1
|
Li Q, Gan B. Disulfidptosis meets antitumour immunity. Nat Cell Biol 2025; 27:886-887. [PMID: 40473855 DOI: 10.1038/s41556-025-01679-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/19/2025]
Affiliation(s)
- Qidong Li
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Boyi Gan
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.
| |
Collapse
|
2
|
Qu Y, Wang X, Li J, Luo H, Liu H, Wang T, Han X. TAMs-derived SPP1, regulated by HIF-1α/STAT3 signaling pathway, influences colorectal cancer malignant progression by activation of EMT via integrin αvβ3. Int Immunopharmacol 2025; 159:114947. [PMID: 40424660 DOI: 10.1016/j.intimp.2025.114947] [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: 02/24/2025] [Revised: 04/30/2025] [Accepted: 05/20/2025] [Indexed: 05/29/2025]
Abstract
Liver metastasis of colorectal cancer (CRC) is characterized by a high recurrence rate after surgery, which may be related to the rerecruitment of residual tumor cells by other factors that promote cancer cell growth in the tumor microenvironment. Tumor-associated macrophages (TAMs), as key immune components, showed high expression of secretory phosphoprotein-1 (SPP1) at the site of liver metastasis in colorectal cancer patients. However, the factors and mechanisms driving the elevated expression of SPP1 in TAMs remain poorly understood, as do the potential effects of SPP1 on colorectal cancer progression. In this study, we investigated the factors that contributed to the high expression of SPP1 in TAMs and its role in promoting the M2 polarization of TAMs. Additionally, we examined the direct impact of SPP1 derived from TAMs on the malignant phenotype of colorectal cancer. The results showed that the two major characteristics of the tumor microenvironment-hypoxia and acidity-synergistically increased the expression of SPP1 in TAMs through the HIF-1α/STAT3 signaling pathway, Moreover, elevated SPP1 protein promoted the M2-like polarization of TAMs by reducing mitochondrial damage and affecting metabolic reprogramming. In addition, TAMs-derived SPP1 could directly influence the malignant progression of colorectal cancer by interacting with αvβ3 integrin through paracrine on the surface of cancer cells. Inhibiting HIF-1α involved in the regulation of SPP1 and blocking the direct action of SPP1 with cancer cells could effectively inhibit liver metastasis of CRC. These findings suggested that blocking the upstream signaling pathway of SPP1 or inhibiting its downstream target could be a promising therapeutic strategy to prevent or reduce liver metastasis recurrence in CRC.
Collapse
Affiliation(s)
- Yaru Qu
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan 250012, China
| | - Xingchen Wang
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan 250012, China
| | - Junnan Li
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan 250012, China
| | - Huiyuan Luo
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan 250012, China
| | - He Liu
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan 250012, China
| | - Tong Wang
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan 250012, China
| | - Xiuzhen Han
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan 250012, China; Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Science, Shandong University, China.
| |
Collapse
|
3
|
Wang X, Liu J, Mao C, Mao Y. Lactylation-regulated biomolecular condensates: metabolic control of phase separation in physiology and disease. Cell Commun Signal 2025; 23:239. [PMID: 40414883 DOI: 10.1186/s12964-025-02244-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 05/09/2025] [Indexed: 05/27/2025] Open
Abstract
Lactate has long been viewed as a "waste product" of anaerobic glycolysis, with its role in health and disease often overlooked. However, recent discoveries of lactylation-a novel post-translational modification involving lactate-have sparked a renewed understanding of lactate's functions. Lactylation alters the molecular structure of proteins with different cellular localizations, enabling the regulation of their functions and aggregation in specific spatiotemporal contexts, with its impact on biomolecular phase separation being one of its primary effects. However, it remains unknown how lactylation dynamically regulates the spatiotemporal specificity of phase separation and its role in diseases. This article provides an overview of the regulatory mechanisms of biomolecular phase separation driven by lactylation, aiming to offer fresh insights into the role of lactylation in normal and disease-related biological processes while deepening our understanding of its research value and biological significance.
Collapse
Affiliation(s)
- Xi Wang
- Department of Nuclear Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Jiameng Liu
- Department of Nuclear Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Chaoming Mao
- Department of Nuclear Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Yufei Mao
- Department of Ultrasound Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
| |
Collapse
|
4
|
Zhang T, Zhou W, Fan T, Yuan Y, Tang X, Zhang Q, Zou J, Li Y. Lactic acid metabolism: gynecological cancer's Achilles' heel. Discov Oncol 2025; 16:657. [PMID: 40314877 PMCID: PMC12048388 DOI: 10.1007/s12672-025-02364-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 04/10/2025] [Indexed: 05/03/2025] Open
Abstract
Lactic acid is significantly expressed in many cancers, including gynecological cancer, and has become a key regulator of the proliferation, development, metastasis and invasion of these cancers. In clinical and experimental studies, the level of lactic acid in gynecological cancer is closely related to metastasis and invasion, tumor recurrence and poor prognosis. Lactic acid can regulate the internal metabolic pathway of gynecological cancer cells and drive the autonomous role of non-cancer cells in gynecological cancer. In addition to being used as a source of energy metabolism by gynecological cancer cells, lactic acid can also be transported from cancer cells to neighboring cancer cells, stroma and vascular endothelial cells (ECs) to further guide metabolic reprogramming. Lactic acid is also involved in promoting inflammation and angiogenesis in gynecologic tumors. Therefore, we reviewed the mechanisms and recent advances in the production and transport of lactic acid in gynecological cancer. These advances and evidence suggest that targeted lactic acid metabolism is a promising cancer treatment.
Collapse
Affiliation(s)
- Ting Zhang
- The Second Affiliated Hospital, Department of Gynecology, Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Hengyang Medical School, Cancer Research Institute, University of South China, Hengyang, Hunan, China
| | - Wenchao Zhou
- The Second Affiliated Hospital, Department of Gynecology, Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Hengyang Medical School, Cancer Research Institute, University of South China, Hengyang, Hunan, China
| | - Tingyu Fan
- The Second Affiliated Hospital, Department of Gynecology, Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Hengyang Medical School, Cancer Research Institute, University of South China, Hengyang, Hunan, China
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
| | - Yuwei Yuan
- The Second Affiliated Hospital, Department of Gynecology, Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Hengyang Medical School, Cancer Research Institute, University of South China, Hengyang, Hunan, China
| | - Xing Tang
- The Second Affiliated Hospital, Department of Gynecology, Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Hengyang Medical School, Cancer Research Institute, University of South China, Hengyang, Hunan, China
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
| | - Qunfeng Zhang
- The Second Affiliated Hospital, Department of Gynecology, Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Hengyang Medical School, Cancer Research Institute, University of South China, Hengyang, Hunan, China.
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China.
| | - Juan Zou
- The Second Affiliated Hospital, Department of Gynecology, Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Hengyang Medical School, Cancer Research Institute, University of South China, Hengyang, Hunan, China.
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China.
| | - Yukun Li
- The Second Affiliated Hospital, Department of Gynecology, Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Hengyang Medical School, Cancer Research Institute, University of South China, Hengyang, Hunan, China.
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China.
| |
Collapse
|
5
|
Miyazaki I, Tsao KK, Kamijo Y, Nasu Y, Terai T, Campbell RE. Synthesis and application of a photocaged-L-lactate for studying the biological roles of L-lactate. Commun Chem 2025; 8:104. [PMID: 40188278 PMCID: PMC11972357 DOI: 10.1038/s42004-025-01495-1] [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: 09/27/2024] [Accepted: 03/20/2025] [Indexed: 04/07/2025] Open
Abstract
L-Lactate, once considered a metabolic waste product of glycolysis, is now recognized as a vitally important metabolite and signaling molecule in multiple biological pathways. However, exploring L-lactate's emerging intra- and extra-cellular roles is hindered by a lack of tools to perturb L-lactate concentration intracellularly and extracellularly. Photocaged compounds are a powerful way to introduce bioactive molecules with spatiotemporal precision using illumination. Here, we report the development of a photocaged derivative of L-lactate, 4-methoxy-7-nitroindolinyl-L-lactate (MNI-L-lac), that releases L-lactate upon illumination. We validated MNI-L-lac in cell culture by demonstrating that the photorelease of L-lactate elicits a response from genetically encoded extra- and intracellular L-lactate biosensors (eLACCO1, eLACCO2.1, R-iLACCO1.2). To demonstrate the utility of MNI-L-lac, we employed the photorelease of L-lactate to activate G protein-coupled receptor 81 (GPR81), as revealed by the inhibition of adenylyl cyclase activity and concomitant decrease of cAMP. These results indicate that MNI-L-lac may be useful for perturbing the concentration of endogenous L-lactate in order to investigate L-lactate's roles in metabolic and signaling pathways.
Collapse
Affiliation(s)
- Ikumi Miyazaki
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Kelvin K Tsao
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.
- Global Standard Science Education Division, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.
| | - Yuki Kamijo
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yusuke Nasu
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- PRESTO, Japan Science and Technology Agency, Chiyoda-ku, Tokyo, Japan
- Institute of Biological Chemistry, Academia Sinica, Nankang, Taipei, Taiwan
| | - Takuya Terai
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.
| | - Robert E Campbell
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.
- CERVO, Brain Research Center and Department of Biochemistry, Microbiology, and Bioinformatics, Université Laval, Québec, Québec, Canada.
| |
Collapse
|
6
|
Zou Y, Cao M, Tai M, Zhou H, Tao L, Wu S, Yang K, Zhang Y, Ge Y, Wang H, Luo S, Ju Z. A Feedback Loop Driven by H4K12 Lactylation and HDAC3 in Macrophages Regulates Lactate-Induced Collagen Synthesis in Fibroblasts Via the TGF-β Signaling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411408. [PMID: 39945346 PMCID: PMC11967864 DOI: 10.1002/advs.202411408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 01/20/2025] [Indexed: 04/05/2025]
Abstract
The decrease in fibroblast collagen is a primary contributor to skin aging. Lactate can participate in collagen synthesis through lysine lactylation by regulating gene transcription. However, the precise mechanism by which lactate influences collagen synthesis requires further investigation. This study demonstrates that the depletion of macrophages mitigates the stimulating effect of lactate on collagen synthesis in fibroblasts. Through joint CUT&Tag and RNA-sequencing analyses, a feedback loop between H4K12 lactylation (H4K12la) and histone deacetylase 3 (HDAC3) in macrophages that drives lactate-induced collagen synthesis are identified. Macrophages can uptake extracellular lactate via monocarboxylate transporter-1 (MCT1), leading to an up-regulation of H4K12la levels through a KAT5-KAT8-dependent mechanism in response to Poly-L-Lactic Acid (PLLA) stimulation, a source of low concentration and persistent lactate, thereby promoting collagen synthesis in fibroblasts. Furthermore, H4K12la is enriched at the promoters of TGF-β1 and TGF-β3, enhancing their transcription. Hyperlactylation of H4K12la inhibits the expression of the eraser HDAC3, while the activation of HDAC3 reduces H4K12la in macrophages and suppresses collagen synthesis in fibroblasts. In conclusion, this study illustrates that macrophages play a critical role in lactate-induced collagen synthesis in the skin, and targeting the lactate-H4K12la-HDAC3-TGF-β axis may represent a novel approach for enhancing collagen production to combat skin aging.
Collapse
Affiliation(s)
- Ying Zou
- Key Laboratory of Regenerative Medicine of Ministry of EducationInstitute of Aging and Regenerative MedicineCollege of Life Science and TechnologyJinan UniversityGuangzhou510632China
- Department of Plastic and Reconstructive SurgeryGuangdong Second Provincial General HospitalJinan UniversityGuangzhou510403China
| | - Mibu Cao
- Department of Plastic and Reconstructive SurgeryGuangdong Second Provincial General HospitalJinan UniversityGuangzhou510403China
| | - Meiling Tai
- R&D CenterInfinitus (China) Company LtdGuangzhou510640China
| | - Haoxian Zhou
- Department of CardiologyGuangdong Provincial Cardiovascular InstituteGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhou510080China
| | - Li Tao
- Key Laboratory of Regenerative Medicine of Ministry of EducationInstitute of Aging and Regenerative MedicineCollege of Life Science and TechnologyJinan UniversityGuangzhou510632China
| | - Shu Wu
- Key Laboratory of Regenerative Medicine of Ministry of EducationInstitute of Aging and Regenerative MedicineCollege of Life Science and TechnologyJinan UniversityGuangzhou510632China
| | - Kaiye Yang
- R&D CenterInfinitus (China) Company LtdGuangzhou510640China
| | - Youliang Zhang
- Department of Plastic and Reconstructive SurgeryGuangdong Second Provincial General HospitalJinan UniversityGuangzhou510403China
| | - Yuanlong Ge
- Key Laboratory of Regenerative Medicine of Ministry of EducationInstitute of Aging and Regenerative MedicineCollege of Life Science and TechnologyJinan UniversityGuangzhou510632China
| | - Hao Wang
- Department of AnesthesiologyThe First Affiliated HospitalJinan UniversityGuangzhou510632China
| | - Shengkang Luo
- Key Laboratory of Regenerative Medicine of Ministry of EducationInstitute of Aging and Regenerative MedicineCollege of Life Science and TechnologyJinan UniversityGuangzhou510632China
- Department of Plastic and Reconstructive SurgeryGuangdong Second Provincial General HospitalJinan UniversityGuangzhou510403China
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of EducationInstitute of Aging and Regenerative MedicineCollege of Life Science and TechnologyJinan UniversityGuangzhou510632China
- Department of Plastic and Reconstructive SurgeryGuangdong Second Provincial General HospitalJinan UniversityGuangzhou510403China
| |
Collapse
|
7
|
Wu Y, Xie BB, Zhang BL, Zhuang QX, Liu SW, Pan HM. Apatinib regulates the glycolysis of vascular endothelial cells through PI3K/AKT/PFKFB3 pathway in hepatocellular carcinoma. World J Gastroenterol 2025; 31:102848. [PMID: 40124275 PMCID: PMC11924011 DOI: 10.3748/wjg.v31.i11.102848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/17/2025] [Accepted: 02/11/2025] [Indexed: 03/13/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a prevalent and aggressive malignancy in the Chinese population; the severe vascularization by the tumor makes it difficult to cure. The high incidence and poor survival rates of this disease indicate the search for new therapeutic alternatives. Apatinib became a drug of choice because it inhibits tyrosine kinase activity, mainly through an effect on vascular endothelial growth factor receptor-2, thereby preventing tumor angiogenesis. This mechanism of action makes apatinib effective in the treatment of HCC. AIM To investigate the effect of apatinib on the glycolysis of vascular endothelial cells (VECs). METHODS This present study has investigated the effects of HCC cells on VECs, paying particular attention to changes in the glycolytic activity of VECs. The co-culture system established in the present study examined key cellular functions such as extracellular acidification rate and oxygen consumption rate. It also discusses participation of apatinib in the above processes. Core to the findings is the phosphatidylinositol 3-kinase (PI3K)/AKT/6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) signaling pathway, emphasizing the function of phosphorylated AKT and its interaction with PFKFB3, an essential regulator of glycolysis. In the investigation, molecular mechanisms by which such a pathway could influence the above VECs functions of proliferation, migration, and tube formation were underlined through coimmunoprecipitation analysis. Besides, supplementary in vivo experiments on nude mice provided additional biological relevance to the obtained results. RESULTS The glycolytic metabolism in VECs co-cultured with HCC cells is highly active, and the increased glycolysis in these endothelial cells accelerates the malignant transformation of HCC cells. Apatinib has been shown to inhibit this glycolytic activity in the VECs. It also hinders the development, multiplication, and movement of these cells while encouraging their programmed cell death. Moreover, biological analysis revealed that apatinib mainly influences VECs by regulating the PI3K/AKT signaling pathway. Subsequent research indicated that apatinib blocks the PI3K/AKT/PFKEB3 pathway, which in turn reduces glycolysis in these cells. CONCLUSION Apatinib influences the glycolytic pathway in the VECs of HCC a through the PI3K/AKT/PFKFB3 signaling pathway.
Collapse
Affiliation(s)
- Yi Wu
- Division of Cancer Medicine, Sir Run Run Shaw Medical Center, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| | - Bin-Bin Xie
- Division of Cancer Medicine, Sir Run Run Shaw Medical Center, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| | - Bing-Liang Zhang
- Section of Oncology, Ningxia Hui Autonomous Region General Hospital, Ningxia Medical University, Yinchuan 750000, Ningxia Hui Autonomous Region, China
| | - Qing-Xin Zhuang
- Section of Oncology, Ningxia Hui Autonomous Region General Hospital, Ningxia Medical University, Yinchuan 750000, Ningxia Hui Autonomous Region, China
| | - Shi-Wei Liu
- Section of Oncology, Ningxia Hui Autonomous Region General Hospital, Ningxia Medical University, Yinchuan 750000, Ningxia Hui Autonomous Region, China
| | - Hong-Ming Pan
- Division of Cancer Medicine, Sir Run Run Shaw Medical Center, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| |
Collapse
|
8
|
Wan Y, Li G, Cui G, Duan S, Chang S. Reprogramming of Thyroid Cancer Metabolism: from Mechanism to Therapeutic Strategy. Mol Cancer 2025; 24:74. [PMID: 40069775 PMCID: PMC11895238 DOI: 10.1186/s12943-025-02263-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 02/06/2025] [Indexed: 03/15/2025] Open
Abstract
Thyroid cancer as one of the most prevalent malignancies of endocrine system, has raised public concern and more research on its mechanism and treatment. And metabolism-based therapies have advanced rapidly, for the exclusive metabolic profiling of thyroid cancer. In thyroid cancer cells, plenty of metabolic pathways are reprogrammed to accommodate tumor microenvironment. In this review, we initiatively summarize recent progress in the full-scale thyroid cancer metabolic rewiring and the interconnection of various metabolites. We also discuss the efficacy and prospect of metabolic targeted detection as well as therapy. Comprehending metabolic mechanism and characteristics of thyroid cancer roundly will be highly beneficial to managing individual patients.
Collapse
Affiliation(s)
- Yuxuan Wan
- Department of General Surgery, Xiangya Hospital Central South University, Changsha, 410008, Hunan, People's Republic of China
- Xiangya School of Medicine, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Guoqing Li
- Department of General Surgery, Xiangya Hospital Central South University, Changsha, 410008, Hunan, People's Republic of China
- Xiangya School of Medicine, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Gaoyuan Cui
- Xiangya School of Medicine, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Saili Duan
- Department of General Surgery, Xiangya Hospital Central South University, Changsha, 410008, Hunan, People's Republic of China.
- Xiangya School of Medicine, Central South University, Changsha, 410008, Hunan, People's Republic of China.
- Department of Cancer Biology, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Shi Chang
- Department of General Surgery, Xiangya Hospital Central South University, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, 410008, Hunan, People's Republic of China.
- Clinical Research Center for Thyroid Disease in Hunan Province, Changsha, 410008, Hunan, People's Republic of China.
- Hunan Provincial Engineering Research Center for Thyroid and Related Diseases Treatment Technology, Changsha, 410008, Hunan, People's Republic of China.
| |
Collapse
|
9
|
Ye Y, Bin B, Chen P, Chen J, Meng A, Yu L, Yang F, Cui H. Advances in the study of the role of gastric microbiota in the progression of gastric cancer. Microb Pathog 2025; 199:107240. [PMID: 39708981 DOI: 10.1016/j.micpath.2024.107240] [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: 07/10/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
Gastric cancer (GC) is a common malignant tumor and the third most common cancer in China in terms of mortality. Stomach microorganisms play complex roles in the development of GC. The carcinogenic mechanism of Helicobacter pylori has been elucidated, and there is much evidence that other microorganisms in the gastric mucosa are also heavily involved in the disease progression of this cancer. However, their carcinogenic mechanisms have not yet been fully elucidated. The microbial compositions associated with the normal stomach, precancerous lesions, and GC are distinctly different and have a complex evolutionary mechanism. The dysregulation of gastric microbiota may play a key role in the oncogenic process from precancerous lesions to malignant gastric tumors. In this review, we explore the potential translational and clinical implications of intragastric microbes in the diagnosis, prognosis, and treatment of GC. Finally, we summarize the research dilemmas and solutions concerning intragastric microbes, emphasizing that they should be at the forefront of strategies for GC prevention and treatment.
Collapse
Affiliation(s)
- Yu Ye
- Inner Mongolia Medical University, No 60, Xi Lin Guo Le South Road, Hohhot, 010020, Inner Mongolia Autonomous Region, PR China
| | - Ba Bin
- Department of Oncology, Ordos Hospital of Traditional Chinese Medicine, No 5, Yongning Street, Kangbashi District, Ordos City, Inner Mongolia Autonomous Region, PR China
| | - Pengfei Chen
- The Affiliated Hospital of Inner Mongolia Medical University, PR China
| | - Jing Chen
- Medical Department of Ordos College of Applied Technology, PR China
| | - Aruna Meng
- Inner Mongolia Medical University, No 60, Xi Lin Guo Le South Road, Hohhot, 010020, Inner Mongolia Autonomous Region, PR China
| | - Lei Yu
- Department of Pharmacy, Traditional Chinese Medicine Hospital of Inner Mongolia Autonomous Region, Hohhot, Inner Mongolia Autonomous Region, 010020, PR China
| | - Fan Yang
- Inner Mongolia Autonomous Region Blood Central, PR China.
| | - Hongwei Cui
- Peking University Cancer Hospital (Inner Mongolia Campus) & Affiliated Cancer Hospital of Inner Mongolia Medical University, No 42, Zhao Wu Da Road, Hohhot, 010020, Inner Mongolia Autonomous Region, PR China.
| |
Collapse
|
10
|
Chen Z, Yu T, Wang Y, Li J, Zhang B, Zhou L. Mechanistic insights into the role of traditional Chinese medicine in treating gastric cancer. Front Oncol 2025; 14:1443686. [PMID: 39906672 PMCID: PMC11790455 DOI: 10.3389/fonc.2024.1443686] [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: 06/04/2024] [Accepted: 12/30/2024] [Indexed: 02/06/2025] Open
Abstract
Gastric cancer remains a leading cause of cancer-related mortality worldwide, with advanced stages presenting significant challenges due to metastasis and drug resistance. Traditional Chinese Medicine (TCM) offers a promising complementary approach characterized by holistic treatment principles and minimal side effects. This review comprehensively explores the multifaceted mechanisms by which TCM addresses gastric cancer. Specifically, we detail how TCM inhibits aerobic glycolysis by downregulating key glycolytic enzymes and metabolic pathways, thereby reducing the energy supply essential for cancer cell proliferation. We examine how TCM suppresses angiogenesis by targeting the vascular endothelial growth factor (VEGF) and cyclooxygenase-2 (COX-2) pathways, effectively starving tumors of nutrients and oxygen required for growth and metastasis. Furthermore, TCM modulates the immune microenvironment by enhancing the activity of effector immune cells such as CD4+ and CD8+ T cells and natural killer (NK) cells while reducing immunosuppressive cells like regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs). These actions collectively contribute to slowing tumor progression, inhibiting metastasis, and enhancing the body's antitumor response. The insights presented underscore the significant potential of TCM as an integral component of comprehensive gastric cancer treatment strategies, highlighting avenues for future research and clinical application to improve patient outcomes.
Collapse
Affiliation(s)
- Ziqiang Chen
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Ting Yu
- Department of Rheumatism, Third Affiliated Clinical Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Yunhe Wang
- Department of Endocrinology, Metabolism and Gastroenterology, Third Affiliated Clinical Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Jiaxin Li
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Bo Zhang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Liya Zhou
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| |
Collapse
|
11
|
Zhang D, Yuan Y, Zeng Q, Xiong J, Gan Y, Jiang K, Xie N. Plant protein-derived anti-breast cancer peptides: sources, therapeutic approaches, mechanisms, and nanoparticle design. Front Pharmacol 2025; 15:1468977. [PMID: 39898323 PMCID: PMC11783187 DOI: 10.3389/fphar.2024.1468977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 11/19/2024] [Indexed: 02/04/2025] Open
Abstract
Breast cancer causes the deaths of approximately 685,000 women annually, posing a severe threat to women's health. Consequently, there is an urgent need for low-cost, low-toxicity and effective therapeutic methods to prevent or mitigate breast cancer progression. PDBP are natural, non-toxic, and affordable substances and have demonstrated excellent anti-breast cancer activities in inhibiting proliferation, migration, and invasion, and promoting apoptosis both in vitro and in vivo, thus effectively preventing or inhibiting breast cancer. However, there are no comprehensive reviews summarizing the effects and mechanisms of PDBP on the treatment of breast cancer. Therefore, this review described the inhibitory effects and mechanisms of active peptides from different plant protein sources on breast cancer. Additionally, we summarized the advantages and preparation methods of plant protein-derived anticancer peptide-encapsulated nanoparticles and their effects in inhibiting breast cancer. This review provides a scientific basis for understanding the anti-breast cancer mechanisms of PDBP and offers guidance for the development of therapeutic adjuvants enriched with these peptides.
Collapse
Affiliation(s)
- Deju Zhang
- Biobank, Shenzhen Second People’s Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Ying Yuan
- Biobank, Shenzhen Second People’s Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
| | - Qingdong Zeng
- Biobank, Shenzhen Second People’s Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China
- Hengyang Medical School, University of South China, Hengyang, China
| | - Juan Xiong
- Biobank, Shenzhen Second People’s Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China
- Hengyang Medical School, University of South China, Hengyang, China
| | - Yiming Gan
- Plant Science, School of Biological Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Kai Jiang
- Eastern Institute for Advanced Study, Eastern Institute of Technology, Ningbo, China
- Department of Thermal Science and Energy Engineering, University of Science and Technology of China, Hefei, China
| | - Ni Xie
- Biobank, Shenzhen Second People’s Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|
12
|
Wang X, Qu Y, Ji J, Liu H, Luo H, Li J, Han X. Colorectal cancer cells establish metabolic reprogramming with cancer-associated fibroblasts (CAFs) through lactate shuttle to enhance invasion, migration, and angiogenesis. Int Immunopharmacol 2024; 143:113470. [PMID: 39471692 DOI: 10.1016/j.intimp.2024.113470] [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: 08/01/2024] [Revised: 10/09/2024] [Accepted: 10/20/2024] [Indexed: 11/01/2024]
Abstract
Fibroblasts undergo metabolic reprogramming after contact with cancer cells in tumor microenvironment, producing lactate to provide a metabolic substrate for neighboring tumor cells. The exchange of lactate between cancer cells and fibroblasts via monocarboxylate transporters (MCTs) is known as the lactate shuttle. Colorectal cancer cells may establish a metabolic coupling akin to the lactate shuttle in collaboration with cancer-associated fibroblasts (CAFs) to augment their invasive and migratory capabilities. However, the specific phenomena and underlying mechanisms are not clear. In this study, we investigated the phenomena and explored the correlation and possible mechanism between CAFs and the invasion and migration of colorectal cancer cells by using two different co-culture models. The results showed that colorectal cancer cells established a lactate metabolic coupling with fibroblasts through the oxidative stress effect, triggering the metabolic reprogramming process of themselves and those of fibroblasts. In addition, lactate enhanced the invasion and migration of colorectal cancer by stabilizing the protein expression levels of nuclear factor kappa-B (NF-κB) and hypoxia-inducible factor-1α (HIF-1α). Blocking oxidative stress and lactate metabolic coupling with reactive oxygen species removers and MCT1-specific inhibitors, respectively, could effectively suppress metastasis in colorectal cancer. These findings suggest that targeting the lactate metabolic coupling between tumor cells and CAFs will offer a new strategy to combat colorectal cancer.
Collapse
Affiliation(s)
- Xingchen Wang
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan 250012, China
| | - Yaru Qu
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan 250012, China
| | - Jianbo Ji
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan 250012, China
| | - He Liu
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan 250012, China
| | - Huiyuan Luo
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan 250012, China
| | - Junnan Li
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan 250012, China
| | - Xiuzhen Han
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan 250012, China; Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Science, Shandong University; Shandong Cancer Hospital and Institute, 440 Jiyan Road, Jinan 250117, Shandong Province, China.
| |
Collapse
|
13
|
Zhang F, Gu T, Li J, Zhu Y, Chu M, Zhou Q, Liu J. Emodin regulated lactate metabolism by inhibiting MCT1 to delay non-small cell lung cancer progression. Hum Cell 2024; 38:11. [PMID: 39465441 DOI: 10.1007/s13577-024-01140-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/09/2024] [Indexed: 10/29/2024]
Abstract
Lung cancer is one of the most common malignant tumors in the world, with high incidence rate and mortality. Monocarboxylate transporter (MCT) 1 has been found to be widely expressed in various tumors and plays a crucial role in regulating energy metabolism. Emodin, as an important traditional Chinese medicine in China, has been reported to inhibit the progression of lung cancer. However, its potential mechanism has not been fully elucidated. The effects of emodin and MCT1 inhibitor AZD3965 on the proliferation, migration, and invasion of lung cancer cells were detected using cell counting kit-8 (CCK-8) assay, wound-healing assay, and transwell small chamber assay. The content of glucose, lactate, and pyruvate in the cell culture medium was detected using a glucose, lactate, and pyruvate detection kit, and also detected protein expression using western blotting. In addition, to investigate the effects of emodin and AZD3965 on lung cancer in vivo, we constructed nude mice subcutaneous transplant tumor model by subcutaneous injection of lung cancer cells. The results showed that emodin and AZD3965 could inhibit the proliferation, migration, and invasion of lung cancer cells. At the same time, they could inhibit the expression of MCT1 in lung cancer cells and promote the release of lactate, but did not affect the content of glucose and pyruvate. In vivo experiments had shown that emodin and AZD3965 could effectively inhibit the growth of lung cancer and inhibit the expression of MCT1. All in all, our data suggested that emodin inhibited the proliferation, migration, and invasion of lung cancer cells, possibly by inhibiting MCT1, providing important theoretical basis for elucidating the mechanism of emodin in treating lung cancer.
Collapse
Affiliation(s)
- Fei Zhang
- First Affiliated Hospital of Guizhou, University of Traditional Chinese Medicine, Guiyang, 550001, China
| | - Tian Gu
- First Affiliated Hospital of Guizhou, University of Traditional Chinese Medicine, Guiyang, 550001, China
| | - Jin Li
- First Affiliated Hospital of Guizhou, University of Traditional Chinese Medicine, Guiyang, 550001, China
| | - Yanqiu Zhu
- First Affiliated Hospital of Guizhou, University of Traditional Chinese Medicine, Guiyang, 550001, China
| | - Mingliang Chu
- First Affiliated Hospital of Guizhou, University of Traditional Chinese Medicine, Guiyang, 550001, China.
| | - Qing Zhou
- First Affiliated Hospital of Guizhou, University of Traditional Chinese Medicine, Guiyang, 550001, China.
| | - Jiemin Liu
- Department of Endoscopy, Guizhou Provincial People's Hospital, Guiyang, 550002, China.
| |
Collapse
|
14
|
Song Y, Wang Z, Xu L, Han B, Sun D. Identification of Genetic Associations of IDH2, LDHA, and LDHB Genes with Milk Yield and Compositions in Dairy Cows. Life (Basel) 2024; 14:1228. [PMID: 39459528 PMCID: PMC11508787 DOI: 10.3390/life14101228] [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: 09/02/2024] [Revised: 09/23/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024] Open
Abstract
Previous study revealed that isocitrate dehydrogenase (NADP (+)) 2, mitochondrial (IDH2), lactate dehydrogenase A (LDHA), and lactate dehydrogenase B (LDHB) genes were significantly differentially expressed in liver tissues of Holstein cows among different lactation periods and associated with lipid and protein metabolism; hence, they were considered as candidates for milk production traits. Herein, the genetic effects of the three genes on milk yield, fat, and protein traits were studied by association analysis using 926 Chinese Holstein cows from 45 sire families. As a result, five single nucleotide polymorphisms (SNPs) in IDH2, one in LDHA, and three in LDHB were identified by re-sequencing, and subsequently, they were genotyped in 926 Chinese Holstein cows by genotyping by target sequencing (GBTS). With the animal model, single-locus association analysis revealed that four SNPs in IDH2 and one SNP in LDHA were significantly associated with milk, fat, and protein yields (p ≤ 0.0491), and three SNPs in LDHB were associated with milk yield, milk fat yield, and fat percentage (p ≤ 0.0285). Further, four IDH2 SNPs were found to form a haplotype block significantly associated with milk yield, fat yield, protein yield, and protein percentage (p ≤ 0.0249). In addition, functional predictions indicated that one SNP in LDHA, g.26304153G>A, may affect transcription factor binding and two SNPs, g.88544541A>G and g.88556310T>C could alter LDHB mRNA secondary structure. In summary, this study profiled the significant genetic effects of IDH2, LDHA, and LDHB on milk yield and composition traits and provided referable genetic markers for genomic selection programs in dairy cattle.
Collapse
Affiliation(s)
| | | | | | | | - Dongxiao Sun
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, National Engineering Laboratory of Animal Breeding, State Key Laboratory of Animal Biotech Breeding, Department of Animal Genetics and Breeding, College of Animal Science and Technology, Agricultural University, Beijing 100193, China; (Y.S.); (Z.W.); (L.X.); (B.H.)
| |
Collapse
|
15
|
Liu J, Zhao F, Qu Y. Lactylation: A Novel Post-Translational Modification with Clinical Implications in CNS Diseases. Biomolecules 2024; 14:1175. [PMID: 39334941 PMCID: PMC11430557 DOI: 10.3390/biom14091175] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/06/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Lactate, an important metabolic product, provides energy to neural cells during energy depletion or high demand and acts as a signaling molecule in the central nervous system. Recent studies revealed that lactate-mediated protein lactylation regulates gene transcription and influences cell fate, metabolic processes, inflammation, and immune responses. This review comprehensively examines the regulatory roles and mechanisms of lactylation in neurodevelopment, neuropsychiatric disorders, brain tumors, and cerebrovascular diseases. This analysis indicates that lactylation has multifaceted effects on central nervous system function and pathology, particularly in hypoxia-induced brain damage. Highlighting its potential as a novel therapeutic target, lactylation may play a significant role in treating neurological diseases. By summarizing current findings, this review aims to provide insights and guide future research and clinical strategies for central nervous system disorders.
Collapse
Affiliation(s)
- Junyan Liu
- Department of Pediatrics/Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education)/NHC Key Laboratory of Chronobiology, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Neonatal Intensive Care Unit, Binzhou Medical University Hospital, Binzhou 256600, China
| | - Fengyan Zhao
- Department of Pediatrics/Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education)/NHC Key Laboratory of Chronobiology, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Yi Qu
- Department of Pediatrics/Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education)/NHC Key Laboratory of Chronobiology, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
16
|
Shi M, Li Z, Tang Z, Zhou H, Huang X, Wei Y, Li X, Li X, Shi H, Qin D. Exploring the pathogenesis and treatment of PSD from the perspective of gut microbiota. Brain Res Bull 2024; 215:111022. [PMID: 38936669 DOI: 10.1016/j.brainresbull.2024.111022] [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: 04/07/2024] [Revised: 05/30/2024] [Accepted: 06/24/2024] [Indexed: 06/29/2024]
Abstract
Post-stroke depression (PSD) is a psychological disease that can occur following a stroke and is associated with serious consequences. Research on the pathogenesis and treatment of PSD is still in the infancy stage. Patients with PSD often exhibit gastrointestinal symptoms; therefore the role of gut microbiota in the pathophysiology and potential treatment effects of PSD has become a hot topic of research. In this review, describe the research on the pathogenesis and therapy of PSD. We also describe how the gut microbiota influences neurotransmitters, the endocrine system, energy metabolism, and the immune system. It was proposed that the gut microbiota is involved in the pathogenesis and treatment of PSD through the regulation of neurotransmitter levels, vagal signaling, hypothalamic-pituitary-adrenal axis activation and inhibition, hormone secretion and release, in addition to immunity and inflammation.
Collapse
Affiliation(s)
- Mingqin Shi
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming Yunnan, China.
| | - Zhenmin Li
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming Yunnan, China.
| | - Zhengxiu Tang
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming Yunnan, China.
| | - Haimei Zhou
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming Yunnan, China.
| | - Xiaoyi Huang
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming Yunnan, China.
| | - Yuanyuan Wei
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming Yunnan, China.
| | - Xinyao Li
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming Yunnan, China.
| | - Xiahuang Li
- The People's Hospital of Mengzi, The Affiliated Hospital of Yunnan University of Chinese Medicine, Mengzi Honghe, China.
| | - Hongling Shi
- Department of Rehabilitation Medicine, The Third People's Hospital of Yunnan Province, Kunming Yunnan, China.
| | - Dongdong Qin
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming Yunnan, China.
| |
Collapse
|
17
|
Meynen J, Adriaensens P, Criel M, Louis E, Vanhove K, Thomeer M, Mesotten L, Derveaux E. Plasma Metabolite Profiling in the Search for Early-Stage Biomarkers for Lung Cancer: Some Important Breakthroughs. Int J Mol Sci 2024; 25:4690. [PMID: 38731909 PMCID: PMC11083579 DOI: 10.3390/ijms25094690] [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: 03/21/2024] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Lung cancer is the leading cause of cancer-related mortality worldwide. In order to improve its overall survival, early diagnosis is required. Since current screening methods still face some pitfalls, such as high false positive rates for low-dose computed tomography, researchers are still looking for early biomarkers to complement existing screening techniques in order to provide a safe, faster, and more accurate diagnosis. Biomarkers are biological molecules found in body fluids, such as plasma, that can be used to diagnose a condition or disease. Metabolomics has already been shown to be a powerful tool in the search for cancer biomarkers since cancer cells are characterized by impaired metabolism, resulting in an adapted plasma metabolite profile. The metabolite profile can be determined using nuclear magnetic resonance, or NMR. Although metabolomics and NMR metabolite profiling of blood plasma are still under investigation, there is already evidence for its potential for early-stage lung cancer diagnosis, therapy response, and follow-up monitoring. This review highlights some key breakthroughs in this research field, where the most significant biomarkers will be discussed in relation to their metabolic pathways and in light of the altered cancer metabolism.
Collapse
Affiliation(s)
- Jill Meynen
- Faculty of Medicine and Life Sciences, Hasselt University, Martelarenlaan 42, B-3500 Hasselt, Belgium; (J.M.); (M.C.); (K.V.); (L.M.)
| | - Peter Adriaensens
- Applied and Analytical Chemistry, NMR Group, Institute for Materials Research (Imo-Imomec), Hasselt University, Agoralaan 1, B-3590 Diepenbeek, Belgium;
| | - Maarten Criel
- Faculty of Medicine and Life Sciences, Hasselt University, Martelarenlaan 42, B-3500 Hasselt, Belgium; (J.M.); (M.C.); (K.V.); (L.M.)
- Department of Respiratory Medicine, Ziekenhuis Oost-Limburg, Synaps Park 1, B-3600 Genk, Belgium;
| | - Evelyne Louis
- Department of Respiratory Medicine, University Hospital Leuven, Herestraat 49, B-3000 Leuven, Belgium;
| | - Karolien Vanhove
- Faculty of Medicine and Life Sciences, Hasselt University, Martelarenlaan 42, B-3500 Hasselt, Belgium; (J.M.); (M.C.); (K.V.); (L.M.)
- Department of Respiratory Medicine, University Hospital Leuven, Herestraat 49, B-3000 Leuven, Belgium;
- Department of Respiratory Medicine, Algemeen Ziekenhuis Vesalius, Hazelereik 51, B-3700 Tongeren, Belgium
| | - Michiel Thomeer
- Department of Respiratory Medicine, Ziekenhuis Oost-Limburg, Synaps Park 1, B-3600 Genk, Belgium;
| | - Liesbet Mesotten
- Faculty of Medicine and Life Sciences, Hasselt University, Martelarenlaan 42, B-3500 Hasselt, Belgium; (J.M.); (M.C.); (K.V.); (L.M.)
- Department of Nuclear Medicine, Ziekenhuis Oost-Limburg, Synaps Park 1, B-3600 Genk, Belgium
| | - Elien Derveaux
- Applied and Analytical Chemistry, NMR Group, Institute for Materials Research (Imo-Imomec), Hasselt University, Agoralaan 1, B-3590 Diepenbeek, Belgium;
| |
Collapse
|
18
|
Wang P, Xie D, Xiao T, Cheng C, Wang D, Sun J, Wu M, Yang Y, Zhang A, Liu Q. H3K18 lactylation promotes the progression of arsenite-related idiopathic pulmonary fibrosis via YTHDF1/m6A/NREP. JOURNAL OF HAZARDOUS MATERIALS 2024; 461:132582. [PMID: 37742376 DOI: 10.1016/j.jhazmat.2023.132582] [Citation(s) in RCA: 52] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/26/2023] [Accepted: 09/17/2023] [Indexed: 09/26/2023]
Abstract
As epigenetic modifications, lactylation and N6-methyladenosine (m6A) have attracted wide attention. Arsenite is an environmental pollutant that has been proven to induce idiopathic pulmonary fibrosis (IPF). However, the molecular mechanisms of lactylation and m6A methylation are unclear in arsenite-related IPF (As-IPF). In view of the limited understanding of molecular mechanism of m6A and lactylation in As-IPF, MeRIP-seq, RNA-seq and ChIP-seq were analyzed to verify the target gene regulated by m6A and H3K18 lactylation (H3K18la). We found that, for As-IPF, the global levels of m6A, levels of YTHDF1 and m6A-modified neuronal protein 3.1 (NREP) were elevated in alveolar epithelial cells (AECs). The secretion levels of TGF-β1 were increased via YTHDF1/m6A/NREP, which promoted the fibroblast-to-myofibroblast transition (FMT). Further, extracellular lactate from myofibroblasts elevated levels of the global lactylation (Kla) and H3K18la via the lactate monocarboxylate transporter 1 (MCT1), and, in AECs, H3K18la facilitated the transcription of Ythdf1. This report highlights the role of crosstalk between AECs and myofibroblasts via lactylation and m6A and the significance of H3K18la regulation of YTHDF1 in the progression of As-IPF, which may be useful for finding effective therapeutic targets.
Collapse
Affiliation(s)
- Peiwen Wang
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Nanjing Medical University, Nanjing 211166, Jiangsu, PR China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, PR China
| | - Daxiao Xie
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Nanjing Medical University, Nanjing 211166, Jiangsu, PR China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, PR China
| | - Tian Xiao
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Nanjing Medical University, Nanjing 211166, Jiangsu, PR China; Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, Jiangsu, PR China
| | - Cheng Cheng
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Nanjing Medical University, Nanjing 211166, Jiangsu, PR China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, PR China
| | - Dapeng Wang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 550025, Guizhou, PR China
| | - Jing Sun
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Nanjing Medical University, Nanjing 211166, Jiangsu, PR China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, PR China
| | - Meng Wu
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Nanjing Medical University, Nanjing 211166, Jiangsu, PR China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, PR China
| | - Yi Yang
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Nanjing Medical University, Nanjing 211166, Jiangsu, PR China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, PR China
| | - Aihua Zhang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 550025, Guizhou, PR China
| | - Qizhan Liu
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Nanjing Medical University, Nanjing 211166, Jiangsu, PR China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, PR China.
| |
Collapse
|
19
|
Xu J, Bian L, You D, Li Z, Wang T, Li Y, Ren X, He Y. PDGF-BB accelerates TSCC via fibroblast lactates limiting miR-26a-5p and boosting mitophagy. Cancer Cell Int 2024; 24:5. [PMID: 38169376 PMCID: PMC10763357 DOI: 10.1186/s12935-023-03172-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 12/03/2023] [Indexed: 01/05/2024] Open
Abstract
The tumor microenvironment and cancer-associated fibroblasts (CAFs) play crucial roles in tumor development, and their metabolic coupling remains unclear. Clinical data showed a positive correlation between PDGF-BB, CAFs, and glycolysis in the tumor microenvironment of oral tongue squamous cell carcinoma patients. In vitro, CAFs are derived from hOMF cells treated with PDGF-BB, which induces their formation and promotes aerobic glycolysis. Mitophagy increased the PDGF-BB-induced formation of CAF phenotypes and aerobic glycolysis, while autophagy inhibition blocked PDGF-BB-induced effects. Downregulation of miR-26a-5p was observed in CAFs; upregulation of miR-26a-5p inhibited the expression of mitophagy-related proteins ULKI, Parkin, PINK1, and LC3 and aerobic glycolysis in PDGF-BB-induced CAFs. PDGF-BB-induced CAFs promoted tumor cell proliferation, invasion, metastasis, NF-κB signaling pathway activation, and PDGF-BB secretion. Thus, PDGF-BB is associated with lactate-induced CAF formation and glucose metabolism reprogramming. These findings indicate potential therapeutic targets in oral tongue squamous cell carcinoma.
Collapse
Affiliation(s)
- Jianguo Xu
- Department of Oral and Maxillofacial Surgery, Kunming Medical University School and Hospital of Stomatology, Kunming, 650106, China
- Yunnan Key Laboratory of Stomatology, Kunming, 650106, China
| | - Li Bian
- Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650106, China
| | - Dingyun You
- School of Public Health, Kunming Medical University, Kunming, 650500, China
| | - Ziliang Li
- Department of oral Implantology, Kunming Medical University School and Hospital of Stomatology, Kunming, 650106, China
| | - Tingting Wang
- Department of Stomatology, The First People's Hospital of Yunnan Province, Kunming, 650032, China
| | - Yiting Li
- Department of Oral and Maxillofacial Surgery, Kunming Medical University School and Hospital of Stomatology, Kunming, 650106, China
| | - Xiaobin Ren
- Department of Periodontology, Kunming Medical University School and Hospital of Stomatology, 1088 Haiyuan Central Road, Kunming, Yunnan, 650106, China.
| | - Yongwen He
- Department of Dental Research, Kunming Medical University School and Hospital of Stomatology, 1088 Haiyuan Central Road, Kunming, Yunnan, 650106, China.
- Qujing Medical College, Qujing, 655011, China.
| |
Collapse
|
20
|
Guo B, Shu H, Luo L, Liu X, Ma Y, Zhang J, Liu Z, Zhang Y, Fu L, Song T, Qiao Y, Zhang C. Lactate Conversion by Lactate Dehydrogenase B Is Involved in Beige Adipocyte Differentiation and Thermogenesis in Mice. Nutrients 2023; 15:4846. [PMID: 38004240 PMCID: PMC10674895 DOI: 10.3390/nu15224846] [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: 10/09/2023] [Revised: 11/06/2023] [Accepted: 11/16/2023] [Indexed: 11/26/2023] Open
Abstract
Adipose tissue (AT) is the primary reservoir of lipid, the major thermogenesis organ during cold exposure, and an important site for lactate production. However, the utilization of lactate as a metabolic substrate by adipocytes, as well as its potential involvement in the regulation of adipocyte thermogenesis, remain unappreciated. In vitro experiments using primary stromal vascular fraction preadipocytes isolated from mouse inguinal white adipose tissue (iWAT) revealed that lactate dehydrogenase B (LDHB), the key glycolytic enzyme that catalyzes the conversion of lactate to pyruvate, is upregulated during adipocyte differentiation, downregulated upon chronic cold stimulation, and regained after prolonged cold exposure. In addition, the global knockout of Ldhb significantly reduced the masses of iWAT and epididymal WAT (eWAT) and impeded the utilization of iWAT during cold exposure. In addition, Ldhb loss of function impaired the mitochondrial function of iWAT under cold conditions. Together, these findings uncover the involvement of LDHB in adipocyte differentiation and thermogenesis.
Collapse
Affiliation(s)
- Bin Guo
- The Tenth Affiliated Hospital of Southern Medical University (Dongguan People’s Hospital), Dongguan 523018, China;
| | - Hui Shu
- Cambridge-Suda Genomic Resource Center, Soochow University, Suzhou 215123, China; (H.S.); (L.L.); (X.L.); (Y.M.); (J.Z.); (Z.L.); (Y.Z.)
| | - Ling Luo
- Cambridge-Suda Genomic Resource Center, Soochow University, Suzhou 215123, China; (H.S.); (L.L.); (X.L.); (Y.M.); (J.Z.); (Z.L.); (Y.Z.)
| | - Xiangpeng Liu
- Cambridge-Suda Genomic Resource Center, Soochow University, Suzhou 215123, China; (H.S.); (L.L.); (X.L.); (Y.M.); (J.Z.); (Z.L.); (Y.Z.)
| | - Yue Ma
- Cambridge-Suda Genomic Resource Center, Soochow University, Suzhou 215123, China; (H.S.); (L.L.); (X.L.); (Y.M.); (J.Z.); (Z.L.); (Y.Z.)
| | - Jie Zhang
- Cambridge-Suda Genomic Resource Center, Soochow University, Suzhou 215123, China; (H.S.); (L.L.); (X.L.); (Y.M.); (J.Z.); (Z.L.); (Y.Z.)
| | - Zhiwei Liu
- Cambridge-Suda Genomic Resource Center, Soochow University, Suzhou 215123, China; (H.S.); (L.L.); (X.L.); (Y.M.); (J.Z.); (Z.L.); (Y.Z.)
| | - Yong Zhang
- Cambridge-Suda Genomic Resource Center, Soochow University, Suzhou 215123, China; (H.S.); (L.L.); (X.L.); (Y.M.); (J.Z.); (Z.L.); (Y.Z.)
| | - Lei Fu
- Wisdom Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China;
| | - Tongxing Song
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China;
| | - Yixue Qiao
- Wisdom Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China;
| | - Chi Zhang
- Cambridge-Suda Genomic Resource Center, Soochow University, Suzhou 215123, China; (H.S.); (L.L.); (X.L.); (Y.M.); (J.Z.); (Z.L.); (Y.Z.)
| |
Collapse
|
21
|
Nasu Y, Aggarwal A, Le GNT, Vo CT, Kambe Y, Wang X, Beinlich FRM, Lee AB, Ram TR, Wang F, Gorzo KA, Kamijo Y, Boisvert M, Nishinami S, Kawamura G, Ozawa T, Toda H, Gordon GR, Ge S, Hirase H, Nedergaard M, Paquet ME, Drobizhev M, Podgorski K, Campbell RE. Lactate biosensors for spectrally and spatially multiplexed fluorescence imaging. Nat Commun 2023; 14:6598. [PMID: 37891202 PMCID: PMC10611801 DOI: 10.1038/s41467-023-42230-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023] Open
Abstract
L-Lactate is increasingly appreciated as a key metabolite and signaling molecule in mammals. However, investigations of the inter- and intra-cellular dynamics of L-lactate are currently hampered by the limited selection and performance of L-lactate-specific genetically encoded biosensors. Here we now report a spectrally and functionally orthogonal pair of high-performance genetically encoded biosensors: a green fluorescent extracellular L-lactate biosensor, designated eLACCO2.1, and a red fluorescent intracellular L-lactate biosensor, designated R-iLACCO1. eLACCO2.1 exhibits excellent membrane localization and robust fluorescence response. To the best of our knowledge, R-iLACCO1 and its affinity variants exhibit larger fluorescence responses than any previously reported intracellular L-lactate biosensor. We demonstrate spectrally and spatially multiplexed imaging of L-lactate dynamics by coexpression of eLACCO2.1 and R-iLACCO1 in cultured cells, and in vivo imaging of extracellular and intracellular L-lactate dynamics in mice.
Collapse
Affiliation(s)
- Yusuke Nasu
- Department of Chemistry, School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan.
- PRESTO, Japan Science and Technology Agency, Chiyoda-ku, Tokyo, 102-0075, Japan.
| | - Abhi Aggarwal
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, 20147, USA
- Allen Institute for Neural Dynamics, Seattle, WA, 98109, USA
| | - Giang N T Le
- Department of Chemistry, School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
- Department of Chemistry, University of Toronto, Toronto, ON, M5S 3H6, Canada
| | - Camilla Trang Vo
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Yuki Kambe
- Department of Pharmacology, Graduate School of Medical and Dental Science, Kagoshima University, Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Xinxing Wang
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Felix R M Beinlich
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Ashley Bomin Lee
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Tina R Ram
- Hotchkiss Brain Institute, Cumming School of Medicine, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Fangying Wang
- Hotchkiss Brain Institute, Cumming School of Medicine, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Kelsea A Gorzo
- Hotchkiss Brain Institute, Cumming School of Medicine, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Yuki Kamijo
- Department of Chemistry, School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Marc Boisvert
- CERVO Brain Research Centre, Québec, QC, G1J 2G3, Canada
- Department of Biochemistry, Microbiology and Bioinformatics, Laval University, Québec, QC, G1E 1T2, Canada
| | - Suguru Nishinami
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Genki Kawamura
- Department of Chemistry, School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Takeaki Ozawa
- Department of Chemistry, School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Hirofumi Toda
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Grant R Gordon
- Hotchkiss Brain Institute, Cumming School of Medicine, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Shaoyu Ge
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Hajime Hirase
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Marie-Eve Paquet
- CERVO Brain Research Centre, Québec, QC, G1J 2G3, Canada
- Department of Biochemistry, Microbiology and Bioinformatics, Laval University, Québec, QC, G1E 1T2, Canada
| | - Mikhail Drobizhev
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, 59717, USA
| | - Kaspar Podgorski
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, 20147, USA
- Allen Institute for Neural Dynamics, Seattle, WA, 98109, USA
| | - Robert E Campbell
- Department of Chemistry, School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan.
- CERVO Brain Research Centre, Québec, QC, G1J 2G3, Canada.
- Department of Biochemistry, Microbiology and Bioinformatics, Laval University, Québec, QC, G1E 1T2, Canada.
| |
Collapse
|
22
|
Liu X, Zou X, Zhou Y, Chen R, Peng Y, Qu M. LDHA and LDHB overexpression promoted the Warburg effect in malignantly transformed GES-1 cells induced by N-nitroso compounds. Food Chem Toxicol 2023; 180:114007. [PMID: 37648104 DOI: 10.1016/j.fct.2023.114007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/26/2023] [Accepted: 08/27/2023] [Indexed: 09/01/2023]
Abstract
N-nitroso compounds (NOCs) exposure is a major risk factor for the development of gastric cancer. However, the carcinogenic mechanisms by which NOCs induce gastric and other cancers, especially the NOCs-induced Warburg effect, have not been comprehensively studied. Lactate dehydrogenase (LDH), which has two subunits (LDHA and LDHB), plays an important role in the Warburg effect of tumor cells. Therefore, we hypothesized that LDHA and LDHB could promote Warburg effect in malignant transformed GES-1 cells induced by Nmethyl-N'-nitro-N-nitrosoguanidine (MNNG). GES-1 cells were exposed to 1 μmol/L MNNG and cultured for 40 passages. During the culturing process, cell proliferation, migration, and soft agar colony formation significantly increased after 30 passages. Following MNNG exposure, lactate, LDH, glucose uptake, and the expression levels of key enzymes in glycolysis were significantly increased. Knocking down LDHA or LDHB alone reduced lactate secretion, inhibited cell viability, and impaired migratory capacities. Knocking down LDHA and LDHB together fully suppressed lactate secretion and effectively suppressed the malignant phenotype of cells transformed by long-term MNNG exposure. Finally, we demonstrated that overexpression of LDHA and LDHB promotes the malignant transformation of GES-1 cells by enhancing the Warburg effect during long-term exposure to NOCs.
Collapse
Affiliation(s)
- Xing Liu
- School of Public Health, Yangzhou University, Yangzhou, 225009, China.
| | - Xihuan Zou
- School of Public Health, Yangzhou University, Yangzhou, 225009, China.
| | - Yueyue Zhou
- School of Public Health, Yangzhou University, Yangzhou, 225009, China.
| | - Ruobing Chen
- School of Public Health, Yangzhou University, Yangzhou, 225009, China.
| | - Yuting Peng
- School of Public Health, Yangzhou University, Yangzhou, 225009, China.
| | - Man Qu
- School of Public Health, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
23
|
Liu T, Han S, Yao Y, Zhang G. Role of Human Monocarboxylate Transporter 1 (hMCT1) and 4 (hMCT4) in Tumor Cells and the Tumor Microenvironment. Cancer Manag Res 2023; 15:957-975. [PMID: 37693221 PMCID: PMC10487743 DOI: 10.2147/cmar.s421771] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 08/30/2023] [Indexed: 09/12/2023] Open
Abstract
In recent years, the abnormal glucose metabolism of tumor cells has attracted increasing attention. Abnormal glucose metabolism is closely related to the occurrence and development of tumors. Monocarboxylate transporters (MCTs) transport the sugar metabolites lactic acid and pyruvate, which affect glucose metabolism and tumor progression in a variety of ways. Thus, research has recently focused on MCTs and their potential functions in cancer. The MCT superfamily consists of 14 members. MCT1 and MCT4 play a crucial role in the maintenance of intracellular pH in tumor cells by transporting monocarboxylic acids (such as lactate, pyruvate and butyrate). MCT1 and MCT4 are highly expressed in a variety of tumor cells and are involved the proliferation, invasion and migration of tumor cells, which are closely related to the prognosis of cancer. Because of their important functions in tumor cells, MCT1 and MCT4 have become potential targets for cancer treatment. In this review, we focus on the structure, function and regulation of MCT1 and MCT4 and discuss the developed inhibitors of MCT1 and MCT4 to provide more comprehensive information that might aid in the development of strategies targeting MCTs in cancer.
Collapse
Affiliation(s)
- Tian Liu
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Shangcong Han
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, People’s Republic of China
| | - Yu Yao
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Guiming Zhang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| |
Collapse
|
24
|
Lee S, Choi Y, Jeong E, Park J, Kim J, Tanaka M, Choi J. Physiological significance of elevated levels of lactate by exercise training in the brain and body. J Biosci Bioeng 2023; 135:167-175. [PMID: 36681523 DOI: 10.1016/j.jbiosc.2022.12.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/15/2022] [Accepted: 12/07/2022] [Indexed: 01/21/2023]
Abstract
For the past 200 years, lactate has been regarded as a metabolic waste end product that causes fatigue during exercise. However, lactate production is closely correlated with energy metabolism. The lactate dehydrogenase-catalyzed reaction uses protons to produce lactate, which delays ongoing metabolic acidosis. Of note, lactate production differs depending on exercise intensity and is not limited to muscles. Importantly, controlling physiological effect of lactate may be a solution to alleviating some chronic diseases. Released through exercise, lactate is an important biomarker for fat oxidation in skeletal muscles. During recovery after sustained strenuous exercise, most of the lactate accumulated during exercise is removed by direct oxidation. However, as the muscle respiration rate decreases, lactate becomes a desirable substrate for hepatic glucose synthesis. Furthermore, improvement in brain function by lactate, particularly, through the expression of vascular endothelial growth factor and brain-derived neurotrophic factor, is being increasingly studied. In addition, it is possible to improve stress-related symptoms, such as depression, by regulating the function of hippocampal mitochondria, and with an increasingly aging society, lactate is being investigated as a preventive agent for brain diseases such as Alzheimer's disease. Therefore, the perception that lactate is equivalent to fatigue should no longer exist. This review focuses on the new perception of lactate and how lactate acts extensively in the skeletal muscles, heart, brain, kidney, and liver. Additionally, lactate is now used to confirm exercise performance and should be further studied to assess its impact on exercise training.
Collapse
Affiliation(s)
- Sungjun Lee
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea; Feynman Institute of Technology, Nanomedicine Corporation, Seoul 06974, Republic of Korea
| | - Yonghyun Choi
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea; Feynman Institute of Technology, Nanomedicine Corporation, Seoul 06974, Republic of Korea
| | - Eunseo Jeong
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Jongjun Park
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Jiwon Kim
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea; Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Masayoshi Tanaka
- Department of Chemical Science and Engineering, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama-shi, Kanagawa 226-8503, Japan
| | - Jonghoon Choi
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea; Feynman Institute of Technology, Nanomedicine Corporation, Seoul 06974, Republic of Korea; Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
25
|
Unraveling the Peculiar Features of Mitochondrial Metabolism and Dynamics in Prostate Cancer. Cancers (Basel) 2023; 15:cancers15041192. [PMID: 36831534 PMCID: PMC9953833 DOI: 10.3390/cancers15041192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023] Open
Abstract
Prostate cancer (PCa) is the second leading cause of cancer deaths among men in Western countries. Mitochondria, the "powerhouse" of cells, undergo distinctive metabolic and structural dynamics in different types of cancer. PCa cells experience peculiar metabolic changes during their progression from normal epithelial cells to early-stage and, progressively, to late-stage cancer cells. Specifically, healthy cells display a truncated tricarboxylic acid (TCA) cycle and inefficient oxidative phosphorylation (OXPHOS) due to the high accumulation of zinc that impairs the activity of m-aconitase, the enzyme of the TCA cycle responsible for the oxidation of citrate. During the early phase of cancer development, intracellular zinc levels decrease leading to the reactivation of m-aconitase, TCA cycle and OXPHOS. PCa cells change their metabolic features again when progressing to the late stage of cancer. In particular, the Warburg effect was consistently shown to be the main metabolic feature of late-stage PCa cells. However, accumulating evidence sustains that both the TCA cycle and the OXPHOS pathway are still present and active in these cells. The androgen receptor axis as well as mutations in mitochondrial genes involved in metabolic rewiring were shown to play a key role in PCa cell metabolic reprogramming. Mitochondrial structural dynamics, such as biogenesis, fusion/fission and mitophagy, were also observed in PCa cells. In this review, we focus on the mitochondrial metabolic and structural dynamics occurring in PCa during tumor development and progression; their role as effective molecular targets for novel therapeutic strategies in PCa patients is also discussed.
Collapse
|
26
|
Dual Roles of Lactate in EGFR-TKI-Resistant Lung Cancer by Targeting GPR81 and MCT1. JOURNAL OF ONCOLOGY 2022; 2022:3425841. [PMID: 36545125 PMCID: PMC9763017 DOI: 10.1155/2022/3425841] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/07/2022] [Accepted: 11/26/2022] [Indexed: 12/15/2022]
Abstract
Lactate is critical in modeling tumor microenvironment causing chemotherapy resistance; however, the role of lactate in tyrosine kinase inhibitor (TKI) resistance has not been fully known. The aim of this study was to evaluate whether lactate could mediate TKI resistance through GPR81 and MCT1 in non-small-cell lung cancer (NSCLC). Here, we showed that lactate enhanced the cell viability and restrained erlotinib-induced apoptosis in PC9 and HCC827 cells. GPR81 and AKT expression were significantly increased with the addition of lactate, and siGPR81 reduced AKT expression resulting in a raised apoptosis rate with erlotinib treatment. Furthermore, we found that lactate also promoted MCT1 exposure, and inhibiting MCT1 with AZD3965 markedly impaired the glycolytic capacity. A significant increase of GPR81 and MCT1 expression was observed in insensitive tissues compared with sensitive ones by immunostaining in NSCLC patients. Our results indicate that lactate adopts dual strategies to promote TKI resistance in NSCLC, not only activating AKT signaling by GPR81, but also giving energy supply through MCT1-mediated input. Targeting GPR81 and MCT1 may provide new therapeutic modalities for TKI resistance in NSCLC.
Collapse
|
27
|
Duan Q, Zhang S, Wang Y, Lu D, Sun Y, Wu Y. Proton-coupled monocarboxylate transporters in cancer: From metabolic crosstalk, immunosuppression and anti-apoptosis to clinical applications. Front Cell Dev Biol 2022; 10:1069555. [PMID: 36506099 PMCID: PMC9727313 DOI: 10.3389/fcell.2022.1069555] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/02/2022] [Indexed: 11/24/2022] Open
Abstract
The Warburg effect is known as the hyperactive glycolysis that provides the energy needed for rapid growth and proliferation in most tumor cells even under the condition of sufficient oxygen. This metabolic pattern can lead to a large accumulation of lactic acid and intracellular acidification, which can affect the growth of tumor cells and lead to cell death. Proton-coupled monocarboxylate transporters (MCTs) belong to the SLC16A gene family, which consists of 14 members. MCT1-4 promotes the passive transport of monocarboxylate (e.g., lactate, pyruvate, and ketone bodies) and proton transport across membranes. MCT1-4-mediated lactate shuttling between glycolytic tumor cells or cancer-associated fibroblasts and oxidative tumor cells plays an important role in the metabolic reprogramming of energy, lipids, and amino acids and maintains the survival of tumor cells. In addition, MCT-mediated lactate signaling can promote tumor angiogenesis, immune suppression and multidrug resistance, migration and metastasis, and ferroptosis resistance and autophagy, which is conducive to the development of tumor cells and avoid death. Although there are certain challenges, the study of targeted drugs against these transporters shows great promise and may form new anticancer treatment options.
Collapse
Affiliation(s)
- Qixin Duan
- Department of Urology, Affiliated Sanming First Hospital of Fujian Medical University, Sanming, Fujian, China,Department of Urology, Nanyang Central Hospital, Nanyang, China
| | - Shuang Zhang
- Department of Nursing, Nanyang Central Hospital, Nanyang, China
| | - Yang Wang
- Department of Urology, Nanyang Central Hospital, Nanyang, China
| | - Dongming Lu
- Department of Urology, Affiliated Sanming First Hospital of Fujian Medical University, Sanming, Fujian, China
| | - Yingming Sun
- Department of Medical and Radiation Oncology, Affiliated Sanming First Hospital of Fujian Medical University, Sanming, Fujian, China,*Correspondence: Yongyang Wu, ; Yingming Sun,
| | - Yongyang Wu
- Department of Urology, Affiliated Sanming First Hospital of Fujian Medical University, Sanming, Fujian, China,*Correspondence: Yongyang Wu, ; Yingming Sun,
| |
Collapse
|
28
|
Mon Hla H, Hunprasit V, Siripoonsup J, Rungsipipat A, Radtanakatikanon A. Diagnostic utility of LDH measurement for determining the etiology of modified transudate pleural effusion in cats. Front Vet Sci 2022; 9:1044192. [PMID: 36406069 PMCID: PMC9669425 DOI: 10.3389/fvets.2022.1044192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022] Open
Abstract
Fluid analysis is an initial approach for determining the underlying causes of body cavity effusions. Modified transudate is commonly diagnosed in pleural effusion in cats, however, it provides limited diagnostic information. Aims of this study were to investigate common etiologies causing different pleural fluid types and to evaluate the usefulness of lactate dehydrogenase (LDH) for differentiating the etiology in modified transudates in cats. Pleural effusion samples from 122 cats were analyzed and classified into three types: transudate, modified transudate, and exudate. Causes of pleural effusion were classified into four conditions: cardiac disease, neoplasia, feline infectious peritonitis (FIP), and pyothorax. The relationship of underlying etiology and fluid types was described. The LDH levels in pleural fluid and plasma were compared between the causes in the samples classified as modified transudate. The fluid analysis of pleural effusion showed that modified transudate was the most common fluid type (44.2%). Neoplasia was predominantly diagnosed (38.5%) as the etiology of pleural effusion. There was no significant correlation between pleural fluid and plasma LDH level in any type of pleural fluid, suggesting that pleural fluid LDH does not appear to be affected by plasma LDH. The occurrence of modified transudate was not associated to its etiologies, however, the LDH level in modified transudates showed significant differences between etiologic groups. The LDH level in modified transudate was excellent in separating cardiac from non-cardiac diseases with a cut-off value of <535 U/L and separating FIP from non-FIP diseases with a cut-off value of >641 U/L. Based on the current findings, pleural fluid LDH can be a useful adjunctive marker for differentiating some causes of modified transudate pleural effusion and should be added in the routine diagnostic work-up of feline patients with pleural effusions.
Collapse
Affiliation(s)
- Hsu Mon Hla
- The International Graduate Program of Veterinary Science and Technology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Vachira Hunprasit
- Department of Veterinary Medicine, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Jedsada Siripoonsup
- Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Anudep Rungsipipat
- Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence for Companion Animal Cancer, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Araya Radtanakatikanon
- Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence for Companion Animal Cancer, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- *Correspondence: Araya Radtanakatikanon
| |
Collapse
|
29
|
Choi SYC, Ribeiro CF, Wang Y, Loda M, Plymate SR, Uo T. Druggable Metabolic Vulnerabilities Are Exposed and Masked during Progression to Castration Resistant Prostate Cancer. Biomolecules 2022; 12:1590. [PMID: 36358940 PMCID: PMC9687810 DOI: 10.3390/biom12111590] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 08/27/2023] Open
Abstract
There is an urgent need for exploring new actionable targets other than androgen receptor to improve outcome from lethal castration-resistant prostate cancer. Tumor metabolism has reemerged as a hallmark of cancer that drives and supports oncogenesis. In this regard, it is important to understand the relationship between distinctive metabolic features, androgen receptor signaling, genetic drivers in prostate cancer, and the tumor microenvironment (symbiotic and competitive metabolic interactions) to identify metabolic vulnerabilities. We explore the links between metabolism and gene regulation, and thus the unique metabolic signatures that define the malignant phenotypes at given stages of prostate tumor progression. We also provide an overview of current metabolism-based pharmacological strategies to be developed or repurposed for metabolism-based therapeutics for castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Stephen Y. C. Choi
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Caroline Fidalgo Ribeiro
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, NY 10021, USA
| | - Yuzhuo Wang
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, NY 10021, USA
- New York Genome Center, New York, NY 10013, USA
| | - Stephen R. Plymate
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, 850 Republican St., Seattle, WA 98109, USA
- Geriatrics Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Takuma Uo
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, 850 Republican St., Seattle, WA 98109, USA
| |
Collapse
|
30
|
Slc2a6 regulates myoblast differentiation by targeting LDHB. Cell Commun Signal 2022; 20:107. [PMID: 35850889 PMCID: PMC9290262 DOI: 10.1186/s12964-022-00915-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 06/10/2022] [Indexed: 11/24/2022] Open
Abstract
Background Type 2 diabetes mellitus is a global health problem. It often leads to a decline in the differentiation capacity of myoblasts and progressive loss of muscle mass, which in turn results in deterioration of skeletal muscle function. However, effective therapies against skeletal muscle diseases are unavailable. Methods Skeletal muscle mass and differentiation ability were determined in db/+ and db/db mice. Transcriptomics and metabolomics approaches were used to explore the genetic mechanism regulating myoblast differentiation in C2C12 myoblasts. Results In this study, the relatively uncharacterized solute carrier family gene Slc2a6 was found significantly up-regulated during myogenic differentiation and down-regulated during diabetes-induced muscle atrophy. Moreover, RNAi of Slc2a6 impaired the differentiation and myotube formation of C2C12 myoblasts. Both metabolomics and RNA-seq analyses showed that the significantly differentially expressed genes (e.g., LDHB) and metabolites (e.g., Lactate) during the myogenic differentiation of C2C12 myoblasts post-Slc2a6-RNAi were enriched in the glycolysis pathway. Furthermore, we show that Slc2a6 regulates the myogenic differentiation of C2C12 myoblasts partly through the glycolysis pathway by targeting LDHB, which affects lactic acid accumulation. Conclusion Our study broadens the understanding of myogenic differentiation and offers the Slc2a6-LDHB axis as a potential therapeutic target for the treatment of diabetes-associated muscle atrophy. Video abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-022-00915-2.
Collapse
|
31
|
Zalpoor H, Akbari A, Nayerain Jazi N, Liaghat M, Bakhtiyari M. Possible role of autophagy induced by COVID-19 in cancer progression, chemo-resistance, and tumor recurrence. Infect Agent Cancer 2022; 17:38. [PMID: 35850916 PMCID: PMC9289088 DOI: 10.1186/s13027-022-00450-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/13/2022] [Indexed: 12/23/2022] Open
Abstract
COVID-19 infection is a serious threat to patients with primary diseases, especially multiple cancers. Studies suggest that cancer patients are one of the most susceptible populations to experience severe COVID-19 and death. In addition, a number of studies suggest various mechanisms for SARS-CoV-2 in cancer progression. In this study, we discussed the role of SARS-CoV-2 in the induction of autophagy and we hypothesized that autophagy induced by COVID-19 not only can contribute to viral replication but also potentially can lead to cancer progression, chemo-resistance, and tumor recurrence in multiple cancer patients. Therefore, targeting autophagy-related signaling pathways and cellular and molecular processes could be a potentially promising therapeutic approach for cancer patients with COVID-19. Hence, this study can shed light on a new window on the management of such patients. However, more investigations in the future are required to understand other pathological effects of COVID-19 infection on cancer patients to provide new therapeutic strategies to combat these complications in these patients.
Collapse
Affiliation(s)
- Hamidreza Zalpoor
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran. .,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran.
| | - Abdullatif Akbari
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Negar Nayerain Jazi
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahsa Liaghat
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran.,Department of Medical Laboratory Sciences, Faculty of Medical Sciences, Kazerun Branch, Islamic Azad University, Kazerun, Iran
| | - Maryam Bakhtiyari
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran.,Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| |
Collapse
|
32
|
Yi C, Yu AM. MicroRNAs in the Regulation of Solute Carrier Proteins Behind Xenobiotic and Nutrient Transport in Cells. Front Mol Biosci 2022; 9:893846. [PMID: 35755805 PMCID: PMC9220936 DOI: 10.3389/fmolb.2022.893846] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 05/02/2022] [Indexed: 11/16/2022] Open
Abstract
Altered metabolism, such as aerobic glycolysis or the Warburg effect, has been recognized as characteristics of tumor cells for almost a century. Since then, there is accumulating evidence to demonstrate the metabolic reprogramming of tumor cells, addiction to excessive uptake and metabolism of key nutrients, to support rapid proliferation and invasion under tumor microenvironment. The solute carrier (SLC) superfamily transporters are responsible for influx or efflux of a wide variety of xenobiotic and metabolites that are needed for the cells to function, as well as some medications. To meet the increased demand for nutrients and energy, SLC transporters are frequently dysregulated in cancer cells. The SLCs responsible for the transport of key nutrients for cancer metabolism and energetics, such as glucose and amino acids, are of particular interest for their roles in tumor progression and metastasis. Meanwhile, rewired metabolism is accompanied by the dysregulation of microRNAs (miRNAs or miRs) that are small, noncoding RNAs governing posttranscriptional gene regulation. Studies have shown that many miRNAs directly regulate the expression of specific SLC transporters in normal or diseased cells. Changes of SLC transporter expression and function can subsequently alter the uptake of nutrients or therapeutics. Given the important role for miRNAs in regulating disease progression, there is growing interest in developing miRNA-based therapies, beyond serving as potential diagnostic or prognostic biomarkers. In this article, we discuss how miRNAs regulate the expression of SLC transporters and highlight potential influence on the supply of essential nutrients for cell metabolism and drug exposure toward desired efficacy.
Collapse
Affiliation(s)
- Colleen Yi
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Ai-Ming Yu
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, United States
| |
Collapse
|
33
|
Resurreccion EP, Fong KW. The Integration of Metabolomics with Other Omics: Insights into Understanding Prostate Cancer. Metabolites 2022; 12:metabo12060488. [PMID: 35736421 PMCID: PMC9230859 DOI: 10.3390/metabo12060488] [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: 04/30/2022] [Revised: 05/21/2022] [Accepted: 05/24/2022] [Indexed: 02/06/2023] Open
Abstract
Our understanding of prostate cancer (PCa) has shifted from solely caused by a few genetic aberrations to a combination of complex biochemical dysregulations with the prostate metabolome at its core. The role of metabolomics in analyzing the pathophysiology of PCa is indispensable. However, to fully elucidate real-time complex dysregulation in prostate cells, an integrated approach based on metabolomics and other omics is warranted. Individually, genomics, transcriptomics, and proteomics are robust, but they are not enough to achieve a holistic view of PCa tumorigenesis. This review is the first of its kind to focus solely on the integration of metabolomics with multi-omic platforms in PCa research, including a detailed emphasis on the metabolomic profile of PCa. The authors intend to provide researchers in the field with a comprehensive knowledge base in PCa metabolomics and offer perspectives on overcoming limitations of the tool to guide future point-of-care applications.
Collapse
Affiliation(s)
- Eleazer P. Resurreccion
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40506, USA;
| | - Ka-wing Fong
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40506, USA;
- Markey Cancer Center, University of Kentucky, Lexington, KY 40506, USA
- Correspondence: ; Tel.: +1-859-562-3455
| |
Collapse
|
34
|
Chhonker SK, Rawat D, Koiri RK. Repurposing PDE5 inhibitor tadalafil and sildenafil as anticancer agent against hepatocellular carcinoma via targeting key events of glucose metabolism and multidrug resistance. J Biochem Mol Toxicol 2022; 36:e23100. [PMID: 35608386 DOI: 10.1002/jbt.23100] [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: 08/21/2021] [Revised: 02/28/2022] [Accepted: 05/11/2022] [Indexed: 11/12/2022]
Abstract
Hepatocellular carcinoma (HCC) has emerged as one of the most common and lethal cancers worldwide and is caused due to contamination of diets with aflatoxin B1 and chronic viral hepatitis. Recent reports suggest that phosphodiesterase-5 inhibitor (PDE5i) exhibits anticancer properties against several forms of cancer but till now has not been evaluated against HCC. We aimed to evaluate the anticancer property of phosphodiesterase-5 inhibitors (PDE5i) tadalafil and sildenafil against aflatoxin B1 HCC. Rats of HCC group were fed with 5% alcohol via drinking water for 3 weeks, followed by administration of AFB1 (1 mg/kg/bw, i.p.) at an interval of two subsequent days. PDE5i (tadalafil and sildenafil, 10 mg/kg bw) was administered along with drinking water after 6 weeks of treatment with AFB1 for 2 weeks. In the present investigation, in HCC elevation in the level of SGOT, SGPT, ALP, and urea vis-à-vis activity of key glycolytic enzyme LDH and mRNA expression of c-myc, Akt, LDH-A, and PFKFB3 was noted. Similarly, the level of multidrug resistance protein (MDR) and breast cancer resistance protein (BCRP/ABCG2) was elevated along with increased expression of angiogenesis marker (HIF-1α, VEGF, and TGF-β1) in HCC. Post-treatment with PDE5 inhibitor (tadalafil and sildenafil) downregulated and brought back the above parameters towards normal and out of two PDE5i (tadalafil and sildenafil), sildenafil effect was more potent as compared to tadalafil. Our findings demonstrate for the first time that PDE5 inhibitors tadalafil and sildenafil are able to prohibit the development and progression of aflatoxin B1 induced HCC.
Collapse
Affiliation(s)
- Saurabh Kumar Chhonker
- Biochemistry Laboratory, Department of Zoology, School of Biological Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh, India
| | - Divya Rawat
- Biochemistry Laboratory, Department of Zoology, School of Biological Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh, India
| | - Raj Kumar Koiri
- Biochemistry Laboratory, Department of Zoology, School of Biological Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh, India
| |
Collapse
|