380 results on '"Cpt1a"'
Search Results
2. TRF‐16 Inhibits Lung Cancer Progression by Hindering the N6‐Methyladenosine Modification of CPT1A mRNA.
- Author
-
Ye, Jiankui, Chen, Yu, Shao, Zhuowei, Wu, Yili, Li, You, Fang, Shuai, and Wu, Shibo
- Subjects
CANCER cell proliferation ,NON-coding RNA ,LUNG cancer ,GENE expression ,CELL physiology - Abstract
Transfer RNA‐derived fragments (tRFs) are a new class of small non‐coding RNAs. Recent studies suggest that tRFs participate in some pathological processes. However, the biological activities and processes of tRFs in lung cancer cells remain mainly unclear. In the present investigation, we employed tRNA‐derived small RNA (tsRNA) sequencing to predict differentially expressed tsRNAs in lung cancer cells, and nine tsRNAs with significant expression alterations were validated using qPCR. Wound healing, colony formation, transwell invasion and CCK‐8 assays were performed to detect the effects of tRF‐16 on cell function. Western blotting evaluated the relationship between tRF‐16 and the IGF2BP1 axis. Our findings demonstrated that tRF‐16 expression was substantially downregulated in lung cancer cells. TRF‐16 could inhibit lung cancer cells' ability to increase, migrate, invade and obtain radiation resistance. Furthermore, tRF‐16 decreases the stability of CPT1A by impairing the binding of IGF2BP1 to CPT1A. As a result, the fatty acid metabolism in lung cancer cells was inhibited. Finally, tRF‐16 also inhibits lung cancer cell proliferation in vivo. This study shows that tRF‐16 plays a crucial regulatory role in the proliferation of lung cancer cells and may represent a novel avenue for their regulation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. Overexpression of CPT1A disrupts the maintenance and regenerative function of muscle stem cells.
- Author
-
Qiu, Jiamin, Yue, Feng, Kim, Kun Ho, Chen, Xiyue, Khedr, Mennatallah A., Chen, Jingjuan, Gu, Lijie, Ren, Junxiao, Ferreira, Christina R., Ellis, Jessica, and Kuang, Shihuan
- Abstract
The skeletal muscle satellite cells (SCs) mediate regeneration of myofibers upon injury. As they switch from maintenance (quiescence) to regeneration, their relative reliance on glucose and fatty acid metabolism alters. To explore the contribution of mitochondrial fatty acid oxidation (FAO) pathway to SCs and myogenesis, we examined the role of carnitine palmitoyltransferase 1A (CPT1A), the rate‐limiting enzyme of FAO. CPT1A is highly expressed in quiescent SCs (QSCs) compared with activated and proliferating SCs, and its expression level decreases during myogenic differentiation. Myod1Cre‐driven overexpression (OE) of Cpt1a in embryonic myoblasts (Cpt1aMTG) reduces muscle weight, grip strength, and contractile force without affecting treadmill endurance of adult mice. Adult Cpt1aMTG mice have reduced number of SC, impairing muscle regeneration and promoting lipid infiltration. Similarly, Pax7CreER‐driven, tamoxifen‐inducible Cpt1a‐OE in QSCs of adult muscles (Cpt1aPTG) leads to depletion of SCs and compromises muscle regeneration. The reduced proliferation of Cpt1a‐OE SCs is associated with elevated level of acyl‐carnitine, and acyl‐carnitine treatment impedes proliferation of wildtype SCs. These findings indicate that aberrant level of CPT1A elevates acyl‐carnitine to impair the maintenance, proliferation and regenerative function of SCs. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
4. Oral Microbiome and CPT1A Function in Fatty Acid Metabolism in Oral Cancer.
- Author
-
Praveen, Zeba, Choi, Sung-Weon, Lee, Jong Ho, Park, Joo Yong, Oh, Hyun Jun, Kwon, Ik Jae, Park, Jin Hee, and Kim, Mi Kyung
- Subjects
- *
TUMOR necrosis factors , *SHORT-chain fatty acids , *FATTY acid oxidation , *CARNITINE palmitoyltransferase , *ORAL cancer , *ACYLTRANSFERASES - Abstract
The oral microbiome is crucial for human health. Although oral dysbiosis may contribute to oral cancer (OC), the detailed relationships between the microbiome and OC remain unclear. In this case-control study, we aimed to elucidate the connection between the oral microbiome and mechanisms potentially involved in oral cancer. The study analyzed 1022 oral saliva samples, including 157 from oral cancer patients and 865 from healthy controls, using 16S ribosomal RNA (16S rRNA) sequencing and a Light Gradient Boosting Machine (LightGBM) model to identify four bacterial genera significantly associated with oral cancer. In patients with oral cancer, the relative abundance of Streptococcus and Parvimonas was higher; Corynebacterium and Prevotella showed decreased relative abundance; and levels of fatty acid oxidation enzymes, including Carnitine palmitoyltransferase 1A (CPT1A), long-chain acyl-CoA synthetase, acyl-CoA dehydrogenase, diacylglycerol choline phosphotransferase, and H+-transporting ATPase, were significantly higher compared to controls. Conversely, healthy controls exhibited increased levels of short-chain fatty acids (SCFAs) and CD4+T-helper cell counts. Survival analysis revealed that higher abundance of Streptococcus and Parvimonas, which correlated positively with interleukin-6, tumor necrosis factor-alpha, and CPT1A, were linked to poorer disease-free survival (DFS) and overall survival (OS) rates, while Prevotella and Corynebacterium were associated with better outcomes. These findings suggest that changes in these bacterial genera are associated with alterations in specific cytokines, CPT1A levels, SCFAs in oral cancer, with lower SCFA levels in patients reinforcing this link. Overall, these microbiome changes, along with cytokine and enzyme alterations, may serve as predictive markers, enhancing diagnostic accuracy for oral cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
5. CircHIPK3/miR-124 affects angiogenesis in early-onset preeclampsia via CPT1A-mediated fatty acid oxidation.
- Author
-
Wu, Yanying, Huang, Jingrui, Liu, Lijuan, Zhang, Xiaowen, Zhang, Weishe, and Li, Qi
- Subjects
- *
FATTY acid oxidation , *PREECLAMPSIA , *NEOVASCULARIZATION , *CARNITINE palmitoyltransferase , *ENDOTHELIAL cells - Abstract
Multiple theories have been proposed to explain the pathogenesis of early-onset preeclampsia (EOPE), and angiogenic dysfunction is an important part of this pathogenesis. Carnitine palmitoyltransferase (CPT1A) is a key rate-limiting enzyme in the metabolic process of fatty acid oxidation (FAO). FAO regulates endothelial cell (EC) proliferation during vascular germination and is also essential for ab initio deoxyribonucleotide synthesis, but its role in EOPE needs to be further elucidated. In the present study, we investigated its functional role in EOPE by targeting the circHIPK3/miR-124-3p/CPT1A axis. In our study, reduced expression of circHIPK3 and CPT1A and increased expression of miR-124-3p in placental tissues from patients with EOPE were associated with EC dysfunction. Here, we confirmed that CPT1A regulates fatty acid oxidative activity, cell proliferation, and tube formation in ECs by regulating FAO. Functionally, knockdown of circHIPK3 suppressed EC angiogenesis by inhibiting CPT1A-mediated fatty acid oxidative activity, which was ameliorated by CPT1A overexpression. In addition, circHIPK3 regulates CPT1A expression by sponging miR-124-3p. Hence, circHIPK3 knockdown reduced fatty acid oxidation in ECs by sponging miR-124-3p in a CPT1A-dependent manner and inhibited EC proliferation and tube formation, which may have led to aberrant angiogenesis in EOPE. Thus, strategies targeting CPT1A-driven FAO may be promising approaches for the treatment of EOPE. Key messages: Decreased Carnitine palmitoyltransferase (CPT1A) expression in preeclampsia(PE). CPT1A overexpression promotes FAO activity and tube formation in ECs. CircHIPK3 can affect CPT1A expression and impaire angiogenesis of EOPE. CircHIPK3 regulates CPT1A expression by acting as a ceRNA of miR-124-3p in HUVECs. Confirming the effect of circHIPK3/miR-124-3p/CPT1A axis on EOPE. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. CPT1A mediates the succinylation of SP5 which activates transcription of PDPK1 to promote the viability and glycolysis of prostate cancer cells
- Author
-
Shufeng Liu, Xiaoguang Chen, Liqi Zhang, and Bo Lu
- Subjects
Prostate cancer ,CPT1A ,SP5 ,PDPK1 ,succinylation ,transcription ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Succinylation modification involves in the progression of human cancers. The present study aimed to investigate the role of CPT1A, which is a succinyltransferase in the progression of prostate cancer (PCa). CCK-8 was used to detect the cell viability. Seahorse was performed to evaluate the cell glycolysis. Luciferase assay was used to detect the transcriptional regulation. ChIP was performed to assess the binding between transcriptional factors with the promoters. Co-IP was used to assess the binding between proteins. We found that CPT1A was highly expressed in PCa tissues and cell lines. Silencing of CPT1A inhibited the viability and glycolysis of PCa cells. Mechanistically, CPT1A promoted the succinylation of SP5, which strengthened the binding between SP5 and the promoter of PDPK1. SP5 activated PDPK1 transcription and PDPK1 activated the AKT/mTOR signal pathway. These findings might provide novel targets for the diagnosis or therapy of prostate cancer.
- Published
- 2024
- Full Text
- View/download PDF
7. CPT1A mediates radiation sensitivity in colorectal cancer
- Author
-
Zhenhui Chen, Lu Yu, Zhihao Zheng, Xusheng Wang, Qiqing Guo, Yuchuan Chen, Yaowei Zhang, Yuqin Zhang, Jianbiao Xiao, Keli Chen, Hongying Fan, and Yi Ding
- Subjects
human ,mouse ,colorectal cancer cells ,CPT1A ,reactive oxygen species ,radiosensitivity ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
The prevalence and mortality rates of colorectal cancer (CRC) are increasing worldwide. Radiation resistance hinders radiotherapy, a standard treatment for advanced CRC, leading to local recurrence and metastasis. Elucidating the molecular mechanisms underlying radioresistance in CRC is critical to enhance therapeutic efficacy and patient outcomes. Bioinformatic analysis and tumour tissue examination were conducted to investigate the CPT1A mRNA and protein levels in CRC and their correlation with radiotherapy efficacy. Furthermore, lentiviral overexpression and CRISPR/Cas9 lentiviral vectors, along with in vitro and in vivo radiation experiments, were used to explore the effect of CPT1A on radiosensitivity. Additionally, transcriptomic sequencing, molecular biology experiments, and bioinformatic analyses were employed to elucidate the molecular mechanisms by which CPT1A regulates radiosensitivity. CPT1A was significantly downregulated in CRC and negatively correlated with responsiveness to neoadjuvant radiotherapy. Functional studies suggested that CPT1A mediates radiosensitivity, influencing reactive oxygen species (ROS) scavenging and DNA damage response. Transcriptomic and molecular analyses highlighted the involvement of the peroxisomal pathway. Mechanistic exploration revealed that CPT1A downregulates the FOXM1-SOD1/SOD2/CAT axis, moderating cellular ROS levels after irradiation and enhancing radiosensitivity. CPT1A downregulation contributes to radioresistance in CRC by augmenting the FOXM1-mediated antioxidant response. Thus, CPT1A is a potential biomarker of radiosensitivity and a novel target for overcoming radioresistance, offering a future direction to enhance CRC radiotherapy.
- Published
- 2024
- Full Text
- View/download PDF
8. Rotenone adaptation promotes migration and invasion of p53-wild-type colon cancer through lipid metabolism
- Author
-
Shi, Yingying, Cao, Zhen, Ge, Ling, Lei, Lin, Tao, Dan, Zhong, Juan, Xu, Dan, Geng, Tao, Li, Xuetao, Li, Ziwei, Xing, Shuaishuai, Wu, Xinyu, Wang, Zhongxu, and Li, Linjun
- Published
- 2024
- Full Text
- View/download PDF
9. TRIM2 promotes metabolic adaptation to glutamine deprivation via enhancement of CPT1A activity.
- Author
-
Liao, Kaimin, Liu, Kaiyue, Wang, Zhongyu, Zhao, Kailiang, and Mei, Yide
- Subjects
- *
SLEEP deprivation , *GLUTAMINE , *TRANSCRIPTION factors , *FATTY acid oxidation , *GLUTAMINE synthetase , *CANCER cells , *CYCLIC-AMP-dependent protein kinase , *TUMOR growth - Abstract
Cancer cells undergo metabolic adaptation to promote their survival and growth under energy stress conditions, yet the underlying mechanisms remain largely unclear. Here, we report that tripartite motif‐containing protein 2 (TRIM2) is upregulated in response to glutamine deprivation by the transcription factor cyclic AMP‐dependent transcription factor (ATF4). TRIM2 is shown to specifically interact with carnitine O‐palmitoyltransferase 1 (CPT1A), a rate‐limiting enzyme of fatty acid oxidation. Via this interaction, TRIM2 enhances the enzymatic activity of CPT1A, thereby regulating intracellular lipid levels and protecting cells from glutamine deprivation‐induced apoptosis. Furthermore, TRIM2 is able to promote both in vitro cell proliferation and in vivo xenograft tumor growth via CPT1A. Together, these findings establish TRIM2 as an important regulator of the metabolic adaptation of cancer cells to glutamine deprivation and implicate TRIM2 as a potential therapeutic target for cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. Microfluidic production of amiodarone loaded nanoparticles and application in drug repositioning in ovarian cancer
- Author
-
Asia Saorin, Gloria Saorin, Fahriye Duzagac, Pietro Parisse, Ni Cao, Giuseppe Corona, Enrico Cavarzerani, and Flavio Rizzolio
- Subjects
Ovarian cancer ,CPT1A ,Amiodarone ,Microfluidics ,Lipidic nanoparticles ,Medicine ,Science - Abstract
Abstract Amiodarone repositioning in cancer treatment is promising, however toxicity limits seem to arise, constraining its exploitability. Notably, amiodarone has been investigated for the treatment of ovarian cancer, a tumour known for metastasizing within the peritoneal cavity. This is associated with an increase of fatty acid oxidation, which strongly depends on CPT1A, a transport protein which has been found overexpressed in ovarian cancer. Amiodarone is an inhibitor of CPT1A but its role still has to be explored. Therefore, in the present study, amiodarone was tested on ovarian cancer cell lines with a focus on lipid alteration, confirming its activity. Moreover, considering that drug delivery systems could lower drug side effects, microfluidics was employed for the development of drug delivery systems of amiodarone obtaining simultaneously liposomes with a high payload and amiodarone particles. Prior to amiodarone loading, microfluidics production was optimized in term of temperature and flow rate ratio. Moreover, stability over time of particles was evaluated. In vitro tests confirmed the efficacy of the drug delivery systems.
- Published
- 2024
- Full Text
- View/download PDF
11. Dysregulated lipid metabolism is associated with kidney allograft fibrosis
- Author
-
Linjie Peng, Chang Wang, Shuangjin Yu, Qihao Li, Guobin Wu, Weijie Lai, Jianliang Min, and Guodong Chen
- Subjects
Lipid metabolism ,CPT1A ,Kidney allograft ,Fibrosis ,IF/TA ,Nutritional diseases. Deficiency diseases ,RC620-627 - Abstract
Abstract Background Interstitial fibrosis and tubular atrophy (IF/TA), a histologic feature of kidney allograft destruction, is linked to decreased allograft survival. The role of lipid metabolism is well-acknowledged in the area of chronic kidney diseases; however, its role in kidney allograft fibrosis is still unclarified. In this study, how lipid metabolism contributes to kidney allografts fibrosis was examined. Methods A comprehensive bioinformatic comparison between IF/TA and normal kidney allograft in the Gene Expression Omnibus (GEO) database was conducted. Further validations through transcriptome profiling or pathological staining of human recipient biopsy samples and in rat models of kidney transplantation were performed. Additionally, the effects of enhanced lipid metabolism on changes in the fibrotic phenotype induced by TGF-β1 were examined in HK-2 cell. Results In-depth analysis of the GEO dataset revealed a notable downregulation of lipid metabolism pathways in human kidney allografts with IF/TA. This decrease was associated with increased level of allograft rejection, inflammatory responses, and epithelial mesenchymal transition (EMT). Pathway enrichment analysis showed the downregulation in mitochondrial LC-fatty acid beta-oxidation, fatty acid beta-oxidation (FAO), and fatty acid biosynthesis. Dysregulated fatty acid metabolism was also observed in biopsy samples from human kidney transplants and in fibrotic rat kidney allografts. Notably, the areas affected by IF/TA had increased immune cell infiltration, during which increased EMT biomarkers and reduced CPT1A expression, a key FAO enzyme, were shown by immunohistochemistry. Moreover, under TGF-β1 induction, activating CPT1A with the compound C75 effectively inhibited migration and EMT process in HK-2 cells. Conclusions This study reveal a critical correlation between dysregulated lipid metabolism and kidney allograft fibrosis. Enhancing lipid metabolism with CPT1A agonists could be a therapeutic approach to mitigate kidney allografts fibrosis.
- Published
- 2024
- Full Text
- View/download PDF
12. HMGB1 Modulates Macrophage Metabolism and Polarization in Ulcerative Colitis by Inhibiting Cpt1a Expression
- Author
-
Fenfen Wang, Linfei Luo, Zhengqiang Wu, Lijun Wan, Fan Li, and Zhili Wen
- Subjects
ulcerative colitis ,hmgb1 ,macrophage polarization ,cell metabolism ,cpt1a ,Biochemistry ,QD415-436 ,Biology (General) ,QH301-705.5 - Abstract
Background: Macrophage polarization is involved in the development of ulcerative colitis (UC). This study investigated the mechanism by which high mobility group box-1 protein (HMGB1) regulates macrophage polarization through metabolic reprogramming, thereby contributing to the pathogenesis of UC. Methods: Dextran sulfate sodium (DSS) was used to induce colitis in mice. RAW264.7 cells were polarized to M1 or M2 macrophages in vitro by stimulating with lipopolysaccharide (LPS)/interferon-γ (IFN-γ) or Interleukin-4 (IL-4), respectively. Macrophage infiltration and distribution within colon tissue were assessed by immunohistochemistry and flow cytometry. Glycolysis, fatty acid oxidation (FAO), and inflammatory factors were evaluated using relevant reagent kits. Chromatin Immunoprecipitation (ChIP) and luciferase reporter experiments were performed to study the regulation of Carnitine palmitoyltransferase 1A (Cpt1a) promoter transcriptional activity by HMGB1. Results: The mouse UC model showed upregulated HMGB1 and increased macrophage infiltration. Overexpression of HMGB1 promoted M1 macrophage polarization, increased glycolysis, and reduced FAO, whereas knockdown of HMGB1 promoted M2 macrophage polarization, reduced glycolysis, and increased FAO. HMGB1 negatively regulated Cpt1a expression by inhibiting transcription of the Cpt1a promoter. Knockdown of Cpt1a reversed the effects of small interfering RNA targeting HMGB1 (si-HMGB1) on macrophage metabolism and polarization. Administration of adeno-associated virus (AAV)-shHMGB1 in vivo caused a reduction in UC symptoms and inflammation. Conclusions: HMGB1 modulates macrophage metabolism in UC by inhibiting Cpt1a expression, leading to increased M1 polarization. This provides a theoretical basis for the clinical application of HMGB1 inhibitors in the treatment of UC.
- Published
- 2024
- Full Text
- View/download PDF
13. Microfluidic production of amiodarone loaded nanoparticles and application in drug repositioning in ovarian cancer
- Author
-
Saorin, Asia, Saorin, Gloria, Duzagac, Fahriye, Parisse, Pietro, Cao, Ni, Corona, Giuseppe, Cavarzerani, Enrico, and Rizzolio, Flavio
- Published
- 2024
- Full Text
- View/download PDF
14. Dysregulated lipid metabolism is associated with kidney allograft fibrosis
- Author
-
Peng, Linjie, Wang, Chang, Yu, Shuangjin, Li, Qihao, Wu, Guobin, Lai, Weijie, Min, Jianliang, and Chen, Guodong
- Published
- 2024
- Full Text
- View/download PDF
15. Immunometabolic features of natural killer cells are associated with infection outcomes in critical illness.
- Author
-
Kuei-Pin Chung, Jia-Ying Su, Yi-Fu Wang, Budiarto, Bugi Ratno, Yu-Chang Yeh, Jui-Chen Cheng, Li-Ta Keng, Yi-Jung Chen, Ya-Ting Lu, Yi-Hsiu Juan, Kiichi Nakahira, Sheng-Yuan Ruan, Jung-Yien Chien, Hou-Tai Chang, Jih-Shuin Jerng, Yen-Tsung Huang, Shih-Yu Chen, and Chong-Jen Yu
- Subjects
KILLER cells ,CRITICALLY ill ,NOSOCOMIAL infections ,MITOCHONDRIA formation ,FATTY acid oxidation - Abstract
Immunosuppression increases the risk of nosocomial infection in patients with chronic critical illness. This exploratory study aimed to determine the immunometabolic signature associated with nosocomial infection during chronic critical illness. We prospectively recruited patients who were admitted to the respiratory care center and who had received mechanical ventilator support for more than 10 days in the intensive care unit. The study subjects were followed for the occurrence of nosocomial infection until 6 weeks after admission, hospital discharge, or death. The cytokine levels in the plasma samples were measured. Single-cell immunometabolic regulome profiling by mass cytometry, which analyzed 16 metabolic regulators in 21 immune subsets, was performed to identify immunometabolic features associated with the risk of nosocomial infection. During the study period, 37 patients were enrolled, and 16 patients (43.2%) developed nosocomial infection. Unsupervised immunologic clustering using multidimensional scaling and logistic regression analyses revealed that expression of nuclear respiratory factor 1 (NRF1) and carnitine palmitoyltransferase 1a (CPT1a), key regulators of mitochondrial biogenesis and fatty acid transport, respectively, in natural killer (NK) cells was significantly associated with nosocomial infection. Downregulated NRF1 and upregulated CPT1a were found in all subsets of NK cells from patients who developed a nosocomial infection. The risk of nosocomial infection is significantly correlated with the predictive score developed by selecting NK cell-specific features using an elastic net algorithm. Findings were further examined in an independent cohort of COVID-19-infected patients, and the results confirm that COVID-19-related mortality is significantly associated with mitochondria biogenesis and fatty acid oxidation pathways in NK cells. In conclusion, this study uncovers that NK cell-specific immunometabolic features are significantly associated with the occurrence and fatal outcomes of infection in critically ill population, and provides mechanistic insights into NK cell-specific immunity against microbial invasion in critical illness. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
16. Cpt1a silencing in AgRP neurons improves cognitive and physical capacity and promotes healthy aging in male mice.
- Author
-
Ibeas, Kevin, Griñán‐Ferré, Christian, del Mar Romero, Maria, Sebastián, David, Bastías‐Pérez, Marianela, Gómez, Roberto, Soler‐Vázquez, M. Carmen, Zagmutt, Sebastián, Pallás, Mercè, Castell, Margarida, Belsham, Denise D., Mera, Paula, Herrero, Laura, and Serra, Dolors
- Subjects
- *
HYPOTHALAMUS , *FATTY acid oxidation , *CARNITINE palmitoyltransferase , *NEURONS , *NEUROPEPTIDE Y , *MICE - Abstract
Orexigenic neurons expressing agouti‐related protein (AgRP) and neuropeptide Y in the arcuate nucleus (ARC) of the hypothalamus are activated in response to dynamic variations in the metabolic state, including exercise. We previously observed that carnitine palmitoyltransferase 1a (CPT1A), a rate‐limiting enzyme of mitochondrial fatty acid oxidation, is a key factor in AgRP neurons, modulating whole‐body energy balance and fluid homeostasis. However, the effect of CPT1A in AgRP neurons in aged mice and during exercise has not been explored yet. We have evaluated the physical and cognitive capacity of adult and aged mutant male mice lacking Cpt1a in AgRP neurons (Cpt1a KO). Adult Cpt1a KO male mice exhibited enhanced endurance performance, motor coordination, locomotion, and exploration compared with control mice. No changes were observed in anxiety‐related behavior, cognition, and muscle strength. Adult Cpt1a KO mice showed a reduction in gastrocnemius and tibialis anterior muscle mass. The cross‐sectional area (CSA) of these muscles were smaller than those of control mice displaying a myofiber remodeling from type II to type I fibers. In aged mice, changes in myofiber remodeling were maintained in Cpt1a KO mice, avoiding loss of physical capacity during aging progression. Additionally, aged Cpt1a KO mice revealed better cognitive skills, reduced inflammation, and oxidative stress in the hypothalamus and hippocampus. In conclusion, CPT1A in AgRP neurons appears to modulate health and protects against aging. Future studies are required to clarify whether CPT1A is a potential antiaging candidate for treating diseases affecting memory and physical activity. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
17. Gestational α-ketoglutarate supplementation ameliorates arsenic-induced hepatic lipid deposition via epigenetic reprogramming of β-oxidation process in female offspring
- Author
-
Qing-Hua Qian, Ya-Ping Song, Yu Zhang, Hao Xue, Wei-Wei Zhang, Yapeng Han, Yán Wāng, and De-Xiang Xu
- Subjects
Arsenite ,Developmental toxicology ,Epigenetic modifications ,Hydroxymethylation ,β-oxidation ,CPT1A ,Environmental sciences ,GE1-350 - Abstract
Inorganic trivalent arsenic (iAsⅢ) at environmentally relevant levels has been found to cause developmental toxicity. Maternal exposure to iAsⅢ leads to enduring hepatic lipid deposition in later adult life. However, the exact mechanism in iAsⅢ induced hepatic developmental hazards is still unclear. In this study, we initially found that gestational exposure to iAsⅢ at an environmentally relevant concentration disturbs lipid metabolism and reduces levels of alpha-ketoglutaric acid (α-KG), an important mitochondrial metabolite during the citric acid cycle, in fetal livers. Further, gestational supplementation of α-KG alleviated hepatic lipid deposition caused by early-life exposure to iAsⅢ. This beneficial effect was particularly pronounced in female offspring. α-KG partially restored the β-oxidation process in hepatic tissues by hydroxymethylation modifications of carnitine palmitoyltransferase 1a (Cpt1a) gene during fetal development. Insufficient β-oxidation capacities probably play a crucial role in hepatic lipid deposition in adulthood following in utero arsenite exposure, which can be efficiently counterbalanced by replenishing α-KG. These results suggest that gestational administration of α-KG can ameliorate hepatic lipid deposition caused by iAsⅢ in female adult offspring partially through epigenetic reprogramming of the β-oxidation pathway. Furthermore, α-KG shows potential as an interventive target to mitigate the harmful effects of arsenic-induced hepatic developmental toxicity.
- Published
- 2024
- Full Text
- View/download PDF
18. CPT1A mediates the succinylation of SP5 which activates transcription of PDPK1 to promote the viability and glycolysis of prostate cancer cells.
- Author
-
Liu, Shufeng, Chen, Xiaoguang, Zhang, Liqi, and Lu, Bo
- Abstract
Succinylation modification involves in the progression of human cancers. The present study aimed to investigate the role of CPT1A, which is a succinyltransferase in the progression of prostate cancer (PCa). CCK-8 was used to detect the cell viability. Seahorse was performed to evaluate the cell glycolysis. Luciferase assay was used to detect the transcriptional regulation. ChIP was performed to assess the binding between transcriptional factors with the promoters. Co-IP was used to assess the binding between proteins. We found that CPT1A was highly expressed in PCa tissues and cell lines. Silencing of CPT1A inhibited the viability and glycolysis of PCa cells. Mechanistically, CPT1A promoted the succinylation of SP5, which strengthened the binding between SP5 and the promoter of PDPK1. SP5 activated PDPK1 transcription and PDPK1 activated the AKT/mTOR signal pathway. These findings might provide novel targets for the diagnosis or therapy of prostate cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
19. Resensitizing Paclitaxel-Resistant Ovarian Cancer via Targeting Lipid Metabolism Key Enzymes CPT1A, SCD and FASN.
- Author
-
Ma, Qinsiyu, Liu, Zhan'ao, Wang, Tengyu, Zhao, Pengfei, Liu, Mingrui, Wang, Yifang, Zhao, Weitong, Yuan, Ying, and Li, Shuo
- Subjects
- *
PACLITAXEL , *LIPID metabolism , *OVARIAN cancer , *ENZYME metabolism , *OVARIAN epithelial cancer , *CELL migration inhibition , *CELL migration - Abstract
Epithelial ovarian cancer (EOC) is a lethal gynecological cancer, of which paclitaxel resistance is the major factor limiting treatment outcomes, and identification of paclitaxel resistance-related genes is arduous. We obtained transcriptomic data from seven paclitaxel-resistant ovarian cancer cell lines and corresponding sensitive cell lines. Define genes significantly up-regulated in at least three resistant cell lines, meanwhile they did not down-regulate in the other resistant cell lines as candidate genes. Candidate genes were then ranked according to the frequencies of significant up-regulation in resistant cell lines, defining genes with the highest rankings as paclitaxel resistance-related genes (PRGs). Patients were grouped based on the median expression of PRGs. The lipid metabolism-related gene set and the oncological gene set were established and took intersections with genes co-upregulated with PRGs, obtaining 229 co-upregulated genes associated with lipid metabolism and tumorigenesis. The PPI network obtained 19 highly confidential synergistic targets (interaction score > 0.7) that directly associated with CPT1A. Finally, FASN and SCD were up-stream substrate provider and competitor of CPT1A, respectively. Western blot and qRT-PCR results confirmed the over-expression of CPT1A, SCD and FASN in the A2780/PTX cell line. The inhibition of CPT1A, SCD and FASN down-regulated cell viability and migration, pharmacological blockade of CPT1A and SCD increased apoptosis rate and paclitaxel sensitivity of A2780/PTX. In summary, our novel bioinformatic methods can overcome difficulties in drug resistance evaluation, providing promising therapeutical strategies for paclitaxel-resistant EOC via taregting lipid metabolism-related enzymes. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
20. NQO1/CPT1A promotes the progression of pancreatic adenocarcinoma via fatty acid oxidation
- Author
-
Xu Ran, Liu Ying, Ma Liang, Sun Yao, Liu Haifeng, Yang Yang, Jin Tiefeng, and Yang Dawei
- Subjects
NQO1 ,PAAD ,metastasis ,fatty acid oxidation ,CPT1A ,Biochemistry ,QD415-436 ,Genetics ,QH426-470 - Abstract
NQO1, a cytosolic enzyme, is closely related to the progression of cancers and poor outcome of cancer patients. However, the molecular biological mechanism of NQO1 tumorigenicity in pancreatic adenocarcinoma (PAAD) has not been clearly understood. In this study, we demonstrate the molecular mechanism of NQO1 in PAAD proliferation, metastasis and fatty acid oxidation (FAO). Multiple databases and western blot analysis show that NQO1 is overexpressed in PAAD and associated with lymph node metastasis and shorter survival. Furthermore, in vitro and in vivo experiments reveal that overexpression of NQO1 improves tumor growth, metastasis and FAO in PAAD. Mechanistically, NQO1 is able to bind to carnitine palmitoyltransferase 1A (CPT1A), a key enzyme controlling FAO. Therefore, Co-IP and a series of rescue experiments demonstrate that NQO1 promotes PAAD progression via CPT1A-mediated FAO. Our findings identify CPT1A-dependent FAO as an essential metabolic pathway for NQO1 to promote the PAAD process. Targeting the NQO1/CPT1A/FAO axis in PAAD to attenuate proliferation and dissemination is a potential approach to promote a better antitumour effect and improve patient outcomes.
- Published
- 2023
- Full Text
- View/download PDF
21. DDIT4 Downregulation by siRNA Approach Increases the Activity of Proteins Regulating Fatty Acid Metabolism upon Aspirin Treatment in Human Breast Cancer Cells
- Author
-
Aistė Savukaitytė, Agnė Bartnykaitė, Justina Bekampytė, Rasa Ugenskienė, and Elona Juozaitytė
- Subjects
aspirin ,breast cancer ,DDIT4 ,ACC1 ,CPT1A ,Biology (General) ,QH301-705.5 - Abstract
Repositioning of aspirin for a more effective breast cancer (BC) treatment requires identification of predictive biomarkers. However, the molecular mechanism underlying the anticancer activity of aspirin remains fully undefined. Cancer cells enhance de novo fatty acid (FA) synthesis and FA oxidation to maintain a malignant phenotype, and the mechanistic target of rapamycin (mTORC1) is required for lipogenesis. We, therefore, aimed to test if the expression of mTORC1 suppressor DNA damage-inducible transcript (DDIT4) affects the activity of main enzymes in FA metabolism after aspirin treatment. MCF-7 and MDA-MB-468 human BC cell lines were transfected with siRNA to downregulate DDIT4. The expression of carnitine palmitoyltransferase 1 A (CPT1A) and serine 79-phosphorylated acetyl-CoA carboxylase 1 (ACC1) were analyzed by Western Blotting. Aspirin enhanced ACC1 phosphorylation by two-fold in MCF-7 cells and had no effect in MDA-MB-468 cells. Aspirin did not change the expression of CPT1A in either cell line. We have recently reported DDIT4 itself to be upregulated by aspirin. DDIT4 knockdown resulted in 1.5-fold decreased ACC1 phosphorylation (dephosphorylation activates the enzyme), 2-fold increased CPT1A expression in MCF-7 cells, and 2.8-fold reduced phosphorylation of ACC1 following aspirin exposure in MDA-MB-468 cells. Thus, DDIT4 downregulation raised the activity of main lipid metabolism enzymes upon aspirin exposure which is an undesired effect as FA synthesis and oxidation are linked to malignant phenotype. This finding may be clinically relevant as DDIT4 expression has been shown to vary in breast tumors. Our findings justify further, more extensive investigation of the role of DDIT4 in aspirin’s effect on fatty acid metabolism in BC cells.
- Published
- 2023
- Full Text
- View/download PDF
22. A novel CPT1A covalent inhibitor modulates fatty acid oxidation and CPT1A-VDAC1 axis with therapeutic potential for colorectal cancer
- Author
-
Anni Hu, Hang Wang, Qianqian Xu, Yuqi Pan, Zeyu Jiang, Sheng Li, Yi Qu, Yili Hu, Hao Wu, and Xinzhi Wang
- Subjects
Colorectal cancer ,2,6-Dihydroxypeperomin B ,CPT1A ,Covalent inhibitor ,VDAC1 ,Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Colorectal cancer (CRC) is a common and deadly disease of the digestive system, but its targeted therapy is hampered by the lack of reliable and specific biomarkers. Hence, discovering new therapeutic targets and agents for CRC is an urgent and challenging task. Here we report that carnitine palmitoyltransferase 1A (CPT1A), a mitochondrial enzyme that catalyzes fatty acid oxidation (FAO), is a potential target for CRC treatment. We show that CPT1A is overexpressed in CRC cells and that its inhibition by a secolignan-type compound, 2,6-dihydroxypeperomin B (DHP-B), isolated from the plant Peperomia dindygulensis, suppresses tumor cell growth and induces apoptosis. We demonstrate that DHP-B covalently binds to Cys96 of CPT1A, blocks FAO, and disrupts the mitochondrial CPT1A-VDAC1 interaction, leading to increased mitochondrial permeability and reduced oxygen consumption and energy metabolism in CRC cells. We also reveal that CPT1A expression correlates with the survival of tumor-bearing animals and that DHP-B exhibits anti-CRC activity in vitro and in vivo. Our study uncovers the molecular mechanism of DHP-B as a novel CPT1A inhibitor and provides a rationale for its preclinical development as well as a new strategy for CRC targeted therapy.
- Published
- 2023
- Full Text
- View/download PDF
23. CPT1A in cancer: Tumorigenic roles and therapeutic implications.
- Author
-
SHENGJIE SONG and ZHIZHOU SHI
- Subjects
- *
FATTY acid oxidation , *CANCER treatment , *ENERGY consumption , *CANCER invasiveness , *THERAPEUTIC use of antineoplastic agents - Abstract
Metabolic reprogramming frequently occurs in the majority of cancers, wherein fatty acid oxidation (FAO) is usually induced and serves as a compensatory mechanism to improve energy consumption. Carnitine palmitoyltransferase 1A (CPT1A) is the rate-limiting enzyme for FAO and is widely involved in tumor growth, metastasis, and chemo-/radio-resistance. This review summarizes the most recent advances in understanding the oncogenic roles and mechanisms of CPT1A in tumorigenesis, including in proliferation and tumor growth, invasion and metastasis, and the tumor microenvironment. Importantly, CPT1A has been shown to be a biomarker for diagnosis and prognosis prediction and proved to be a candidate therapeutic target, especially for the treatment of drug- and radiation-resistant tumors. In summary, CPT1A plays remarkable roles in promoting cancer progression and is a potential anticancer therapeutic target. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
24. Expression Variation of CPT1A Induces Lipid Reconstruction in Goat Intramuscular Precursor Adipocytes.
- Author
-
Tang, Yinmei, Zhang, Wenyang, Wang, Yinggui, Li, Haiyang, Zhang, Changhui, Wang, Yong, Lin, Yaqiu, Shi, Hengbo, Xiang, Hua, Huang, Lian, and Zhu, Jiangjiang
- Subjects
- *
ADIPOGENESIS , *GENE expression , *CARNITINE palmitoyltransferase , *FAT cells , *CELL cycle , *INHIBITION of cellular proliferation - Abstract
Intramuscular fat (IMF) deposition is one of the most important factors affecting meat quality and is closely associated with the expression of carnitine palmitoyl transferase 1A (CPT1A) which facilitates the transfer of long-chain fatty acids (LCFAs) into the mitochondria. However, the role of how CPT1A regulates the IMF formation remains unclear. Herein, we established the temporal expression profile of CPT1A during the differentiation of goat intramuscular precursor adipocytes. Functionally, the knockdown of CPT1A by siRNA treatment significantly increased the mRNA expression of adipogenic genes and promoted lipid deposition in goat intramuscular precursor adipocytes. Meanwhile, a CPT1A deficiency inhibited cell proliferation and promoted cell apoptosis significantly. CPT1A was then supported by the overexpression of CPT1A which significantly suppressed the cellular triglyceride deposition and promoted cell proliferation although the cell apoptosis also was increased. For RNA sequencing, a total of 167 differential expression genes (DEGs), including 125 upregulated DEGs and 42 downregulated DEGs, were observed after the RNA silencing of CPT1A compared to the control, and were predicted to enrich in the focal adhesion pathway, cell cycle, apoptosis and the MAPK signaling pathway by KEGG analysis. Specifically, blocking the MAPK signaling pathway by a specific inhibitor (PD169316) rescued the promotion of cell proliferation in CPT1A overexpression adipocytes. In conclusion, the expression variation of CPT1A may reconstruct the lipid distribution between cellular triglyceride deposition and cell proliferation in goat intramuscular precursor adipocyte. Furthermore, we demonstrate that CPT1A promotes the proliferation of goat adipocytes through the MAPK signaling pathway. This work widened the genetic regulator networks of IMF formation and delivered theoretical support for improving meat quality from the aspect of IMF deposition. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
25. Targeting carnitine palmitoyltransferase 1 isoforms in the hypothalamus: A promising strategy to regulate energy balance.
- Author
-
Rodríguez‐Rodríguez, Rosalía, Fosch, Anna, Garcia‐Chica, Jesús, Zagmutt, Sebastián, and Casals, Nuria
- Subjects
- *
CARNITINE palmitoyltransferase , *HYPOTHALAMUS , *LIPID metabolism , *FOOD consumption , *NUTRITIONAL status - Abstract
Tackling the growing incidence and prevalence of obesity urgently requires uncovering new molecular pathways with therapeutic potential. The brain, and in particular the hypothalamus, is a major integrator of metabolic signals from peripheral tissues that regulate functions such as feeding behavior and energy expenditure. In obesity, hypothalamic capacity to sense nutritional status and regulate these functions is altered. An emerging line of research is that hypothalamic lipid metabolism plays a critical role in regulating energy balance. Here, we focus on the carnitine palmitoyltransferase 1 (CPT1) enzyme family responsible for long‐chain fatty acid metabolism. The evidence suggests that two of its isoforms expressed in the brain, CPT1A and CPT1C, play a crucial role in hypothalamic lipid metabolism, and their promise as targets in food intake and bodyweight management is currently being intensively investigated. In this review we describe and discuss the metabolic actions and potential up‐ and downstream effectors of hypothalamic CPT1 isoforms, and posit the need to develop innovative nanomedicine platforms for selective targeting of CPT1 and related nutrient sensors in specific brain areas as potential next‐generation therapy to treat obesity. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
26. CPT1A in AgRP neurons is required for sex-dependent regulation of feeding and thirst
- Author
-
Sebastián Zagmutt, Paula Mera, Ismael González-García, Kevin Ibeas, María del Mar Romero, Arnaud Obri, Beatriz Martin, Anna Esteve-Codina, M. Carmen Soler-Vázquez, Marianela Bastias-Pérez, Laia Cañes, Elisabeth Augé, Carme Pelegri, Jordi Vilaplana, Xavier Ariza, Jordi García, José Martinez-González, Núria Casals, Miguel López, Richard Palmiter, Elisenda Sanz, Albert Quintana, Laura Herrero, and Dolors Serra
- Subjects
CPT1A ,Fatty acid metabolism ,AgRP neurons ,Energy balance ,Food intake ,Thirst ,Medicine ,Physiology ,QP1-981 - Abstract
Highlights Fatty acid metabolism and CPT1A in AgRP neurons show marked sex-dependent differences in the control of feeding. Cpt1a gene deletion in AgRP neurons increases energy expenditure in females but not in males. CPT1A in AgRP neurons is involved in the control of thirst and fluid homeostasis. Cpt1a gene deletion in AgRP neurons induces morphological, mitochondrial, and gene expression alterations in a sex-dependent manner.
- Published
- 2023
- Full Text
- View/download PDF
27. WY-14643 attenuates lipid deposition via activation of the PPARα/CPT1A axis by targeting Gly335 to inhibit cell proliferation and migration in ccRCC
- Author
-
Rui Wang, Jun Zhao, Jiacheng Jin, Yun Tian, Lan Lan, Xuejian Wang, Liang Zhu, and Jianbo Wang
- Subjects
ccRCC ,WY-14643 ,Lipid accumulation ,PPARα ,CPT1A ,Nutritional diseases. Deficiency diseases ,RC620-627 - Abstract
Abstract Background Histologically, cytoplasmic deposits of lipids and glycogen are common in clear cell renal cell carcinoma (ccRCC). Owing to the significance of lipid deposition in ccRCC, numerous trials targeting lipid metabolism have shown certain therapeutic potential. The agonism of peroxisome proliferator-activated receptor-α (PPARα) via ligands, including WY-14,643, has been considered a promising intervention for cancers. Methods First, the effects of WY-14,643 on malignant behaviors were investigated in ccRCC in vitro. After RNA sequencing, the changes in lipid metabolism, especially neutral lipids and glycerol, were further evaluated. Finally, the underlying mechanisms were revealed. Results Phenotypically, the proliferation and migration of ccRCC cells treated with WY-14,643 were significantly inhibited in vitro. A theoretical functional mechanism was proposed in ccRCC: WY-14,643 mediates lipid consumption by recognizing carnitine palmitoyltransferase 1 A (CPT1A). Activation of PPARα using WY-14,643 reduces lipid deposition by increasing the CPT1A level, which also suppresses the NF-κB signaling pathway. Spatially, WY-14,643 binds and activates PPARα by targeting Gly335. Conclusion Overall, WY-14,643 suppresses the biological behaviors of ccRCC in terms of cell proliferation, migration, and cell cycle arrest. Furthermore, its anticancer properties are mediated by the inhibition of lipid accumulation, at least in part, through the PPARα/CPT1A axis by targeting Gly335, as part of the process, NF-κB signaling is also suppressed. Pharmacological activation of PPARα might offer a new treatment option for ccRCC.
- Published
- 2022
- Full Text
- View/download PDF
28. Epigenetic Histone Methylation of PPARγ and CPT1A Signaling Contributes to Betahistine Preventing Olanzapine-Induced Dyslipidemia.
- Author
-
Su, Yueqing, Deng, Chao, Liu, Xuemei, and Lian, Jiamei
- Subjects
- *
HISTONE methylation , *HISTAMINE receptors , *GENE expression , *DYSLIPIDEMIA , *PEROXISOME proliferator-activated receptors , *FATTY acid oxidation - Abstract
As a partial histamine H1 receptor agonist and H3 antagonist, betahistine has been reported to partially prevent olanzapine-induced dyslipidemia and obesity through a combination therapy, although the underlying epigenetic mechanisms are still not known. Recent studies have revealed that histone regulation of key genes for lipogenesis and adipogenesis in the liver is one of the crucial mechanisms for olanzapine-induced metabolic disorders. This study investigated the role of epigenetic histone regulation in betahistine co-treatment preventing dyslipidemia and fatty liver caused by chronic olanzapine treatment in a rat model. In addition to abnormal lipid metabolism, the upregulation of peroxisome proliferator-activated receptor γ (PPARγ) and CCAAT/enhancer binding protein (C/EBPα), as well as the downregulation of carnitine palmitoyltransferase 1A (CPT1A) in the liver induced by olanzapine, were significantly attenuated by betahistine co-treatment. In addition, betahistine co-treatment significantly enhanced the global expression of H3K4me and the enrichment of H3K4me binding on the promoter of Cpt1a gene as revealed by ChIP-qPCR, but inhibited the expression of one of its site-specific demethylases, lysine (K)-specific demethylase 1A (KDM1A). Betahistine co-treatment also significantly enhanced the global expression of H3K9me and the enrichment of H3K9me binding on the promoter of the Pparg gene, but inhibited the expression of two of its site-specific demethylases, lysine demethylase 4B (KDM4B) and PHD finger protein 2 (PHF2). These results suggest that betahistine attenuates abnormal adipogenesis and lipogenesis triggered by olanzapine through modulating hepatic histone methylation, and thus inhibiting the PPARγ pathway-mediated lipid storage, while at the same time promoting CP1A-mediated fatty acid oxidation. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
29. Mitochondrial CPT1A: Insights into structure, function, and basis for drug development.
- Author
-
Kai Liang
- Subjects
DRUG development ,DRUG resistance in cancer cells ,DISEASE risk factors ,DRUG target ,METASTASIS - Abstract
Carnitine Palmitoyl-Transferase1A (CPT1A) is the rate-limiting enzyme in the fatty acid β-oxidation, and its deficiency or abnormal regulation can result in diseases like metabolic disorders and various cancers. Therefore, CPT1A is a desirable drug target for clinical therapy. The deep comprehension of human CPT1A is crucial for developing the therapeutic inhibitors like Etomoxir. CPT1A is an appealing druggable target for cancer therapies since it is essential for the survival, proliferation, and drug resistance of cancer cells. It will help to lower the risk of cancer recurrence and metastasis, reduce mortality, and offer prospective therapy options for clinical treatment if the effects of CPT1A on the lipid metabolism of cancer cells are inhibited. Targeted inhibition of CPT1A can be developed as an effective treatment strategy for cancers from a metabolic perspective. However, the pathogenic mechanism and recent progress of CPT1A in diseases have not been systematically summarized. Here we discuss the functions of CPT1A in health and diseases, and prospective therapies targeting CPT1A. This review summarizes the current knowledge of CPT1A, hoping to prompt further understanding of it, and provide foundation for CPT1Atargeting drug development. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
30. Ror1 promotes PPARα‐mediated fatty acid metabolism in astrocytes.
- Author
-
Tanaka, Yuki, Minami, Yasuhiro, and Endo, Mitsuharu
- Subjects
- *
FATTY acids , *ASTROCYTES , *CELL polarity , *PEROXISOME proliferator-activated receptors , *GENE expression , *DEVELOPMENTAL neurobiology , *METABOLISM , *ADIPOGENESIS - Abstract
Ror1 signaling regulates cell polarity, migration, proliferation, and differentiation during developmental morphogenesis, and plays an important role in regulating neurogenesis in the embryonic neocortices. However, the role of Ror1 signaling in the brains after birth remains largely unknown. Here, we found that expression levels of Ror1 in the mouse neocortices increase during the postnatal period, when astrocytes mature and start expressing GFAP. Indeed, Ror1 is highly expressed in cultured postmitotic mature astrocytes. RNA‐Seq analysis revealed that Ror1 expressed in cultured astrocytes mediates upregulated expression of genes related to fatty acid (FA) metabolism, including the gene encoding carnitine palmitoyl‐transferase 1a (Cpt1a), the rate‐limiting enzyme of mitochondrial fatty acid β‐oxidation (FAO). We also found that Ror1 promotes the degradation of lipid droplets (LDs) accumulated in the cytoplasm of cultured astrocytes after oleic acid loading, and that suppressed expression of Ror1 decreases the amount of FAs localized at mitochondria, intracellular ATP levels, and expression levels of peroxisome proliferator‐activated receptor α (PPARα) target genes, including Cpt1a. Collectively, these findings indicate that Ror1 signaling promotes PPARα‐mediated transcription of FA metabolism‐related genes, thereby facilitating the availability of FAs derived from LDs for mitochondrial FAO in the mature astrocytes. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
31. CPT1A in AgRP neurons is required for sex-dependent regulation of feeding and thirst.
- Author
-
Zagmutt, Sebastián, Mera, Paula, González-García, Ismael, Ibeas, Kevin, Romero, María del Mar, Obri, Arnaud, Martin, Beatriz, Esteve-Codina, Anna, Soler-Vázquez, M. Carmen, Bastias-Pérez, Marianela, Cañes, Laia, Augé, Elisabeth, Pelegri, Carme, Vilaplana, Jordi, Ariza, Xavier, García, Jordi, Martinez-González, José, Casals, Núria, López, Miguel, and Palmiter, Richard
- Subjects
- *
INGESTION , *INNERVATION , *BROWN adipose tissue , *WATER-electrolyte balance (Physiology) , *DRINKING (Physiology) , *GHRELIN receptors , *NEURONS , *FATTY acid oxidation - Abstract
Background: Fatty acid metabolism in the hypothalamus has an important role in food intake, but its specific role in AgRP neurons is poorly understood. Here, we examined whether carnitinea palmitoyltransferase 1A (CPT1A), a key enzyme in mitochondrial fatty acid oxidation, affects energy balance. Methods: To obtain Cpt1aKO mice and their control littermates, Cpt1a(flox/flox) mice were crossed with tamoxifen-inducible AgRPCreERT2 mice. Food intake and body weight were analyzed weekly in both males and females. At 12 weeks of age, metabolic flexibility was determined by ghrelin-induced food intake and fasting–refeeding satiety tests. Energy expenditure was analyzed by calorimetric system and thermogenic activity of brown adipose tissue. To study fluid balance the analysis of urine and water intake volumes; osmolality of urine and plasma; as well as serum levels of angiotensin and components of RAAS (renin–angiotensin–aldosterone system) were measured. At the central level, changes in AgRP neurons were determined by: (1) analyzing specific AgRP gene expression in RiboTag–Cpt1aKO mice obtained by crossing Cpt1aKO mice with RiboTag mice; (2) measuring presynaptic terminal formation in the AgRP neurons with the injection of the AAV1-EF1a-DIO-synaptophysin-GFP in the arcuate nucleus of the hypothalamus; (3) analyzing AgRP neuronal viability and spine formations by the injection AAV9-EF1a-DIO-mCherry in the arcuate nucleus of the hypothalamus; (4) analyzing in situ the specific AgRP mitochondria in the ZsGreen-Cpt1aKO obtained by breeding ZsGreen mice with Cpt1aKO mice. Two-way ANOVA analyses were performed to determine the contributions of the effect of lack of CPT1A in AgRP neurons in the sex. Results: Changes in food intake were just seen in male Cpt1aKO mice while only female Cpt1aKO mice increased energy expenditure. The lack of Cpt1a in the AgRP neurons enhanced brown adipose tissue activity, mainly in females, and induced a substantial reduction in fat deposits and body weight. Strikingly, both male and female Cpt1aKO mice showed polydipsia and polyuria, with more reduced serum vasopressin levels in females and without osmolality alterations, indicating a direct involvement of Cpt1a in AgRP neurons in fluid balance. AgRP neurons from Cpt1aKO mice showed a sex-dependent gene expression pattern, reduced mitochondria and decreased presynaptic innervation to the paraventricular nucleus, without neuronal viability alterations. Conclusions: Our results highlight that fatty acid metabolism and CPT1A in AgRP neurons show marked sex differences and play a relevant role in the neuronal processes necessary for the maintenance of whole-body fluid and energy balance. Highlights: Fatty acid metabolism and CPT1A in AgRP neurons show marked sex-dependent differences in the control of feeding. Cpt1a gene deletion in AgRP neurons increases energy expenditure in females but not in males. CPT1A in AgRP neurons is involved in the control of thirst and fluid homeostasis. Cpt1a gene deletion in AgRP neurons induces morphological, mitochondrial, and gene expression alterations in a sex-dependent manner. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
32. CPT1A modulates PI3K/Akt/mTOR pathway to promote preeclampsia.
- Author
-
Chen, Miaomiao, Chao, Bingdi, Xu, Jiacheng, Liu, Zheng, Tao, Yuelan, He, Jie, Wang, Jie, Yang, Huan, Luo, Xin, and Qi, Hongbo
- Abstract
Preeclampsia (PE) refers to a syndrome of new-onset hypertension with multisystem involvement and damage after 20 weeks of gestation. Defective placentation due to dysregulated behaviors of trophoblast cells is considered a predominant cause of PE. Immunofluorescence (if) and Western blot were used to detect the expression and localization of Carnitine palmitoyltransferase 1A (CPT1A) in placenta. CPT1A protein was overexpressed/knocked down in HTR8/SVneo cells by lentiviral/siRNA interference method. CCK-8 Assay, Western blot, flow cytometry, Wound healing and Transwell assay were used to detect the functional impact of CPT1A on HTR8/SVneo cells. Transcriptomics and bioinformatics analysis were used to predict the possible pathway of CPT1A participating in PE. CPT1A was upregulated in preeclamptic placentas when compared with normal controls. The abnormal expression of CPT1A in HTR8/SVneo cells is associated with the invasion and migration of HTR8/SVneo cells but is not related to the proliferation, cycle, and apoptosis of HTR8/SVneo cells. The results of Transcriptomic and Western blots suggest that phosphatidylinositol-3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/Akt/mTOR) pathway are activated in the si-CPT1A-1796 group. Compared with the si-NC group, the epithelial-mesenchymal transition (EMT) process of HTR8/SVneo cells in the si-CPT1A- 1796 group was significantly enhanced. CPT1A may participate in the pathogenesis of PE by inhibiting the EMT process of HTR8/SVneo cells through the PI3K/AKT/mTOR signaling axis. Thus, the newly unveiled novel function of CPT1A in PE via the PI3K/Akt/mTOR pathway provides a novel insight into the pathogenesis of PE. • CPT1A (a rate-limiting enzyme of fatty acid beta-oxidation) is over-expressed in PE placenta. • CPT1A modulate invasion and migration of HTR8/SVeno cells and did not affect their proliferation and apoptosis. • CPT1A contribute to PE through modulating placental EMT level by inhibiting PI3K/AKT signaling pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
33. Genotype–phenotype correlations in CPT1A deficiency detected by newborn screening in Pacific populations
- Author
-
Isaac Bernhardt, Emma Glamuzina, Leah K. Dowsett, Dianne Webster, Detlef Knoll, Kevin Carpenter, Michael J. Bennett, Michelle Maeda, and Callum Wilson
- Subjects
carnitine palmitoyltransferase type 1 deficiency ,CPT1A ,fatty acid oxidation disorders ,newborn screening ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 ,Genetics ,QH426-470 - Abstract
Abstract Carnitine palmitoyltransferase 1A (CPT1A) deficiency is a long chain fatty acid oxidation disorder, typically presenting with hypoketotic hypoglycaemia and liver dysfunction during fasting and intercurrent illness. Classical CPT1A deficiency is a rare disease, although a milder ‘Arctic variant' (p.P479L) is common in the Inuit population. Since the introduction of expanded metabolic screening (EMS), the newborn screening programmes of Hawai'i and New Zealand (NZ) have detected a significant increase in the incidence of CPT1A deficiency. We report 22 individuals of Micronesian descent (12 in NZ and 10 in Hawai'i), homozygous for a CPT1A c.100T>C (p.S34P) variant detected by EMS or ascertained following diagnosis of a family member. No individuals with the Micronesian variant presented clinically with metabolic decompensation prior to diagnosis or during follow‐up. Three asymptomatic homozygous adults were detected following the diagnosis of their children by EMS. CPT1A activity in cultured skin fibroblasts showed residual enzyme activity of 26% of normal controls. Secondly, we report three individuals from two unrelated Niuean families who presented clinically with symptoms of classic CPT1A deficiency, prior to the introduction of EMS. All were found to be homozygous for a CPT1A c.2122A>C (p.S708R) variant. CPT1A activity in fibroblasts of all three individuals was severely reduced at 4% of normal controls. Migration pressure, in part due to climate change may lead to increased frequency of presentation of Pacific peoples to regional metabolic services around the world. Knowledge of genotype–phenotype correlations in these populations will therefore inform counselling and treatment of those detected by newborn screening.
- Published
- 2022
- Full Text
- View/download PDF
34. CircITGB5 regulates the proliferation and adipogenic differentiation of chicken intramuscular preadipocytes through the miR-181b-5p/CPT1A axis.
- Author
-
Li, Hongtai, Zhang, Hongyuan, Dai, Yaqi, Li, Shuaihao, Gu, Jinxing, Wu, Ran, Jia, Jiyu, Shen, Jingqi, Zhang, Yanhua, Li, Hong, Liu, Xiaojun, Tian, Yadong, Kang, Xiangtao, Zhao, Yinli, and Li, Guoxi
- Subjects
- *
COMPETITIVE endogenous RNA , *LINCRNA , *GENE expression , *CHICKEN as food , *NON-coding RNA , *CIRCULAR RNA - Abstract
Non-coding RNA is known to play a crucial role in the generation and deposition of intramuscular fat (IMF) in vertebrates by regulating the expression of genes involved in the synthesis, degradation and transportation of IMF. With the introduction of the competing endogenous RNA (ceRNA) hypothesis, circular RNA (circRNA) and long non-coding RNA (lncRNA) have been identified as natural "sponges" for microRNA (miRNA), yet their precise mechanisms and biological functions in chicken IMF are still not fully understood. In earlier research, we observed a significant association between Carnitine O-palmitoyltransferase 1, liver isoform (CPT1A) and fatty acid metabolism in chicken breast muscle. In this study, we utilized bioinformatics and previous sequencing data to construct a ceRNA regulatory network linked to intramuscular adipogenesis and deposition in chickens. This investigation aimed to unravel the post-transcriptional regulatory mechanism of CPT1A. The results showed that, functionally, CPT1A inhibited the proliferation and differentiation of intramuscular preadipocytes and affected the fatty acid composition of intramuscular adipocytes, and serving as a direct target of miR-181b-5p. In terms of mechanism, through a combination of bioinformatics analysis, dual luciferase reporter assays, quantitative real-time PCR (qRT-PCR), and Western blotting (WB), it was confirmed the interaction of a novel circRNA named circITGB5 with miR-181b-5p. CircITGB5 increased the expression of CPT1A mRNA and protein through post-transcriptional regulation. Acting as a ceRNA, circITGB5 competed with miR-181b-5p for binding to CPT1A , thereby shielding CPT1A from miR-181b-5p-mediated degradation. Subsequent rescue experiments confirmed the above conclusions. In conclusion, circITGB5 regulates the expression of the downstream target gene CPT1A by adsorption of miR-181b-5p, thereby regulating the proliferation and adipogenic differentiation of intramuscular preadipocytes. This discovery may have implications for achieving balanced breeding strategies aimed at enhancing the quality of chicken. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
35. CPT1A/2-Mediated FAO Enhancement—A Metabolic Target in Radioresistant Breast Cancer
- Author
-
Han, Shujun, Wei, Ryan, Zhang, Xiaodi, Jiang, Nian, Fan, Ming, Huang, Jie Hunter, Xie, Bowen, Zhang, Lu, Miao, Weili, Butler, Ashley Chen-Ping, Coleman, Matthew A, Vaughan, Andrew T, Wang, Yinsheng, Chen, Hong-Wu, Liu, Jiankang, and Li, Jian Jian
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Stem Cell Research ,Radiation Oncology ,Stem Cell Research - Nonembryonic - Non-Human ,Stem Cell Research - Nonembryonic - Human ,Genetics ,Nutrition ,Breast Cancer ,Women's Health ,Cancer ,2.1 Biological and endogenous factors ,breast cancer stem cells ,CPT1A ,CPT2 ,FAO ,metabolism ,radioresistance ,breast cancer ,CPT1A/CPT2 ,Clinical sciences ,Oncology and carcinogenesis - Abstract
Tumor cells, including cancer stem cells (CSCs) resistant to radio- and chemotherapy, must enhance metabolism to meet the extra energy demands to repair and survive such genotoxic conditions. However, such stress-induced adaptive metabolic alterations, especially in cancer cells that survive radiotherapy, remain unresolved. In this study, we found that CPT1 (Carnitine palmitoyl transferase I) and CPT2 (Carnitine palmitoyl transferase II), a pair of rate-limiting enzymes for mitochondrial fatty acid transportation, play a critical role in increasing fatty acid oxidation (FAO) required for the cellular fuel demands in radioresistant breast cancer cells (RBCs) and radiation-derived breast cancer stem cells (RD-BCSCs). Enhanced CPT1A/CPT2 expression was detected in the recurrent human breast cancers and associated with a worse prognosis in breast cancer patients. Blocking FAO via a FAO inhibitor or by CRISPR-mediated CPT1A/CPT2 gene deficiency inhibited radiation-induced ERK activation and aggressive growth and radioresistance of RBCs and RD-BCSCs. These results revealed that switching to FAO contributes to radiation-induced mitochondrial energy metabolism, and CPT1A/CPT2 is a potential metabolic target in cancer radiotherapy.
- Published
- 2019
36. Alfalfa Xeno-miR168b Target CPT1A to Regulate Milk Fat Synthesis in Bovine Mammary Epithelial Cells.
- Author
-
Jia, Jingying, Duan, Hongjuan, Liu, Baobao, Ma, Yanfen, Ma, Yun, and Cai, Xiaoyan
- Subjects
MILKFAT ,EPITHELIAL cells ,FAT content of milk ,LIPID metabolism ,ALFALFA ,STAINS & staining (Microscopy) - Abstract
It was shown that microRNAs (miRNAs) play an important role in the synthesis of milk fat; thus, this manuscript evaluated whether exogenous miRNA (xeno-miRNAs) from alfalfa could influence the milk fat content in dairy cows. At first, mtr-miR168b was screened from dairy cow milk and blood. Then, EdU staining, flow cytometry, Oil Red O staining, qRT-PCR, and WB were applied to explore the effect of xeno-miR168b on the proliferation, apoptosis, and lipid metabolism of bovine mammary epithelial cells (BMECs). Finally, in order to clarify the pathway that regulated the lipid metabolism of BMECs using xeno-miR168b, a double-luciferase reporter assay was used to verify the target gene related to milk fat. These results showed that overexpression of xeno-miR168b inhibited cell proliferation but promoted apoptosis, which also decreased the expression of several lipid metabolism genes, including PPARγ, SCD1, C/EBPβ, and SREBP1, significantly inhibited lipid droplet formation, and reduced triglyceride content in BMECs. Furthermore, the targeting relationship between CPT1A and xeno-miR168b was determined and it was confirmed that CPT1A silencing reduced the expression of lipid metabolism genes and inhibited fat accumulation in BMECs. These findings identified xeno-miR168b from alfalfa as a cross-kingdom regulatory element that could influence milk fat content in dairy cows by modulating CPT1A expression. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
37. WY-14643 attenuates lipid deposition via activation of the PPARα/CPT1A axis by targeting Gly335 to inhibit cell proliferation and migration in ccRCC.
- Author
-
Wang, Rui, Zhao, Jun, Jin, Jiacheng, Tian, Yun, Lan, Lan, Wang, Xuejian, Zhu, Liang, and Wang, Jianbo
- Subjects
- *
INHIBITION of cellular proliferation , *CARNITINE palmitoyltransferase , *LIPID metabolism , *RENAL cell carcinoma , *LIPIDS , *RNA sequencing , *RNA metabolism - Abstract
Background: Histologically, cytoplasmic deposits of lipids and glycogen are common in clear cell renal cell carcinoma (ccRCC). Owing to the significance of lipid deposition in ccRCC, numerous trials targeting lipid metabolism have shown certain therapeutic potential. The agonism of peroxisome proliferator-activated receptor-α (PPARα) via ligands, including WY-14,643, has been considered a promising intervention for cancers. Methods: First, the effects of WY-14,643 on malignant behaviors were investigated in ccRCC in vitro. After RNA sequencing, the changes in lipid metabolism, especially neutral lipids and glycerol, were further evaluated. Finally, the underlying mechanisms were revealed. Results: Phenotypically, the proliferation and migration of ccRCC cells treated with WY-14,643 were significantly inhibited in vitro. A theoretical functional mechanism was proposed in ccRCC: WY-14,643 mediates lipid consumption by recognizing carnitine palmitoyltransferase 1 A (CPT1A). Activation of PPARα using WY-14,643 reduces lipid deposition by increasing the CPT1A level, which also suppresses the NF-κB signaling pathway. Spatially, WY-14,643 binds and activates PPARα by targeting Gly335. Conclusion: Overall, WY-14,643 suppresses the biological behaviors of ccRCC in terms of cell proliferation, migration, and cell cycle arrest. Furthermore, its anticancer properties are mediated by the inhibition of lipid accumulation, at least in part, through the PPARα/CPT1A axis by targeting Gly335, as part of the process, NF-κB signaling is also suppressed. Pharmacological activation of PPARα might offer a new treatment option for ccRCC. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
38. The Intersection of Anoikis Resistance and Fatty Acid Metabolism in Cancer
- Author
-
Bapat, Jaidev, Bitler, Benjamin G., and Frisch, Steven M., editor
- Published
- 2021
- Full Text
- View/download PDF
39. RNF5 exacerbates steatotic HCC by enhancing fatty acid oxidation via the improvement of CPT1A stability.
- Author
-
Chen X, Zhang YW, Ren H, Dai C, Zhang M, Li X, Xu K, Li J, Ju Y, Pan X, Xia P, Ma W, He W, Wu T, and Yuan Y
- Abstract
Non-alcoholic fatty liver disease (NAFLD) is expected to become the leading risk factor for liver cancer, surpassing viral hepatitis. Unlike viral hepatitis-related hepatocellular carcinoma (HCC), the role of excessive nutrient supply in steatotic HCC is not well understood, hindering effective prevention and treatment strategies. Therefore, it is crucial to identify key molecules in the pathogenesis of steatotic HCC, investigate changes in metabolic reprogramming due to excessive fatty acid (FA) supply, understand its molecular mechanisms, and find potential therapeutic targets. Trans-species transcriptome analysis identified Ring Finger Protein 5 (RNF5) as a critical regulator of steatotic HCC. RNF5 upregulation is associated with poor prognosis in steatotic HCC compared to canonical HCC. In vitro and in vivo studies showed that RNF5 exacerbates HCC in the presence of additional FA supply. Lipidomics and transcriptome analyses revealed that RNF5 significantly increases carnitine palmitoyltransferase 1A (CPT1A) mRNA levels and is positively correlated with fatty acid oxidation (FAO). Protein interaction analysis demonstrated that RNF5 promotes K63-type ubiquitination of insulin-like growth factor-2 mRNA-binding protein 1 (IGF2BP1), enhancing CPT1A mRNA stabilization through m6A modification. Additionally, peroxisome proliferator-activated receptor gamma (PPARγ) was found to activate RNF5 expression specifically in HCC cells. Mechanistically, excessive exogenous FAs reorganize FA metabolism in HCC cells, worsening steatotic HCC via the PPARγ-RNF5-IGF2BP1-CPT1A axis. This study highlights a distinct FA metabolism pattern in steatotic HCC, providing valuable insights for potential therapeutic targets., Competing Interests: Declaration of Competing Interest ☒ The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024. Published by Elsevier B.V.)
- Published
- 2024
- Full Text
- View/download PDF
40. ANGPTL3 overcomes sorafenib resistance via suppression of SNAI1 and CPT1A in liver cancer.
- Author
-
Lin YH, Chen CY, Chi HC, Wu MH, Lai MW, and Yeh CT
- Abstract
Liver cancer, encompassing hepatocellular carcinoma (HCC) and hepatoblastoma, the latter of which primarily occurs in early childhood, is the most common malignant tumor arising from liver and is responsible for a significant number of cancer-related deaths worldwide. Targeted drugs have been used for anti-liver cancer treatment in the advanced stage, while their efficacy is greatly compromised by development of drug resistance. Drug resistance is a complicated process regulated by intrinsic and extrinsic signals and has been associated with poorer prognosis in cancer patients. In the current study, online available dataset analysis uncovered that angiopoietin-like protein 3 (ANGPTL3) manifested lower expression in sorafenib-resistant liver cancer cell lines. Additionally, ANGPTL3 was downregulated in HCC tissues, with its expression positively correlated with good prognosis. Functionally, ectopic expression of ANGPTL3 re-sensitized sorafenib-resistant cells, enhancing the sorafenib-induced reduction in cell viability and migration by suppressing zinc finger protein SNAI1 (SNAI1) expression and the protein stability of carnitine O-palmitoyltransferase 1, liver isoform (CPT1A). Clinical correlation analysis revealed that ANGPTL3 was negatively associated with SNAI1 expression. In conclusion, we identify a novel association between ANGPTL3, SNAI1 and CPT1A on sorafenib therapeutic response. Targeting ANGPTL3/SNAI1/CPT1A axis may serve as a therapeutic approach to improve prognosis of liver cancer patients with sorafenib resistance., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024. Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
41. CPT1A mediates radiation sensitivity in colorectal cancer.
- Author
-
Chen Z, Yu L, Zheng Z, Wang X, Guo Q, Chen Y, Zhang Y, Zhang Y, Xiao J, Chen K, Fan H, and Ding Y
- Subjects
- Humans, Mice, Animals, Carnitine O-Palmitoyltransferase metabolism, Carnitine O-Palmitoyltransferase genetics, Cell Line, Tumor, Gene Expression Regulation, Neoplastic, Male, Reactive Oxygen Species metabolism, Female, Colorectal Neoplasms genetics, Colorectal Neoplasms radiotherapy, Radiation Tolerance genetics
- Abstract
The prevalence and mortality rates of colorectal cancer (CRC) are increasing worldwide. Radiation resistance hinders radiotherapy, a standard treatment for advanced CRC, leading to local recurrence and metastasis. Elucidating the molecular mechanisms underlying radioresistance in CRC is critical to enhance therapeutic efficacy and patient outcomes. Bioinformatic analysis and tumour tissue examination were conducted to investigate the CPT1A mRNA and protein levels in CRC and their correlation with radiotherapy efficacy. Furthermore, lentiviral overexpression and CRISPR/Cas9 lentiviral vectors, along with in vitro and in vivo radiation experiments, were used to explore the effect of CPT1A on radiosensitivity. Additionally, transcriptomic sequencing, molecular biology experiments, and bioinformatic analyses were employed to elucidate the molecular mechanisms by which CPT1A regulates radiosensitivity. CPT1A was significantly downregulated in CRC and negatively correlated with responsiveness to neoadjuvant radiotherapy. Functional studies suggested that CPT1A mediates radiosensitivity, influencing reactive oxygen species (ROS) scavenging and DNA damage response. Transcriptomic and molecular analyses highlighted the involvement of the peroxisomal pathway. Mechanistic exploration revealed that CPT1A downregulates the FOXM1-SOD1/SOD2/CAT axis, moderating cellular ROS levels after irradiation and enhancing radiosensitivity. CPT1A downregulation contributes to radioresistance in CRC by augmenting the FOXM1-mediated antioxidant response. Thus, CPT1A is a potential biomarker of radiosensitivity and a novel target for overcoming radioresistance, offering a future direction to enhance CRC radiotherapy., Competing Interests: ZC, LY, ZZ, XW, QG, YC, YZ, YZ, JX, KC, HF, YD No competing interests declared, (© 2024, Chen, Yu, Zheng et al.)
- Published
- 2024
- Full Text
- View/download PDF
42. HMGB1 Modulates Macrophage Metabolism and Polarization in Ulcerative Colitis by Inhibiting Cpt1a Expression.
- Author
-
Wang F, Luo L, Wu Z, Wan L, Li F, and Wen Z
- Subjects
- Animals, Mice, RAW 264.7 Cells, Mice, Inbred C57BL, Male, Glycolysis, Dextran Sulfate toxicity, Macrophage Activation, Disease Models, Animal, HMGB1 Protein metabolism, HMGB1 Protein genetics, Colitis, Ulcerative metabolism, Colitis, Ulcerative genetics, Carnitine O-Palmitoyltransferase metabolism, Carnitine O-Palmitoyltransferase genetics, Macrophages metabolism
- Abstract
Background: Macrophage polarization is involved in the development of ulcerative colitis (UC). This study investigated the mechanism by which high mobility group box-1 protein (HMGB1) regulates macrophage polarization through metabolic reprogramming, thereby contributing to the pathogenesis of UC., Methods: Dextran sulfate sodium (DSS) was used to induce colitis in mice. RAW264.7 cells were polarized to M1 or M2 macrophages in vitro by stimulating with lipopolysaccharide (LPS)/interferon-γ (IFN-γ) or Interleukin-4 (IL-4), respectively. Macrophage infiltration and distribution within colon tissue were assessed by immunohistochemistry and flow cytometry. Glycolysis, fatty acid oxidation (FAO), and inflammatory factors were evaluated using relevant reagent kits. Chromatin Immunoprecipitation (ChIP) and luciferase reporter experiments were performed to study the regulation of Carnitine palmitoyltransferase 1A (Cpt1a) promoter transcriptional activity by HMGB1., Results: The mouse UC model showed upregulated HMGB1 and increased macrophage infiltration. Overexpression of HMGB1 promoted M1 macrophage polarization, increased glycolysis, and reduced FAO, whereas knockdown of HMGB1 promoted M2 macrophage polarization, reduced glycolysis, and increased FAO. HMGB1 negatively regulated Cpt1a expression by inhibiting transcription of the Cpt1a promoter. Knockdown of Cpt1a reversed the effects of small interfering RNA targeting HMGB1 (si-HMGB1) on macrophage metabolism and polarization. Administration of adeno-associated virus (AAV)-shHMGB1 in vivo caused a reduction in UC symptoms and inflammation., Conclusions: HMGB1 modulates macrophage metabolism in UC by inhibiting Cpt1a expression, leading to increased M1 polarization. This provides a theoretical basis for the clinical application of HMGB1 inhibitors in the treatment of UC., (© 2024 The Author(s). Published by IMR Press.)
- Published
- 2024
- Full Text
- View/download PDF
43. Overexpression CPT1A reduces lipid accumulation via PPARα/CD36 axis to suppress the cell proliferation in ccRCC
- Author
-
Yang Hui, Zhao Hongbo, Ren Zhongkun, Yi Xiaojia, Zhang Qiao, Yang Zhe, Kuang Yingmin, and Zhu Yuechun
- Subjects
CD36 ,cholesterol ,CPT1A ,growth ,PPARα ,renal cancer ,Biochemistry ,QD415-436 ,Genetics ,QH426-470 - Abstract
Clear cell renal carcinoma (ccRCC) is histologically defined by its cytoplasmic lipid deposits. Lipid metabolism disorder largely increases the risk of ccRCC. In this study, we aimed to investigate the biological functions and molecular mechanisms of carnitine palmitoyl transferase 1A (CPT1A) in ccRCC. Our results showed that CPT1A is decreased in ccRCC clinical samples and cell lines compared with that in normal samples. Lentivirus overexpressing CPT1A was used to investigate the neoplastic phenotypes of ccRCC, and the results showed that lipid accumulation and tumor growth are attenuated both in vitro and in vivo. In addition, CPT1A prevents cholesterol uptake and lipid accumulation by increasing the peroxisome proliferator-activated receptor α (PPARα) level through regulation of Class B scavenger receptor type 1 (SRB1) and cluster of differentiation 36 (CD36). Furthermore, PI3K/Akt signaling pathway promotes tumor cell proliferation in ccRCC, which is related to the enhanced expression of CD36. Functionally, weakened CPT1A expression is critical for lipid accumulation to promote ccRCC development. Collectively, our research unveiled a novel function of CPT1A in lipid metabolism via PPARα/CD36 axis, which provides a new theoretical explanation for the pathogenesis of ccRCC. Targeting CPT1A may be a potential therapeutic strategy to treat ccRCC.
- Published
- 2021
- Full Text
- View/download PDF
44. CPT1A promotes anoikis resistance in esophageal squamous cell carcinoma via redox homeostasis
- Author
-
Tian Tian, Yunxin Lu, Jinfei Lin, Miao Chen, Huijuan Qiu, Wancui Zhu, Haohui Sun, Jinsheng Huang, Han Yang, and Wuguo Deng
- Subjects
CPT1A ,Fatty acid oxidation ,Anoikis resistance ,Esophageal squamous cell carcinoma ,Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Anoikis resistance was a prominent hallmark of cancer metastasis, and lipo-genic characteristics have been identified as another metabolic alteration during tumorigenesis. However, their crosstalk has not been fully elucidated, especially in advanced esophageal squamous cell carcinoma (ESCC). In this study, we showed, for the first time, that the key enzyme carnitine O-palmitoyl transferase 1 (CPT1A), which is involved in fatty acid oxidation (FAO), was markedly upregulated in ESCC cells upon detached culture via a metabolism PCR array. Overexpression of CPT1A was associated with poor survival of ESCC patients and could protect ESCC cells from apoptosis via maintaining redox homeostasis through supply of GSH and NADPH. Mechanistically, detached culture conditions enhanced the expression of the transcription factor ETV4 and suppressed the expression of the ubiquitin enzyme RNF2, which were responsible for the elevated expression of CPT1A at the mRNA and protein levels, respectively. Moreover, genetic or pharmacologic disruption of CPT1A switched off the NADPH supply and therefore prevented the anchorage-independent growth of ESCC cells in vitro and lung metastases of xenografted tumor models in vivo. Collectively, our results provide novel insights into how ESCC cancer cells exploit metabolic switching to form distant metastases and some evidence for the link between anoikis and FAO.
- Published
- 2022
- Full Text
- View/download PDF
45. Downregulation of CPT1A exerts a protective effect in dextran sulfate sodium‐induced ulcerative colitis partially by inhibiting PPARα signaling pathway.
- Author
-
Chen, Wenxiao, Zou, Jinyan, Shi, Xinyuan, and Huang, Huifeng
- Subjects
- *
ULCERATIVE colitis , *DEXTRAN sulfate , *INFLAMMATORY bowel diseases , *CELLULAR signal transduction , *DOWNREGULATION , *PEROXISOME proliferator-activated receptors - Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease that may progress to colorectal cancer in severe cases. Carnitine palmitoyltransferase‐1A (CPT1A) has been reported to be upregulated in colorectal cancer. This paper aims to explore the role of CPT1A in UC and its pathogenesis. An in vivo mice model of UC was constructed by administrating 3% dextran sulfate sodium (DSS). The expression level of CPT1A was examined by quantitative real‐time polymerase chain reaction and Western blot. The intestinal damage, inflammatory response and oxidative stress were assessed by hematoxylin and eosin staining, colon length, and commercial kits. Thereafter, an in vitro cell model of UC was established by stimulating HT‐29 cells with 2% DSS. The peroxisome proliferator‐activated receptor α (PPARα) signaling agonist GW7647 was used for treatment. Cell viability and apoptosis was assayed by cell counting kit‐8 assay and terminal dUTP nick‐end labeling assay, respectively. The inflammatory cytokines and oxidative stress‐related factors was evaluated using corresponding commercial detection kits. In DSS‐induced mice model of UC, CPT1A expression was upregulated. Interference of CPT1A attenuated histological damage, the disease activity index and colon length in colitis. We also found downregulation of CPT1A inhibited inflammatory response and oxidative stress, and inhibited PPARα signaling pathway in UC mice. Additionally, in DSS‐induced HT‐29 cells, downregulation of CPT1A promoted cell viability, reduced cell apoptosis, inflammatory response, and oxidative stress, which was partly abolished by additional treatment with GW7647. In summary, downregulation of CPT1A exerts a protective effect in DSS‐induced UC partially through suppressing PPARα signaling, suggesting that CPT1A might be a potential target for the treatment of UC. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
46. Metabolic disease in the Pacific: Lessons for indigenous populations.
- Abstract
Twenty‐five percent of the New Zealand population is either Māori or Pacific and are thus indigenous to the region. The New Zealand National Metabolic Service has considerable experience in diagnosing and managing metabolic diseases in this population. The frequencies and phenotypes of inborn errors of metabolism in indigenous people differ from that in Western European populations. Metabolic services need to be aware of these local variations and adapt their screening and treatment protocols accordingly. Likewise, the services themselves need to adopt culturally appropriate practices. This includes an understanding of the language, ideally employment of indigenous people and targeting of the service to meet the needs of the people. Knowledge of the metabolic diseases common within particular ethnic groups is important for the rapid delivery of appropriate management. Newborn screening protocols need to reflect the local populations. With the advent of expanded newborn screening relatively benign forms of fatty acid oxidation disorders have been commonly encountered. This high prevalence may reflect a selective evolutionary advantage as similar conditions have been found in other ethnic groups with traditionally high fat and low carbohydrate diets. HLA haplotypes of indigenous populations are less represented in international stem cell transplant databanks thereby making the option of human stem cell transplant more challenging. The recent discovery that short‐chain enoyl‐CoA hydratase deficiency is particularly common in New Zealand with nearly a dozen cases identified this year suggests there is still a lot to learn regarding Māori and Pacific and indeed an indigenous metabolic disease. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
47. Knockdown of CPT1A Induce Chicken Adipocyte Differentiation to Form Lipid Droplets
- Author
-
ZQ Li, JJ Li, ZZ Lin, DH Zhang, GF Zhang, JS Ran, Y Wang, HD Yin, and YP Liu
- Subjects
Adipocyte differentiation ,CPT1A ,lipid droplets ,Animal culture ,SF1-1100 ,Veterinary medicine ,SF600-1100 ,Zoology ,QL1-991 - Abstract
ABSTRACT Lipid metabolism dysfunction is closely related to obesity, inflammation, diabetes, lipodystrophy, cardiovascular disease. Along with having a positive effect on energy homeostasis during fasting or prolonged exercise through mitochondrial fatty acid oxidation (FAO), more than two dozen enzymes and transport proteins are involved in the activation and transport of fatty acids into the mitochondrial, providing insights into their critical roles in metabolism. CPT1A has been reported to be expressed ubiquitously in the body and associated with dire consequences affecting fat deposition as the key rate-limiting enzyme of FAO. However, there is a dearth of data on the specific role of CPT1A on adipogenic differentiation and adipocyte lipolysis on chicken. This study assessed CPT1A’s function in adipocyte differentiation andadipocyte lipolysis, and the mechanisms were investigated. We found that CPT1A knockdown (KD) promotes the differentiation of chicken preadipocytes into mature adipocytes. CPT1A KD increased PPARγ protein expression level. Expression levels of lipid synthesis-related genes were increased, and lipolysis genes were reduced. Also, CPT1A KD can encourage the formation of lipid droplets. So our results confirmed that knockdown of CPT1A induced the lipid differentiation and inhibited the β-oxidation process to promote the formation of lipid droplets. These findings may deepen our understanding on CPT1A function, especially its regulatory role in adipocyte biology.
- Published
- 2022
- Full Text
- View/download PDF
48. Enhancing cancer‐associated fibroblast fatty acid catabolism within a metabolically challenging tumor microenvironment drives colon cancer peritoneal metastasis
- Author
-
Shaoyong Peng, Daici Chen, Jian Cai, Zixu Yuan, Binjie Huang, Yichen Li, Huaiming Wang, Qianxin Luo, Yingyi Kuang, Wenfeng Liang, Zhihang Liu, Qian Wang, Yanmei Cui, Hui Wang, and Xiaoxia Liu
- Subjects
CAF ,colorectal cancer ,CPT1A ,FAO ,glycolysis ,peritoneal metastases ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Most cancer‐related deaths result from the progressive growth of metastases. Patients with peritoneal metastatic (PM) colorectal cancer have reduced overall survival. Currently, it is still unclear why colorectal cancer (CRC) cells home to and proliferate inside the peritoneal cavity, and there is no effective consolidation therapy for improved survival. Using a proteomic approach, we found that key enzymes of fatty acid oxidation (FAO) were decreased in patients with PM colorectal cancer. Furthermore, we confirmed that carnitine palmitoyltransferase IA (CPT1A), a rate‐limiting enzyme of FAO, was expressed at significantly low levels in patients with PM colorectal cancer, as determined by RT‐qPCR, IHC, and GEO dataset analysis. However, lipidomics revealed no difference in FFA levels between PM and non‐PM primary tumors. Here, we showed that cancer‐associated fibroblasts (CAFs) promote the proliferation, migration, and invasion of colon cancer cells via upregulating CPT1A to actively oxidize FAs and conduct minimal glycolysis. In addition, coculture‐induced glycolysis increased in cancer cells while fatty acid catabolism decreased with lower adiponectin levels. Importantly, inhibition of glycolysis significantly reduced the survival of CRC cells after incubation with conditioned medium from CAFsCPT1A‐OE in vitro and impaired the survival and growth of organoids derived from CRC‐PM. Finally, we found that directly blocking FAO in CAFsCPT1A‐OE with etomoxir inhibits migration and invasion in vitro and decreases tumor growth and intraperitoneal dissemination in vivo, revealing a role for CAF CPT1A in promoting tumor growth and invasion. In conclusion, our results suggest the possibility of testing FAO inhibition as a novel approach and clinical strategy against CAF‐induced colorectal cancer with peritoneal dissemination/metastases.
- Published
- 2021
- Full Text
- View/download PDF
49. Cryptotanshinone alleviates liver fibrosis via inhibiting STAT3/CPT1A-dependent fatty acid oxidation in hepatic stellate cells.
- Author
-
Li, Zibo, Zheng, Yaqiu, Zhang, Lin, and Xu, Erping
- Subjects
- *
HEPATIC fibrosis , *FATTY acid oxidation , *LIVER cells , *ACETYLCOENZYME A , *BIOTRANSFORMATION (Metabolism) , *SALVIA miltiorrhiza - Abstract
Hepatic stellate cells (HSCs) are a major source of fibrogenic cells and play a central role in liver fibrogenesis. HSC activation depends on metabolic activation, for which it is well established that fatty acid oxidation (FAO) sustains their rapid proliferative rate. Studies have indicated that tanshinones inhibit HSC activation, however, the anti-fibrosis mechanisms of tanshinones are remain unclear. Herein, we reported that cryptotanshinone (CTS), a lipid-soluble ingredient of Salvia miltiorrhiza Bunge, exhibited the strongest inhibitory effects on HSC-LX2 proliferation and activation. CTS could induce lipocyte phenotype in mouse primary HSC and HSC-LX2. Transcriptomic sequencing and qPCR revealed that CTS regulated fatty acid metabolism and inhibited CPT1A and CPT1B expression. Target prediction suggested CTS regulates lipid metabolism by targeting STAT3. Mechanistically, the level of ATP and acetyl-CoA were reduced by the treatment of CTS, indicating that CTS could inhibit the level of FAO. Furthermore, CTS could inhibit the phosphorylation and nuclear translocation of STAT3. Additionally, CPT1A overexpression reversed the efficacy of CTS. Finally, CTS (40 mg/kg/day) attenuated CCl 4 -induced liver fibrosis and inhibited collagen production and HSC activation. Moreover, the results of immunofluorescence showed that α-SMA and p-STAT3 were co-located, and CTS could reduce the levels of p-STAT3 and α-SMA. In summary, CTS alleviated liver fibrosis by inhibiting the p-STAT3/CPT1A - dependent FAO both in vitro and in vivo , making it a potential candidate drug for the treatment of liver fibrosis. [Display omitted] • Cryptotanshinone alleviates CCl 4 -induced liver fibrosis by inhibiting the activation of hepatic stellate cells in mice. • STAT3/CPT1A-dependent fatty acid oxidation is essential for the activation of hepatic stellate cells. • Cryptotanshinone inhibits STAT3/CPT1A-dependent fatty acid oxidation in hepatic stellate cells. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
50. Inhibition of carnitine palmitoyltransferase 1A in hepatic stellate cells protects against fibrosis.
- Author
-
Fondevila, Marcos F., Fernandez, Uxia, Heras, Violeta, Parracho, Tamara, Gonzalez-Rellan, Maria J., Novoa, Eva, Porteiro, Begoña, Alonso, Cristina, Mayo, Rebeca, da Silva Lima, Natalia, Iglesias, Cristina, Filliol, Aveline A., Senra, Ana, Delgado, Teresa C., Woodhoo, Ashwin, Herrero, Laura, Serra, Dolors, Prevot, Vincent, Schwaninger, Markus, and López, Miguel
- Subjects
- *
CARNITINE palmitoyltransferase , *LIVER cells , *TRANSFORMING growth factors , *SATURATED fatty acids , *HEPATIC fibrosis , *CHOLINE - Abstract
The pathogenesis of liver fibrosis requires activation of hepatic stellate cells (HSCs); once activated, HSCs lose intracellular fatty acids but the role of fatty acid oxidation and carnitine palmitoyltransferase 1A (CPT1A) in this process remains largely unexplored. CPT1A was found in HSCs of patients with fibrosis. Pharmacological and genetic manipulation of CPT1A were performed in human HSC cell lines and primary HCSs. Finally, we induced fibrosis in mice lacking CPT1A specifically in HSCs. Herein, we show that CPT1A expression is elevated in HSCs of patients with non-alcoholic steatohepatitis, showing a positive correlation with the fibrosis score. This was corroborated in rodents with fibrosis, as well as in primary human HSCs and LX-2 cells activated by transforming growth factor β1 (TGFβ1) and fetal bovine serum (FBS). Furthermore, both pharmacological and genetic silencing of CPT1A prevent TGFβ1- and FBS-induced HSC activation by reducing mitochondrial activity. The overexpression of CPT1A, induced by saturated fatty acids and reactive oxygen species, triggers mitochondrial activity and the expression of fibrogenic markers. Finally, mice lacking CPT1A specifically in HSCs are protected against fibrosis induced by a choline-deficient high-fat diet, a methionine- and choline-deficient diet, or treatment with carbon tetrachloride. These results indicate that CPT1A plays a critical role in the activation of HSCs and is implicated in the development of liver fibrosis, making it a potentially actionable target for fibrosis treatment. We show that the enzyme carnitine palmitoyltransferase 1A (CPT1A) is elevated in hepatic stellate cells (HSCs) in patients with fibrosis and mouse models of fibrosis, and that CPT1A induces the activation of these cells. Inhibition of CPT1A ameliorates fibrosis by preventing the activation of HSCs. [Display omitted] • CPT1A is overexpressed in HSCs from patients with fibrosis and positively correlates with fibrosis and NAFLD activity score. • Fatty acid oxidation is increased in activated HSCs and CPT1A inhibition blunts HSC activation by reducing mitochondrial activity. • In experimental models, CPT1A-induced fibrogenesis is dependent on ATP availability. • The specific deletion of CPT1A in mouse HSCs protects against fibrosis. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.