effects on production performance. Using transcriptome sequencing of the broiler livers, 327 differentially expressed genes (DEG) were identified, and highly enriched in fatty acid biosynthesis, fatty acid degradation, and the pyruvate metabolism pathway. When compared to the CC group, the mRNA levels of ACAA1, ACAT2, ACSL1, CPT1A, LDHB, and PCK1 in the H5 group were increased at 22 d (P < 0.05). The LDHB mRNA level was upregulated in the H5 group at 29 d compared to the CC group (P < 0.05). After 21 d of IMCS (at 36 d), the mRNA expression levels of ACAT2 and PCK1 were found to be significantly increased in the H5 group compared to the CC group (P < 0.05). Seven days after the IMCS had ended (at 43 d), the mRNA levels of ACAA1, ACAT2, and LDHB in the H5 group were higher than in the CC group (P < 0.05). The mRNA levels of heat shock protein (HSP) 70, HSP90, and HSP110 in the H5 group were higher than in the CC group after 6 h of ACS (P < 0.05). The protein levels of HSP70 and HSP90 in the H5 group were downregulated after 12 h of ACS, compared to the CC group (P < 0.05). These results indicated that IMCS at 3 °C lower than the normal temperature could improve energy metabolism and stress resistance in the livers of broilers, alleviate the damage of short-term ACS on broilers, help broilers adapt to the low temperature, and maintain stable of energy metabolism in the body.
Collapse
Affiliation(s)
- Rixin Gong
- College of Life Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Lu Xing
- College of Life Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Jingwen Yin
- College of Life Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Yuqing Ding
- College of Life Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Xiaotao Liu
- College of Life Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Jun Bao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Jianhong Li
- College of Life Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| |
Collapse
|