Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 279
Filter
1.
Cureus ; 16(7): e63852, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39099978

ABSTRACT

The element that causes hypoxia when the von Hippel-Lindau (VHL) protein is not functioning is hypoxia-inducible factor 1-alpha (HIF-1α), which is the essential protein linked to cell control under hypoxia. Consequently, in situations where cells are oxygen-deficient, HIF-1α carries out a variety of essential functions. Citations to relevant literature support the notion that HIF-1α regulates the mitochondrial and glycolytic pathways, as well as the transition from the former to the latter. Cells with limited oxygen supply benefit from this change, which is especially beneficial for the inhibition of the mitochondrial electron transport chain and enhanced uptake of glucose and lactate. During hypoxic stress, HIF-1α also controls proline and glycolytic transporters such as lactate dehydrogenase A (LDHA) and glucose transporter 1 (GLUT1). These mechanisms help the cell return to homeostasis. Therefore, through metabolic change promoting adenosine triphosphate (ATP) synthesis and reducing reactive oxygen species (ROS) creation, HIF-1α may have a role in reducing oxidative stress in cells. This evidence, which describes the function of HIF-1α in many molecular pathways, further supports the notion that it is prognostic and that it contributes to hypoxic cell adaption. Understanding more about disorders, including inflammation, cancer, and ischemia, is possible because of HIF-1α's effect on metabolic changes. Gaining knowledge about the battle between metabolism, which is directed by HIF-1α, would help advance the research on pathophysiological situations involving dysregulated hypoxia and metabolism.

2.
Cell Rep ; 43(8): 114632, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39159042

ABSTRACT

Tumor cells undergo uncontrolled proliferation driven by enhanced anabolic metabolism including glycolysis and glutaminolysis. Targeting these pathways to inhibit cancer growth is a strategy for cancer treatment. Critically, however, tumor-responsive T cells share metabolic features with cancer cells, making them susceptible to these treatments as well. Here, we assess the impact on anti-tumor T cell immunity and T cell exhaustion by genetic ablation of lactate dehydrogenase A (LDHA) and glutaminase1 (GLS1), key enzymes in aerobic glycolysis and glutaminolysis. Loss of LDHA severely impairs expansion of T cells in response to tumors and chronic infection. In contrast, T cells lacking GLS1 can compensate for impaired glutaminolysis by engaging alternative pathways, including upregulation of asparagine synthetase, and thus efficiently respond to tumor challenge and chronic infection as well as immune checkpoint blockade. Targeting GLS1-dependent glutaminolysis, but not aerobic glycolysis, may therefore be a successful strategy in cancer treatment, particularly in combination with immunotherapy.


Subject(s)
Glutaminase , Glutamine , Glycolysis , Glutaminase/metabolism , Glutaminase/antagonists & inhibitors , Glutamine/metabolism , Animals , Mice , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Humans , Mice, Inbred C57BL , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Lactate Dehydrogenase 5/metabolism , Cell Line, Tumor , Immunity
3.
Asian Pac J Cancer Prev ; 25(8): 2929-2944, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39205592

ABSTRACT

BACKGROUND: Understanding the heterogeneous nature of breast cancer, including the role of LDHA expression regulation via non-coding RNAs in prognosis, is still unknown, highlighting the need for more research into its molecular roles and diagnostic approaches. METHODS: The study utilized various computer tools to analyze the differences between LDHA in tissues and cancer cells. It used data from TIMER 2.0, UALCAN, and TISIDB to study gene expression and survival outcomes in breast cancer patients. The study also used the Breast Cancer Gene Expression Miner to examine the relationship between LDHA gene expression and breast cancer type. Other tools included TCGAPortal, TNMplot, ctcRbase, GSCA, Enrichr, TISIDB, Oncomx, and TANRIC. The study then explored the relationship between tumor-infiltrating immune cells and LDHA formation using the GSCA and TISIDB repositories. We used Auto Dock Tools 1.5.6 to perform ligand binding analysis for LDHA, withanolides, and the known inhibitor LDH-IN-1. LigPlot+ and Pymol were used for visualization of protein-ligand complexes. RESULTS: LDHA overexpression in breast cancer cells, metastatic tissue, and circulating tumor cells leads to shortened recurrence-free survival, overall survival, and distant metastasis-free survival. In invasive breast cell carcinoma, we observed that LDHA/HIF-1α /TMPO-AS1 are overexpressed while miR-383-5p is downregulated. This overexpression is associated with poor prognosis and may lead to Act_DC infiltration into the tumor microenvironment. Withanolides, viz., Withaferine A and Withanolide D, have shown high binding affinity with LDHA, with binding energies of -9.3kcal/mol and -10kcal/mol respectively. These could be attractive choices for small-molecule inhibitor design against LDHA.


Subject(s)
MicroRNAs , RNA, Long Noncoding , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/metabolism , Female , RNA, Long Noncoding/genetics , MicroRNAs/genetics , Prognosis , Gene Expression Regulation, Neoplastic , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Cell Proliferation , Survival Rate , L-Lactate Dehydrogenase
4.
Autoimmunity ; 57(1): 2384889, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39086231

ABSTRACT

Osteoarthritis (OA) is a worldwide joint disease, leading to the physical pain, stiffness, and even disability. Lactate dehydrogenase A (LDHA) is known as a lactylation mediator that can regulate histone lactylation of its target genes. However, the role of LDHA-mediated histone H3 lysine 18 lactylation (H3K18la) in OA progression is yet to be clarified. Our study aims at revealing the role and mechanism of LDHA-mediated histone lactylation in the glycolysis of chondrocytes. In this study, we determined at first that the H3K18la level was enhanced in OA. Energy metabolism such as glycolysis is often altered in OA progress. Therefore, we further explored the mechanism mediating glycolysis and thus promoting OA progress. Moreover, glycolysis was enhanced in LPS-induced OA cell model, as evidenced by the increased glucose consumption and lactate production. Furthermore, we silenced LDHA for loss-of-function assays. The results showed that knockdown of LDHA suppressed glycolysis of LPS-induced chondrocytes. In vivo animal study demonstrated that knockout of LDHA recovered cartilage injury of OA mice. Mechanistically, we uncovered that LDHA-mediated H3K18la in TPI1 promoter enhanced the transcription activity of TPI1. Mutation of K69 site was found to ameliorate LPS-induced glycolysis in OA cell model. In conclusion, our study reveals the role of LDHA-mediated H3K18la of TPI1 promoter in OA progress.


Subject(s)
Chondrocytes , Glycolysis , Histones , Osteoarthritis , Osteoarthritis/metabolism , Osteoarthritis/genetics , Osteoarthritis/pathology , Animals , Mice , Histones/metabolism , Humans , Chondrocytes/metabolism , Disease Models, Animal , Lactate Dehydrogenase 5/metabolism , Male , Gene Expression Regulation , Mice, Knockout , Promoter Regions, Genetic , Transcription, Genetic
5.
J Tradit Complement Med ; 14(4): 414-423, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39035691

ABSTRACT

Background and aim: Hypoxia of the cartilage has been considered as a potential pathogenic factor in knee osteoarthritis (KOA). Studies have shown that impaired blood perfusion of joint leads to cartilage hypoxia. Electroacupuncture (EA) has proven effects on pain relief and improving microcirculation. This study aimed to explore the effect of EA on articular microcirculation and cartilage anoxic and the underlying mechanisms. Procedures: Videman's method was used for 6 weeks to establish the KOA model. EA intervention was performed in four points around the knee for 3 weeks after KOA modeling. The Lequesne MG score was used to assess ethology. We recorded the oxygen tension of synovial fluid and the synovial microcirculation in vivo. HE-staining was used to assess cartilage morphology, and immunohistochemistry (IHC), Western blotting, and RT-PCR were used to assess expression of the major glycolytic enzymes glucosetransporter1 (GLUT1), pyruvate kinase M2(PKM2), and lactate dehydrogenase A (LDHA). Enzyme-linked immunosorbent assay (Elisa) was used to detect lactate content. Results and conclusion: There was a significant decrease in Lequesne MG score and improvement in Mankin score after EA intervention (P < 0.01), a significant increase in synovial microcirculation (P < 0.05) and synovial fluid oxygen tension (P < 0.01), and there was significant decrease in the expression of GLUT1, PKM2 and LDHA (P < 0.01) and lactate (P < 0.05). This study suggested that EA ameliorate cartilage hypoxia and regulate glycolytic metabolism in chondrocytes in KOA model rabbits by improving articular microcirculation and oxygen tension.

6.
Phytomedicine ; 133: 155874, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39079314

ABSTRACT

BACKGROUND: The presence of distant metastasis at the time of initial diagnosis is a prevalent issue in non-small cell lung cancer (NSCLC), affecting around 30-40 % of the patients. Acidic tumor microenvironment (TME) provides favorable conditions that increase the invasiveness and aggressiveness of NSCLC. The activity of the glycolytic enzyme lactate dehydrogenase (LDHA) increases intracellular lactate accumulation, which creates an acidic TME. However, it is not yet known whether LDHA is involved in enhancing the metastatic potential of NSCLC and if LDHA is a druggable therapeutic target for NSCLC. PURPOSE: We aimed to investigate the molecular mechanisms underlying the enhanced NSCLC metastasis in acidic TME, and to explore whether sulforaphane (SFN), an active compound in Raphani Semen, can serve as a LDHA inhibitor to inhibit NSCLC metastasis in the acidic TME. METHODS: To mimic the acidic TME, NSCLC cells were cultured in acidic medium (pH 6.6), normal medium (pH 7.4) served as control. Western blotting, bioinformatic analysis, luciferase assay and rescue experiments were used to explore the mechanism and investigate the anti-metastatic effect of SFN both in vitro and in vivo. RESULTS: Acidic environment increases the expression of LDHA which in turn increases the production of lactic acid that contributes to the acidity of TME. Interestingly, elevated LDHA expression results from increased c-Myc expression, which transactivates LDHA. c-Myc expression is directly regulated by miR-7-5p. In vitro study shows that overexpression of miR-7-5p reverses the acidic pH-enhanced c-Myc and LDHA expressions and also abolishes the enhanced NSCLC cell migration. More importantly, SFN significantly inhibits NSCLC growth and metastasis by reducing lactate production via the miR-7-5p/c-Myc/LDHA axis. Besides, it also regulates the expressions of monocarboxylate transporter 1 (MCT1) and MCT4 that transport lactate across cell membrane. CONCLUSIONS: The miR-7-5p/c-Myc/LDHA axis is involved in the enhanced NSCLC metastasis in the acidic TME. SFN, a novel LDHA inhibitor, reduces lactate production by targeting the miR-7-5p/c-Myc/LDHA axis, and hence inhibits NSCLC metastasis. Our findings not only delineate a novel mechanism, but also support the clinical translation of SFN as a novel therapeutic agent for treating metastatic NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Isothiocyanates , L-Lactate Dehydrogenase , Lung Neoplasms , MicroRNAs , Proto-Oncogene Proteins c-myc , Sulfoxides , Tumor Microenvironment , Isothiocyanates/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Tumor Microenvironment/drug effects , Sulfoxides/pharmacology , MicroRNAs/metabolism , Humans , Lung Neoplasms/drug therapy , Animals , Proto-Oncogene Proteins c-myc/metabolism , Cell Line, Tumor , L-Lactate Dehydrogenase/metabolism , Mice, Nude , Mice , Hydrogen-Ion Concentration , Mice, Inbred BALB C , Neoplasm Metastasis , Cell Movement/drug effects
7.
J Transl Med ; 22(1): 676, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39044184

ABSTRACT

BACKGROUND: Breast cancer manifests as a heterogeneous pathology marked by complex metabolic reprogramming essential to satisfy its energy demands. Oncogenic signals boost the metabolism, modifying fatty acid synthesis and glucose use from the onset to progression and therapy resistant-forms. However, the exact contribution of metabolic dependencies during tumor evolution remains unclear. METHODS: In this study, we elucidate the connection between FASN and LDHA, pivotal metabolic genes, and their correlation with tumor grade and therapy response using datasets from public repositories. Subsequently, we evaluated the metabolic and proliferative functions upon FASN and LDHA inhibition in breast cancer models. Lastly, we integrated metabolomic and lipidomic analysis to define the contributions of metabolites, lipids, and precursors to the metabolic phenotypes. RESULTS: Collectively, our findings indicate metabolic shifts during breast cancer progression, unvealling two distinct functional energy phenotypes associated with aggressiveness and therapy response. Specifically, FASN exhibits reduced expression in advance-grade tumors and therapy-resistant forms, whereas LDHA demonstrates higher expression. Additionally, the biological and metabolic impact of blocking the enzymatic activity of FASN and LDHA was correlated with resistant conditions. CONCLUSIONS: These observations emphasize the intrinsic metabolic heterogeneity within breast cancer, thereby highlighting the relevance of metabolic interventions in the field of precision medicine.


Subject(s)
Breast Neoplasms , Fatty Acid Synthase, Type I , Humans , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/enzymology , Female , Fatty Acid Synthase, Type I/metabolism , Fatty Acid Synthase, Type I/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm , Cell Proliferation , Gene Expression Regulation, Neoplastic , Lipidomics , Metabolomics , L-Lactate Dehydrogenase
8.
Front Mol Biosci ; 11: 1301099, 2024.
Article in English | MEDLINE | ID: mdl-38993839

ABSTRACT

Introduction: Hepatocellular carcinoma (HCC), which is closely associated with chronicinflammation, is the most common liver cancer and primarily involves dysregulated immune responses in the precancerous microenvironment. Currently, most studies have been limited to HCC incidence. However, the immunopathogenic mechanisms underlying precancerous lesions remain unknown. Methods: We obtained single-cell sequencing data (GSE136103) from two nonalcoholic fatty liver disease (NAFLD) cirrhosis samples and five healthy samples. Using pseudo-time analysis, we systematically identified five different T-cell differentiation states. Ten machine-learning algorithms were used in 81 combinations to integrate the frameworks and establish the best T-cell differentiation-related prognostic signature in a multi-cohort bulk transcriptome analysis. Results: LDHA was considered a core gene, and the results were validated using multiple external datasets. In addition, we validated LDHA expression using immunohistochemistry and flow cytometry. Conclusion: LDHA is a crucial marker gene in T cells for the progression of NAFLD cirrhosis to HCC.

9.
Eur J Med Res ; 29(1): 310, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38840262

ABSTRACT

KDM6A (lysine demethylase 6A) has been reported to undergo inactivating mutations in colorectal cancer, but its function in the progression of colorectal cancer has not been evaluated using animal models of colorectal cancer. In this study, we found that knocking out KDM6A expression in mouse intestinal epithelium increased the length of villus and crypt, promoting the development of AOM (azoxymethane)/DSS (dextran sulfate sodium salt)-induced colorectal cancer. On the other hand, knocking down KDM6A expression promoted the growth of colorectal cancer cells. In molecular mechanism studies, we found that KDM6A interacts with HIF-1α; knocking down KDM6A promotes the binding of HIF-1α to the LDHA promoter, thereby promoting LDHA expression and lactate production, enhancing glycolysis. Knocking down LDHA reversed the malignant phenotype caused by KDM6A expression loss. In summary, this study using animal models revealed that KDM6A loss promotes the progression of colorectal cancer through reprogramming the metabolism of the colorectal cancer cells, suggesting that restoring the function of KDM6A is likely to be one of the strategies for colorectal cancer treatment.


Subject(s)
Colorectal Neoplasms , Disease Progression , Glycolysis , Histone Demethylases , Animals , Humans , Mice , Cell Proliferation/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Histone Demethylases/metabolism , Histone Demethylases/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics
10.
Int Immunopharmacol ; 136: 112338, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38850787

ABSTRACT

Cardiac fibrosis is a typical feature of cardiac pathological remodeling, which is associated with adverse clinical outcomes and has no effective therapy. Nicotine is an important risk factor for cardiac fibrosis, yet its underlying molecular mechanism remains poorly understood. This study aimed to identify its potential molecular mechanism in nicotine-induced cardiac fibrosis. Our results showed nicotine exposure led to the proliferation and transformation of cardiac fibroblasts (CFs) into myofibroblasts (MFs) by impairing autophagy flux. Through the use of drug affinity responsive target stability (DARTS) assay, cellular thermal shift assay (CETSA), and surface plasmon resonance (SPR) technology, it was discovered that nicotine directly increased the stability and protein levels of lactate dehydrogenase A (LDHA) by binding to it. Nicotine treatment impaired autophagy flux by regulating the AMPK/mTOR signaling pathway, impeding the nuclear translocation of transcription factor EB (TFEB), and reducing the activity of cathepsin B (CTSB). In vivo, nicotine treatment exacerbated cardiac fibrosis induced in spontaneously hypertensive rats (SHR) and worsened cardiac function. Interestingly, the absence of LDHA reversed these effects both in vitro and in vivo. Our study identified LDHA as a novel nicotine-binding protein that plays a crucial role in mediating cardiac fibrosis by blocking autophagy flux. The findings suggest that LDHA could potentially serve as a promising target for the treatment of cardiac fibrosis.


Subject(s)
Autophagy , Fibrosis , Nicotine , Animals , Autophagy/drug effects , Rats , Male , Rats, Inbred SHR , Signal Transduction/drug effects , Myocardium/pathology , Myocardium/metabolism , Lactate Dehydrogenase 5/metabolism , Cells, Cultured , Humans , Fibroblasts/drug effects , Fibroblasts/metabolism , TOR Serine-Threonine Kinases/metabolism , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Rats, Sprague-Dawley
11.
Wound Repair Regen ; 2024 May 19.
Article in English | MEDLINE | ID: mdl-38764180

ABSTRACT

Hyperplastic scar (HS) is an overreaction of tissue to skin injury caused by local fibroblast proliferation and excessive collagen production. Histone posttranslational modification patterns are important epigenetic processes that control various biological activities. This study was designed to investigate the effects of histone lactylation on HS and the underlying mechanism. Western blot was used to analyse the lactylation level in HS patients and fibroblasts (HSFs). In vitro experiments, western blot, cell counting kit-8, and immunofluorescence staining were performed to detect the collagen level, cell viability, and autophagy, respectively. The relationship between snai2 (SLUG) and phosphatase and tensin homologue (PTEN) was assessed by RNA immunoprecipitation and dual-luciferase reporter assays. The results showed that the histone lactylation level was upregulated in HS tissues and HSFs. HSFs showed increased collagen production and cell viability, and decreased autophagy. Silencing of lactate dehydrogenase A (LDHA) promoted the transcription of PTEN by inhibiting SLUG, thus promoting autophagy. Knockdown of LDHA inhibited collagen deposition and cell viability, and increased autophagy in HSFs, and the results were reversed after PTEN inhibition. In summary, histone lactylation inhibited the transcription activity of PTEN by promoting SLUG, thereby suppressing autophagy and promoting collagen deposition and cell viability of HSFs, which might provide effective therapeutic strategies in HS.

12.
Free Radic Biol Med ; 220: 312-323, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38740101

ABSTRACT

Podocytes are crucial for regulating glomerular permeability. They have foot processes that are integral to the renal filtration barrier. Understanding their energy metabolism could shed light on the pathogenesis of filtration barrier injury. Lactate has been increasingly recognized as more than a waste product and has emerged as a significant metabolic fuel and reserve. The recent identification of lactate transporters in podocytes, the expression of which is modulated by glucose levels and lactate, highlights lactate's relevance. The present study investigated the impact of lactate on podocyte respiratory efficiency and mitochondrial dynamics. We confirmed lactate oxidation in podocytes, suggesting its role in cellular energy production. Under conditions of glucose deprivation or lactate supplementation, a significant shift was seen toward oxidative phosphorylation, reflected by an increase in the oxygen consumption rate/extracellular acidification rate ratio. Notably, lactate dehydrogenase A (LDHA) and lactate dehydrogenase B (LDHB) isoforms, which are involved in lactate conversion to pyruvate, were detected in podocytes for the first time. The presence of lactate led to higher intracellular pyruvate levels, greater LDH activity, and higher LDHB expression. Furthermore, lactate exposure increased mitochondrial DNA-to-nuclear DNA ratios and resulted in upregulation of the mitochondrial biogenesis markers peroxisome proliferator-activated receptor coactivator-1α and transcription factor A mitochondrial, regardless of glucose availability. Changes in mitochondrial size and shape were observed in lactate-exposed podocytes. These findings suggest that lactate is a pivotal energy source for podocytes, especially during energy fluctuations. Understanding lactate's role in podocyte metabolism could offer insights into renal function and pathologies that involve podocyte injury.


Subject(s)
L-Lactate Dehydrogenase , Lactic Acid , Mitochondrial Dynamics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Podocytes , Podocytes/metabolism , Podocytes/pathology , Animals , Rats , Lactic Acid/metabolism , L-Lactate Dehydrogenase/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Mitochondria/metabolism , Mitochondria/pathology , Glucose/metabolism , Energy Metabolism , Lactate Dehydrogenase 5/metabolism , Oxidative Phosphorylation/drug effects , DNA, Mitochondrial/metabolism , DNA, Mitochondrial/genetics , Oxygen Consumption , Cells, Cultured , Pyruvic Acid/metabolism , Isoenzymes
13.
Int Immunopharmacol ; 134: 112187, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38733825

ABSTRACT

OBJECTIVE: Glioblastoma (GBM) has poor clinical prognosis due to limited treatment options. In addition, the current treatment regimens for GBM may only slightly prolong patient survival. The aim of this study was to assess the role of BMAL1 in the immune microenvironment and drug resistance of GBM. METHODS: GBM cell lines with stable BMAL1 knockdown or LDHA overexpression were constructed, and functionally characterized by the CCK8, EdU incorporation, and transwell assays. In vivo GBM model was established in C57BL/6J mice. Flow cytometry, ELISA, immunofluorescence, and RT-qPCR were performed to detect macrophage polarization. Lactate production, pathological changes, and the expression of glycolytic proteins were analyzed by HE staining, immunohistochemistry, biochemical assays, and Western blotting. RESULTS: BMAL1 silencing inhibited the malignant characteristics, lactate production, and expression of glycolytic proteins in GBM cells, and these changes were abrogated by overexpression of LDHA or exogenous lactate supplementation. Furthermore, BMAL1 knockdown induced M1 polarization of macrophages, and inhibited M2 polarization and angiogenesis in GBM cells in conditioned media. Overexpression of LDHA or presence of exogenous lactate inhibited BMAL1-induced M1 polarization and angiogenesis. Finally, BMAL1 silencing and bevacizumab synergistically inhibited glycolysis, angiogenesis and M2 polarization, and promoted M1 polarization in vivo, thereby suppressing GBM growth. CONCLUSION: BMAL1 silencing can sensitize GBM cells to bevacizumab by promoting M1/M2 polarization through the LDHA/lactate axis.


Subject(s)
ARNTL Transcription Factors , Bevacizumab , Glioblastoma , Lactic Acid , Mice, Inbred C57BL , Animals , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Glioblastoma/metabolism , Glioblastoma/drug therapy , Glioblastoma/genetics , Humans , Cell Line, Tumor , Bevacizumab/therapeutic use , Bevacizumab/pharmacology , Mice , Lactic Acid/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Tumor Microenvironment/drug effects , Macrophages/immunology , Macrophages/drug effects , Macrophages/metabolism , Glycolysis/drug effects , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Agents, Immunological/pharmacology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/drug therapy , Gene Silencing , L-Lactate Dehydrogenase
14.
J Anim Sci Biotechnol ; 15(1): 68, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38725063

ABSTRACT

BACKGROUND: In poultry, the smooth transition of follicles from the preovulatory-to-postovulatory phase impacts egg production in hens and can benefit the poultry industry. However, the regulatory mechanism underlying follicular ovulation in avians is a complex biological process that remains unclear. RESULTS: Critical biochemical events involved in ovulation in domestic chickens (Gallus gallus) were evaluated by transcriptomics, proteomics, and in vitro assays. Comparative transcriptome analyses of the largest preovulatory follicle (F1) and postovulatory follicle (POF1) in continuous laying (CL) and intermittent laying (IL) chickens indicated the greatest difference between CL_F1 and IL_F1, with 950 differentially expressed genes (DEGs), and the smallest difference between CL_POF1 and IL_POF1, with 14 DEGs. Additionally, data-independent acquisition proteomics revealed 252 differentially abundant proteins between CL_F1 and IL_F1. Perivitelline membrane synthesis, steroid biosynthesis, lysosomes, and oxidative phosphorylation were identified as pivotal pathways contributing to ovulation regulation. In particular, the regulation of zona pellucida sperm-binding protein 3, plasminogen activator, cathepsin A, and lactate dehydrogenase A (LDHA) was shown to be essential for ovulation. Furthermore, the inhibition of LDHA decreased cell viability and promoted apoptosis of ovarian follicles in vitro. CONCLUSIONS: This study reveals several important biochemical events involved in the process of ovulation, as well as crucial role of LDHA. These findings improve our understanding of ovulation and its regulatory mechanisms in avian species.

15.
Ecotoxicol Environ Saf ; 279: 116500, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38795416

ABSTRACT

Hexavalent chromium [Cr(VI)] is one of the most common environmental contaminants due to its tremendous industrial applications, but its effects and mechanism remain to be investigated. Our previous studies showed that Cr(VI) exposure caused malignant transformation and tumorigenesis. This study showed that glycolytic proteins HK2 and LDHA levels were statistically significant changed in blood samples of Cr(VI)-exposed workers and in Cr-T cells compared to the control subjects and parental cells. HK2 and LDHA knockdown inhibited cell proliferation and angiogenesis, and higher HK2 and LDHA expression levels are associated with advanced stages and poor prognosis of lung cancer. We found that miR-218 levels were significantly decreased and miR-218 directly targeted HK2 and LDHA for inhibiting their expression. Overexpression of miR-218 inhibited glucose consumption and lactate production in Cr-T cells. Further study found that miR-218 inhibited tumor growth and angiogenesis by decreasing HK2 and LDHA expression in vivo. MiR-218 levels were negatively correlated with HK2 and LDHA expression levels and cancer development in human lung and other cancers. These results demonstrated that miR-218/HK2/LDHA pathway is vital for regulating Cr(VI)-induced carcinogenesis and human cancer development.


Subject(s)
Carcinogenesis , Chromium , Hexokinase , Lung Neoplasms , MicroRNAs , Up-Regulation , MicroRNAs/genetics , Humans , Chromium/toxicity , Hexokinase/genetics , Hexokinase/metabolism , Carcinogenesis/chemically induced , Lung Neoplasms/chemically induced , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Prognosis , Animals , Cell Proliferation/drug effects , L-Lactate Dehydrogenase/metabolism , Occupational Exposure/adverse effects , Mice , Isoenzymes
16.
Bone ; 185: 117132, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38789096

ABSTRACT

The mechanosensitivity of inflammation can alter cellular mechanotransduction. However, the underlying mechanism remains unclear. This study aims to investigate the metabolic mechanism of inflammation under mechanical force to guide tissue remodeling better. Herein, we found that inflammation hindered bone remodeling under mechanical force, accompanied by a simultaneous enhancement of oxidative phosphorylation (OXPHOS) and glycolysis. The control of metabolism direction through GNE-140 and Visomitin revealed that enhanced glycolysis might act as a compensatory mechanism to resist OXPHOS-induced osteoclastogenesis by promoting osteogenesis. The inhibited osteogenesis induced by inflammatory mechanical stimuli was concomitant with a reduced expression of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α). PGC-1α knockdown impeded osteogenesis under mechanical force and facilitated osteoclastogenesis by enhancing OXPHOS. Conversely, PGC-1α overexpression attenuated the impairment of bone remodeling by inflammatory mechanical signals through promoting glycolysis. This process benefited from the PGC-1α regulation on the transcriptional and translational activity of lactate dehydrogenase A (LDHA) and the tight control of the extracellular acidic environment. Additionally, the increased binding between PGC-1α and LDHA proteins might contribute to the glycolysis promotion within the inflammatory mechanical environment. Notably, LDHA suppression effectively eliminated the bone repair effect mediated by PGC-1α overexpression within inflammatory mechanical environments. In conclusion, this study demonstrated a novel molecular mechanism illustrating how inflammation orchestrated glucose metabolism through glycolysis and OXPHOS to affect mechanically induced bone remodeling.


Subject(s)
Bone Remodeling , Glycolysis , Inflammation , Osteogenesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Signal Transduction , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Animals , Bone Remodeling/physiology , Inflammation/metabolism , Inflammation/pathology , Osteogenesis/physiology , Mice , Mice, Inbred C57BL , L-Lactate Dehydrogenase/metabolism , Oxidative Phosphorylation , Cellular Microenvironment , Male
17.
Aging (Albany NY) ; 16(9): 8000-8018, 2024 05 03.
Article in English | MEDLINE | ID: mdl-38709280

ABSTRACT

Lactate dehydrogenase A (LDHA), a critical enzyme involved in glycolysis, is broadly involved multiple biological functions in human cancers. It is reported that LDHA can impact tumor immune surveillance and induce the transformation of tumor-associated macrophages, highlighting its unnoticed function of LDHA in immune system. However, in human cancers, the role of LDHA in prognosis and immunotherapy hasn't been investigated. In this study, we analyzed the expression pattern and prognostic value of LDHA in pan-cancer and explored its association between tumor microenvironment (TME), immune infiltration subtype, stemness scores, tumor mutation burden (TMB), and immunotherapy resistance. We found that LDHA expression is tumor heterogeneous and that its high expression is associated with poor prognosis in multiple human cancers. In addition, LDHA expression was positively correlated with the presence of mononuclear/macrophage cells, and also promoted the infiltration of a range of immune cells. Genomic alteration of LDHA was common in different types of cancer, while with prognostic value in pan-cancers. Pan-cancer analysis revealed that the significant correlations existed between LDHA expression and tumor microenvironment (including stromal cells and immune cells) as well as stemness scores (DNAss and RNAss) across cancer types. Drug sensitivity analysis also revealed that LDHA was able to predict response to chemotherapy and immunotherapy. Furthermore, it was confirmed that knockdown of LDHA reduced proliferation and migration ability of lung cancer cells. Taken together, LDHA could serve as a prognostic biomarker and a potential immunotherapy marker.


Subject(s)
Drug Resistance, Neoplasm , Immunotherapy , Neoplasms , Tumor Microenvironment , Humans , Tumor Microenvironment/immunology , Prognosis , Neoplasms/immunology , Neoplasms/genetics , Neoplasms/therapy , Drug Resistance, Neoplasm/genetics , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , L-Lactate Dehydrogenase/metabolism , L-Lactate Dehydrogenase/genetics , Cell Line, Tumor
18.
Front Biosci (Landmark Ed) ; 29(4): 166, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38682188

ABSTRACT

OBJECTIVE: The aims of this study were to evaluate the correlation between chemokine (C-X-C) ligand 7 (CXCL7) expression and glycolysis and to explore the prognostic significance of CXCL7 in colorectal cancer (CRC). METHODS: The expression of CXCL7 and lactate dehydrogenase A (LDH-A) was measured by immunohistochemistry in tissue from 158 CRC patients. Patients were divided into high expression and low expression groups based on receiver operating characteristic curves and a cut-off value. The correlation between CXCL7 and LDH-A expression was evaluated. The overall survival (OS) times of CRC patients were explored. The risk factors related to prognosis were assessed. RESULTS: Significantly higher expression of CXCL7 and LDH-A was detected in CRC tissue than in non-CRC tissue, and was associated with N stage and tumor-node-metastasis (TNM) stage. CXCL7 expression was strongly correlated with LDH-A expression in CRC tissue. High expression of CXCL7 was validated as an independent risk factor for OS. CONCLUSION: Increased expression of CXCL7 was positively correlated with LDH-A expression and was an independent risk factor for CRC prognosis.


Subject(s)
Biomarkers, Tumor , Colorectal Neoplasms , L-Lactate Dehydrogenase , Adult , Aged , Female , Humans , Male , Middle Aged , Biomarkers, Tumor/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/mortality , Colorectal Neoplasms/genetics , Immunohistochemistry , L-Lactate Dehydrogenase/metabolism , Prognosis , Risk Factors
19.
Cancer Lett ; 590: 216869, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38593918

ABSTRACT

Lysine acetyltransferase 7 (KAT7), a histone acetyltransferase, has recently been identified as an oncoprotein and has been implicated in the development of various malignancies. However, its specific role in head and neck squamous carcinoma (HNSCC) has not been fully elucidated. Our study revealed that high expression of KAT7 in HNSCC patients is associated with poor survival prognosis and silencing KAT7 inhibits the Warburg effect, leading to reduced proliferation, invasion, and metastatic potential of HNSCC. Further investigation uncovered a link between the high expression of KAT7 in HNSCC and tumor-specific glycolytic metabolism. Notably, KAT7 positively regulates Lactate dehydrogenase A (LDHA), a key enzyme in metabolism, to promote lactate production and create a conducive environment for tumor proliferation and metastasis. Additionally, KAT7 enhances LDHA activity and upregulates LDHA protein expression by acetylating the lysine 118 site of LDHA. Treatment with WM3835, a KAT7 inhibitor, effectively suppressed the growth of subcutaneously implanted HNSCC cells in mice. In conclusion, our findings suggest that KAT7 exerts pro-cancer effects in HNSCC by acetylating LDHA and may serve as a potential therapeutic target. Inhibiting KAT7 or LDHA expression holds promise as a therapeutic strategy to suppress the growth and progression of HNSCC.


Subject(s)
Cell Proliferation , Head and Neck Neoplasms , Histone Acetyltransferases , Squamous Cell Carcinoma of Head and Neck , Humans , Animals , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism , Acetylation , Cell Line, Tumor , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/genetics , Mice , L-Lactate Dehydrogenase/metabolism , L-Lactate Dehydrogenase/genetics , Lysine Acetyltransferases/metabolism , Lysine Acetyltransferases/genetics , Gene Expression Regulation, Neoplastic , Mice, Nude , Warburg Effect, Oncologic , Male , Female , Cell Movement , Xenograft Model Antitumor Assays , Neoplasm Invasiveness , Isoenzymes/metabolism , Isoenzymes/genetics
20.
Glia ; 72(8): 1374-1391, 2024 08.
Article in English | MEDLINE | ID: mdl-38587131

ABSTRACT

Oligodendrocytes and astrocytes are metabolically coupled to neuronal compartments. Pyruvate and lactate can shuttle between glial cells and axons via monocarboxylate transporters. However, lactate can only be synthesized or used in metabolic reactions with the help of lactate dehydrogenase (LDH), a tetramer of LDHA and LDHB subunits in varying compositions. Here we show that mice with a cell type-specific disruption of both Ldha and Ldhb genes in oligodendrocytes lack a pathological phenotype that would be indicative of oligodendroglial dysfunctions or lack of axonal metabolic support. Indeed, when combining immunohistochemical, electron microscopical, and in situ hybridization analyses in adult mice, we found that the vast majority of mature oligodendrocytes lack detectable expression of LDH. Even in neurodegenerative disease models and in mice under metabolic stress LDH was not increased. In contrast, at early development and in the remyelinating brain, LDHA was readily detectable in immature oligodendrocytes. Interestingly, by immunoelectron microscopy LDHA was particularly enriched at gap junctions formed between adjacent astrocytes and at junctions between astrocytes and oligodendrocytes. Our data suggest that oligodendrocytes metabolize lactate during development and remyelination. In contrast, for metabolic support of axons mature oligodendrocytes may export their own glycolysis products as pyruvate rather than lactate. Lacking LDH, these oligodendrocytes can also "funnel" lactate through their "myelinic" channels between gap junction-coupled astrocytes and axons without metabolizing it. We suggest a working model, in which the unequal cellular distribution of LDH in white matter tracts facilitates a rapid and efficient transport of glycolysis products among glial and axonal compartments.


Subject(s)
Axons , Glycolysis , L-Lactate Dehydrogenase , Oligodendroglia , Animals , Oligodendroglia/metabolism , Axons/metabolism , L-Lactate Dehydrogenase/metabolism , L-Lactate Dehydrogenase/genetics , Glycolysis/physiology , Mice , Down-Regulation/physiology , Mice, Inbred C57BL , Lactate Dehydrogenase 5/metabolism , Astrocytes/metabolism , Astrocytes/ultrastructure , Mice, Transgenic , Isoenzymes/metabolism , Isoenzymes/genetics , Gap Junctions/metabolism , Gap Junctions/ultrastructure , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL