Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Publication year range
1.
J Transl Med ; 22(1): 381, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38654380

ABSTRACT

BACKGROUND: Gastric cancer (GC) is a common and aggressive type of cancer worldwide. Despite recent advancements in its treatment, the prognosis for patients with GC remains poor. Understanding the mechanisms of cell death in GC, particularly those related to mitochondrial function, is crucial for its development and progression. However, more research is needed to investigate the significance of the interaction between mitochondrial function and GC cell death. METHODS: We employed a robust computational framework to investigate the role of mitochondria-associated proteins in the progression of GC in a cohort of 1,199 GC patients. Ten machine learning algorithms were utilized and combined into 101 unique combinations. Ultimately, we developed a Mitochondrial-related-Score (MitoScore) using the machine learning model that exhibited the best performance. We observed the upregulation of LEMT2 and further explored its function in tumor progression. Mitochondrial functions were assessed by measuring mitochondrial ATP, mitochondrial membrane potential, and levels of lactate, pyruvate, and glucose. RESULTS: MitoScore showed significant correlations with GC immune and metabolic functions. The higher MitoScore subgroup exhibited enriched metabolic pathways and higher immune activity. Overexpression of LETM2 (leucine zipper and EF-hand containing transmembrane protein 2) significantly enhanced tumor proliferation and metastasis. LETM2 plays a role in promoting GC cell proliferation by activating the mTOR pathway, maintaining mitochondrial homeostasis, and promoting glycolysis. CONCLUSION: The powerful machine learning framework highlights the significant potential of MitoScore in providing valuable insights and accurate assessments for individuals with GC. This study also enhances our understanding of LETM2 as an oncogene signature in GC. LETM2 may promote tumor progression by maintaining mitochondrial health and activating glycolysis, offering potential targets for diagnosis, treatment, and prognosis of GC.


Subject(s)
Machine Learning , Mitochondria , Stomach Neoplasms , Stomach Neoplasms/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Humans , Mitochondria/metabolism , Prognosis , Cohort Studies , Male , Female , Models, Biological , Cell Proliferation , Middle Aged , Gene Expression Regulation, Neoplastic , Membrane Proteins/metabolism , Multiomics
2.
Cancers (Basel) ; 14(19)2022 Sep 28.
Article in English | MEDLINE | ID: mdl-36230646

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the highest mortalities malignant tumors, which is characterized by difficult diagnosis, rapid progression and high recurrence rate. Nevertheless, PDAC responds poorly to conventional therapies, which highlights the urgency to identify novel prognostic and therapeutic targets. LEMT2 was a newly discovered protein-encoding gene with little cancer research and an unclear mechanism. Thus, this study aimed to illustrate LETM2 as the crucial oncogene for tumor progression in PDAC. In this study, we analyzed the expression level and prognostic value of LETM2 in multiple cancers using pan-cancer analysis. The analyses based on the TCGA-GTEx dataset indicated that the LETM2 expression was obviously elevated in several cancers, and it was the most significantly related to the dismal prognosis of PDAC. Subsequently, we demonstrated the functional role and mechanism of LETM2 by clinical sample evaluation, and in in vitro and in vivo experiments. Immunohistochemical analyses showed that high expression of LETM2 was correlated with poor outcomes of PDAC. Moreover, we demonstrated that LETM2 knockdown significantly inhibited tumor proliferation and metastasis, and promoted cell apoptosis, while LETM2 overexpression exerted the opposite effects. Finally, the impairment caused by LETM2-knockdown could be recovered via excitation of the PI3k-Akt pathway in vitro and in vivo animal models, which suggested that LETM2 could activate the downstream PI3K-Akt pathway to participate in PDAC progression. In conclusion, the study enhanced our understanding of LETM2 as an oncogene hallmark of PDAC. LETM2 may facilitate tumor progression by activating the PI3K-Akt signaling pathway, which provides potential targets for the diagnosis, treatment, and prognosis of pancreatic cancer.

3.
Int J Mol Sci ; 20(2)2019 Jan 12.
Article in English | MEDLINE | ID: mdl-30642051

ABSTRACT

Mitochondrial calcium (Ca2+) uptake shapes cytosolic Ca2+ signals involved in countless cellular processes and more directly regulates numerous mitochondrial functions including ATP production, autophagy and apoptosis. Given the intimate link to both life and death processes, it is imperative that mitochondria tightly regulate intramitochondrial Ca2+ levels with a high degree of precision. Among the Ca2+ handling tools of mitochondria, the leucine zipper EF-hand containing transmembrane protein-1 (LETM1) is a transporter protein localized to the inner mitochondrial membrane shown to constitute a Ca2+/H⁺ exchanger activity. The significance of LETM1 to mitochondrial Ca2+ regulation is evident from Wolf-Hirschhorn syndrome patients that harbor a haplodeficiency in LETM1 expression, leading to dysfunctional mitochondrial Ca2+ handling and from numerous types of cancer cells that show an upregulation of LETM1 expression. Despite the significance of LETM1 to cell physiology and pathophysiology, the molecular mechanisms of LETM1 function remain poorly defined. In this review, we aim to provide an overview of the current understanding of LETM1 structure and function and pinpoint the knowledge gaps that need to be filled in order to unravel the underlying mechanistic basis for LETM1 function.


Subject(s)
Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neoplasms/genetics , Wolf-Hirschhorn Syndrome/genetics , Calcium/metabolism , Calcium-Binding Proteins/chemistry , Haploinsufficiency , HeLa Cells , Humans , Membrane Proteins/chemistry , Mitochondria/metabolism , Models, Molecular , Neoplasms/metabolism , Protein Conformation , Up-Regulation , Wolf-Hirschhorn Syndrome/metabolism
4.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-513066

ABSTRACT

Objective:To analyze the expression of LETM2 in KYSE150 and ECA109 cell lines and its effect on the proliferation, migra-tion, and invasion of esophageal squamous cell carcinoma (ESCC). Methods:The expression level of the LETM2 protein in 90 paired hu-man ESCC tissues and matched adjacent normal tissues was determined through immunohistochemistry. The expression level of LETM2 in ESCC cell lines was detected by real-time PCR and Western blot. The expression levels of LETM2 in KYSE150 and ECA109 cell lines were knocked down using lentivirus. MTT assays were performed to examine the effect of LETM2 on the proliferation of ESCC cells. Colony formation assay was used to detect the colony formation ability. Flow cytometry was performed to analyze the cell cycle. The effect of LETM2 depletion on the migration and invasion of ESCC cells was determined by Transwell assay. Results:LETM2 expres-sion was frequently upregulated in the ESCC tissues than in the adjacent normal tissues. The suppressed exogenous expression of LETM2 led to the inhibition of cell proliferation and colony formation. However, cell migration and invasion were not affected. The re-sults on the cell cycle distribution revealed that LETM2 knockdown acts as a negative regulator of the cell cycle at the G1 to S phase transition. Conclusion:LETM2 acts as a tumor-driven gene in the development and progression of ESCC. This finding suggests that LETM2 can be used as an efficient prognosis biomarker and a potential therapeutic target for ESCC.

SELECTION OF CITATIONS
SEARCH DETAIL
...