Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Brief Bioinform ; 24(1)2023 01 19.
Article in English | MEDLINE | ID: mdl-36575567

ABSTRACT

Long noncoding ribonucleic acids (RNAs; LncRNAs) endowed with both protein-coding and noncoding functions are referred to as 'dual functional lncRNAs'. Recently, dual functional lncRNAs have been intensively studied and identified as involved in various fundamental cellular processes. However, apart from time-consuming and cell-type-specific experiments, there is virtually no in silico method for predicting the identity of dual functional lncRNAs. Here, we developed a deep-learning model with a multi-head self-attention mechanism, LncReader, to identify dual functional lncRNAs. Our data demonstrated that LncReader showed multiple advantages compared to various classical machine learning methods using benchmark datasets from our previously reported cncRNAdb project. Moreover, to obtain independent in-house datasets for robust testing, mass spectrometry proteomics combined with RNA-seq and Ribo-seq were applied in four leukaemia cell lines, which further confirmed that LncReader achieved the best performance compared to other tools. Therefore, LncReader provides an accurate and practical tool that enables fast dual functional lncRNA identification.


Subject(s)
RNA, Long Noncoding , RNA, Long Noncoding/genetics , RNA, Long Noncoding/chemistry , RNA-Seq
3.
Genes Dev ; 31(20): 2099-2112, 2017 10 15.
Article in English | MEDLINE | ID: mdl-29118048

ABSTRACT

Lung cancer is the leading cause of cancer deaths. Besides smoking, epidemiological studies have linked female sex hormones to lung cancer in women; however, the underlying mechanisms remain unclear. Here we report that the receptor activator of nuclear factor-kB (RANK), the key regulator of osteoclastogenesis, is frequently expressed in primary lung tumors, an active RANK pathway correlates with decreased survival, and pharmacologic RANK inhibition reduces tumor growth in patient-derived lung cancer xenografts. Clonal genetic inactivation of KRasG12D in mouse lung epithelial cells markedly impairs the progression of KRasG12D -driven lung cancer, resulting in a significant survival advantage. Mechanistically, RANK rewires energy homeostasis in human and murine lung cancer cells and promotes expansion of lung cancer stem-like cells, which is blocked by inhibiting mitochondrial respiration. Our data also indicate survival differences in KRasG12D -driven lung cancer between male and female mice, and we show that female sex hormones can promote lung cancer progression via the RANK pathway. These data uncover a direct role for RANK in lung cancer and may explain why female sex hormones accelerate lung cancer development. Inhibition of RANK using the approved drug denosumab may be a therapeutic drug candidate for primary lung cancer.


Subject(s)
Lung Neoplasms/metabolism , Receptor Activator of Nuclear Factor-kappa B/physiology , Alveolar Epithelial Cells/metabolism , Animals , Cell Respiration , Cells, Cultured , Energy Metabolism , Female , Gonadal Steroid Hormones/physiology , Homeostasis , Humans , Lung/metabolism , Lung Neoplasms/drug therapy , Male , Mice , Mitochondria/metabolism , Neoplastic Stem Cells/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Receptor Activator of Nuclear Factor-kappa B/antagonists & inhibitors , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptor Activator of Nuclear Factor-kappa B/metabolism , Respiratory Mucosa/metabolism
4.
Nature ; 563(7732): 564-568, 2018 11.
Article in English | MEDLINE | ID: mdl-30405245

ABSTRACT

Genetic regulators and environmental stimuli modulate T cell activation in autoimmunity and cancer. The enzyme co-factor tetrahydrobiopterin (BH4) is involved in the production of monoamine neurotransmitters, the generation of nitric oxide, and pain1,2. Here we uncover a link between these processes, identifying a fundamental role for BH4 in T cell biology. We find that genetic inactivation of GTP cyclohydrolase 1 (GCH1, the rate-limiting enzyme in the synthesis of BH4) and inhibition of sepiapterin reductase (the terminal enzyme in the synthetic pathway for BH4) severely impair the proliferation of mature mouse and human T cells. BH4 production in activated T cells is linked to alterations in iron metabolism and mitochondrial bioenergetics. In vivo blockade of BH4 synthesis abrogates T-cell-mediated autoimmunity and allergic inflammation, and enhancing BH4 levels through GCH1 overexpression augments responses by CD4- and CD8-expressing T cells, increasing their antitumour activity in vivo. Administration of BH4 to mice markedly reduces tumour growth and expands the population of intratumoral effector T cells. Kynurenine-a tryptophan metabolite that blocks antitumour immunity-inhibits T cell proliferation in a manner that can be rescued by BH4. Finally, we report the development of a potent SPR antagonist for possible clinical use. Our data uncover GCH1, SPR and their downstream metabolite BH4 as critical regulators of T cell biology that can be readily manipulated to either block autoimmunity or enhance anticancer immunity.


Subject(s)
Autoimmune Diseases/immunology , Biopterins/analogs & derivatives , Neoplasms/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Administration, Oral , Alcohol Oxidoreductases/antagonists & inhibitors , Alcohol Oxidoreductases/metabolism , Animals , Autoimmune Diseases/drug therapy , Autoimmune Diseases/pathology , Biopterins/biosynthesis , Biopterins/metabolism , Biopterins/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Coenzymes/metabolism , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Female , GTP Cyclohydrolase/genetics , GTP Cyclohydrolase/metabolism , Humans , Hypersensitivity/immunology , Iron/metabolism , Kynurenine/metabolism , Kynurenine/pharmacology , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
5.
Nucleic Acids Res ; 49(15): 8520-8534, 2021 09 07.
Article in English | MEDLINE | ID: mdl-34331449

ABSTRACT

With the dramatic development of single-cell RNA sequencing (scRNA-seq) technologies, the systematic decoding of cell-cell communication has received great research interest. To date, several in-silico methods have been developed, but most of them lack the ability to predict the communication pathways connecting the insides and outsides of cells. Here, we developed CellCall, a toolkit to infer inter- and intracellular communication pathways by integrating paired ligand-receptor and transcription factor (TF) activity. Moreover, CellCall uses an embedded pathway activity analysis method to identify the significantly activated pathways involved in intercellular crosstalk between certain cell types. Additionally, CellCall offers a rich suite of visualization options (Circos plot, Sankey plot, bubble plot, ridge plot, etc.) to present the analysis results. Case studies on scRNA-seq datasets of human testicular cells and the tumor immune microenvironment demonstrated the reliable and unique functionality of CellCall in intercellular communication analysis and internal TF activity exploration, which were further validated experimentally. Comparative analysis of CellCall and other tools indicated that CellCall was more accurate and offered more functions. In summary, CellCall provides a sophisticated and practical tool allowing researchers to decipher intercellular communication and related internal regulatory signals based on scRNA-seq data. CellCall is freely available at https://github.com/ShellyCoder/cellcall.


Subject(s)
Cell Communication/genetics , RNA, Small Cytoplasmic/genetics , Single-Cell Analysis , Transcription Factors , Algorithms , Base Sequence/genetics , Computational Biology , Gene Expression Regulation/genetics , Humans , Ligands , Sequence Analysis, RNA , Transcription Factors/genetics
6.
Nucleic Acids Res ; 48(7): 3816-3831, 2020 04 17.
Article in English | MEDLINE | ID: mdl-31996915

ABSTRACT

N 6-Methyladenosine (m6A) is the most abundant RNA modification in mammal mRNAs and increasing evidence suggests the key roles of m6A in human tumorigenesis. However, whether m6A, especially its 'reader' YTHDF1, targets a gene involving in protein translation and thus affects overall protein production in cancer cells is largely unexplored. Here, using multi-omics analysis for ovarian cancer, we identified a novel mechanism involving EIF3C, a subunit of the protein translation initiation factor EIF3, as the direct target of the YTHDF1. YTHDF1 augments the translation of EIF3C in an m6A-dependent manner by binding to m6A-modified EIF3C mRNA and concomitantly promotes the overall translational output, thereby facilitating tumorigenesis and metastasis of ovarian cancer. YTHDF1 is frequently amplified in ovarian cancer and up-regulation of YTHDF1 is associated with the adverse prognosis of ovarian cancer patients. Furthermore, the protein but not the RNA abundance of EIF3C is increased in ovarian cancer and positively correlates with the protein expression of YTHDF1 in ovarian cancer patients, suggesting modification of EIF3C mRNA is more relevant to its role in cancer. Collectively, we identify the novel YTHDF1-EIF3C axis critical for ovarian cancer progression which can serve as a target to develop therapeutics for cancer treatment.


Subject(s)
Eukaryotic Initiation Factor-3/genetics , Gene Expression Regulation, Neoplastic , Ovarian Neoplasms/genetics , Protein Biosynthesis , RNA-Binding Proteins/metabolism , Adenosine/analogs & derivatives , Animals , Carcinogenesis , Cell Line , Cell Line, Tumor , Cell Movement , Cell Proliferation , Eukaryotic Initiation Factor-3/biosynthesis , Female , Humans , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Oncogenes , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , RNA, Messenger/chemistry , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/physiology
7.
Cancer Cell Int ; 20: 423, 2020.
Article in English | MEDLINE | ID: mdl-32884446

ABSTRACT

[This corrects the article DOI: 10.1186/s12935-019-1045-1.].

8.
Cancer Cell Int ; 19: 321, 2019.
Article in English | MEDLINE | ID: mdl-31827395

ABSTRACT

BACKGROUND: Since FTO was recognized as the first m6A demethylase, the understanding of its biological function has been widely expanded. However, the role of FTO in cervical cancer tumorigenesis remains unclear. METHODS: In this study, we first analyzed the expression of FTO in two independent human cancer datasets and evaluated the correlation between FTO level and cervical cancer progression. Using small hairpin RNA technology, we explored the function of FTO in cervical cancer cell line Hela and SiHa cells, respectively. We then determined the FTO targets by performing transcriptional profile with FTO deficient and competent Hela cells, and finally validated these targets with ribosome profiling and functional rescue experiments. RESULTS: Our data suggested that FTO was frequently overexpressed in human cervical cancer tissues and highly correlated with cervical cancer progression. FTO serves as an oncogenic regulator for cervical cancer cells' proliferation and migration which is vastly depended on its demethylase activity. Mechanistically, FTO interacts with transcripts of E2F1 and Myc, inhibition of FTO significantly impairs the translation efficiency of E2F1 and Myc, however, either overexpress E2F1 or Myc sufficiently compensates the FTO deficiency which decreases cell proliferation and migration. CONCLUSIONS: Our study indicates that FTO plays important oncogenic role in regulating cervical cancer cells' proliferation and migration via controlling m6A modification of E2F1 and Myc transcripts. FTO represents a potential drug candidate for cervical cancer therapy.

9.
PLoS Genet ; 8(12): e1003071, 2012.
Article in English | MEDLINE | ID: mdl-23236288

ABSTRACT

The ability to perceive noxious stimuli is critical for an animal's survival in the face of environmental danger, and thus pain perception is likely to be under stringent evolutionary pressure. Using a neuronal-specific RNAi knock-down strategy in adult Drosophila, we recently completed a genome-wide functional annotation of heat nociception that allowed us to identify α2δ3 as a novel pain gene. Here we report construction of an evolutionary-conserved, system-level, global molecular pain network map. Our systems map is markedly enriched for multiple genes associated with human pain and predicts a plethora of novel candidate pain pathways. One central node of this pain network is phospholipid signaling, which has been implicated before in pain processing. To further investigate the role of phospholipid signaling in mammalian heat pain perception, we analysed the phenotype of PIP5Kα and PI3Kγ mutant mice. Intriguingly, both of these mice exhibit pronounced hypersensitivity to noxious heat and capsaicin-induced pain, which directly mapped through PI3Kγ kinase-dead knock-in mice to PI3Kγ lipid kinase activity. Using single primary sensory neuron recording, PI3Kγ function was mechanistically linked to a negative regulation of TRPV1 channel transduction. Our data provide a systems map for heat nociception and reinforces the extraordinary conservation of molecular mechanisms of nociception across different species.


Subject(s)
Drosophila , Gene Regulatory Networks , Nociceptive Pain , Phospholipids , Signal Transduction , Animals , Capsaicin/toxicity , Class Ib Phosphatidylinositol 3-Kinase/genetics , Class Ib Phosphatidylinositol 3-Kinase/physiology , Drosophila/genetics , Drosophila/physiology , Hot Temperature , Humans , Hypersensitivity/genetics , Mice , Neurons, Afferent/metabolism , Nociceptive Pain/chemically induced , Nociceptive Pain/genetics , Nociceptive Pain/physiopathology , Phospholipids/genetics , Phospholipids/metabolism , Phospholipids/physiology , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/physiology , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , TRPV Cation Channels/physiology
10.
Cancer Med ; 13(11): e7283, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38826095

ABSTRACT

BACKGROUND: Lung cancer remains the foremost reason of cancer-related mortality, with invasion and metastasis profoundly influencing patient prognosis. N-acetyltransferase 10 (NAT10) catalyzes the exclusive N (4)-acetylcytidine (ac4C) modification in eukaryotic RNA. NAT10 dysregulation is linked to various diseases, yet its role in non-small cell lung cancer (NSCLC) invasion and metastasis remains unclear. Our study delves into the clinical significance and functional aspects of NAT10 in NSCLC. METHODS: We investigated NAT10's clinical relevance using The Cancer Genome Atlas (TCGA) and a group of 98 NSCLC patients. Employing WB, qRT-PCR, and IHC analyses, we assessed NAT10 expression in NSCLC tissues, bronchial epithelial cells (BECs), NSCLC cell lines, and mouse xenografts. Further, knockdown and overexpression techniques (siRNA, shRNA, and plasmid) were employed to evaluate NAT10's effects. A series of assays were carried out, including CCK-8, colony formation, wound healing, and transwell assays, to elucidate NAT10's role in proliferation, invasion, and metastasis. Additionally, we utilized lung cancer patient-derived 3D organoids, mouse xenograft models, and Remodelin (NAT10 inhibitor) to corroborate these findings. RESULTS: Our investigations revealed high NAT10 expression in NSCLC tissues, cell lines and mouse xenograft models. High NAT10 level correlated with advanced T stage, lymph node metastasis and poor overall survive. NAT10 knockdown curtailed proliferation, invasion, and migration, whereas NAT10 overexpression yielded contrary effects. Furthermore, diminished NAT10 levels correlated with increased E-cadherin level whereas decreased N-cadherin and vimentin expressions, while heightened NAT10 expression displayed contrasting results. Notably, Remodelin efficiently attenuated NSCLC proliferation, invasion, and migration by inhibiting NAT10 through the epithelial-mesenchymal transition (EMT) pathway. CONCLUSIONS: Our data underscore NAT10 as a potential therapeutic target for NSCLC, presenting avenues for targeted intervention against lung cancer through NAT10 inhibition.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Cell Proliferation , Epithelial-Mesenchymal Transition , Lung Neoplasms , N-Terminal Acetyltransferase E , Humans , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Animals , Mice , N-Terminal Acetyltransferase E/metabolism , N-Terminal Acetyltransferase E/genetics , Male , Female , Disease Progression , Cell Movement , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , Xenograft Model Antitumor Assays , Mice, Nude , Middle Aged , N-Terminal Acetyltransferases
11.
Nat Cell Biol ; 26(3): 464-477, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38321204

ABSTRACT

Leukaemia stem cells (LSCs) in acute myeloid leukaemia present a considerable treatment challenge due to their resistance to chemotherapy and immunosurveillance. The connection between these properties in LSCs remains poorly understood. Here we demonstrate that inhibition of tyrosine phosphatase SHP-1 in LSCs increases their glycolysis and oxidative phosphorylation, enhancing their sensitivity to chemotherapy and vulnerability to immunosurveillance. Mechanistically, SHP-1 inhibition leads to the upregulation of phosphofructokinase platelet (PFKP) through the AKT-ß-catenin pathway. The increase in PFKP elevates energy metabolic activities and, as a consequence, enhances the sensitivity of LSCs to chemotherapeutic agents. Moreover, the upregulation of PFKP promotes MYC degradation and, consequently, reduces the immune evasion abilities of LSCs. Overall, our study demonstrates that targeting SHP-1 disrupts the metabolic balance in LSCs, thereby increasing their vulnerability to chemotherapy and immunosurveillance. This approach offers a promising strategy to overcome LSC resistance in acute myeloid leukaemia.


Subject(s)
Leukemia, Myeloid, Acute , Metabolic Reprogramming , Humans , Monitoring, Immunologic , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Stem Cells , Neoplastic Stem Cells/metabolism
12.
bioRxiv ; 2023 May 09.
Article in English | MEDLINE | ID: mdl-37214906

ABSTRACT

Infections with defined Herpesviruses, such as Pseudorabies virus (PRV) and Varicella zoster virus (VZV) can cause neuropathic itch, referred to as "mad itch" in multiple species. The underlying mechanisms involved in neuropathic "mad itch" are poorly understood. Here, we show that PRV infections hijack the RNA helicase DDX3X in sensory neurons to facilitate anterograde transport of the virus along axons. PRV induces re-localization of DDX3X from the cell body to the axons which ultimately leads to death of the infected sensory neurons. Inducible genetic ablation of Ddx3x in sensory neurons results in neuronal death and "mad itch" in mice. This neuropathic "mad itch" is propagated through activation of the opioid system making the animals "addicted to itch". Moreover, we show that PRV co-opts and diverts T cell development in the thymus via a sensory neuron-IL-6-hypothalamus-corticosterone stress pathway. Our data reveal how PRV, through regulation of DDX3X in sensory neurons, travels along axons and triggers neuropathic itch and immune deviations to initiate pathophysiological programs which facilitate its spread to enhance infectivity.

13.
bioRxiv ; 2023 May 08.
Article in English | MEDLINE | ID: mdl-37214873

ABSTRACT

Dopa-responsive dystonia (DRD) and Parkinson's disease (PD) are movement disorders caused by the dysfunction of nigrostriatal dopaminergic neurons. Identifying druggable pathways and biomarkers for guiding therapies is crucial due to the debilitating nature of these disorders. Recent genetic studies have identified variants of GTP cyclohydrolase-1 (GCH1), the rate-limiting enzyme in tetrahydrobiopterin (BH4) synthesis, as causative for these movement disorders. Here, we show that genetic and pharmacological inhibition of BH4 synthesis in mice and human midbrain-like organoids accurately recapitulates motor, behavioral and biochemical characteristics of these human diseases, with severity of the phenotype correlating with extent of BH4 deficiency. We also show that BH4 deficiency increases sensitivities to several PD-related stressors in mice and PD human cells, resulting in worse behavioral and physiological outcomes. Conversely, genetic and pharmacological augmentation of BH4 protects mice from genetically- and chemically induced PD-related stressors. Importantly, increasing BH4 levels also protects primary cells from PD-affected individuals and human midbrain-like organoids (hMLOs) from these stressors. Mechanistically, BH4 not only serves as an essential cofactor for dopamine synthesis, but also independently regulates tyrosine hydroxylase levels, protects against ferroptosis, scavenges mitochondrial ROS, maintains neuronal excitability and promotes mitochondrial ATP production, thereby enhancing mitochondrial fitness and cellular respiration in multiple preclinical PD animal models, human dopaminergic midbrain-like organoids and primary cells from PD-affected individuals. Our findings pinpoint the BH4 pathway as a key metabolic program at the intersection of multiple protective mechanisms for the health and function of midbrain dopaminergic neurons, identifying it as a potential therapeutic target for PD.

14.
Nat Med ; 11(8): 875-9, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16007097

ABSTRACT

During several months of 2003, a newly identified illness termed severe acute respiratory syndrome (SARS) spread rapidly through the world. A new coronavirus (SARS-CoV) was identified as the SARS pathogen, which triggered severe pneumonia and acute, often lethal, lung failure. Moreover, among infected individuals influenza such as the Spanish flu and the emergence of new respiratory disease viruses have caused high lethality resulting from acute lung failure. In cell lines, angiotensin-converting enzyme 2 (ACE2) has been identified as a potential SARS-CoV receptor. The high lethality of SARS-CoV infections, its enormous economic and social impact, fears of renewed outbreaks as well as the potential misuse of such viruses as biologic weapons make it paramount to understand the pathogenesis of SARS-CoV. Here we provide the first genetic proof that ACE2 is a crucial SARS-CoV receptor in vivo. SARS-CoV infections and the Spike protein of the SARS-CoV reduce ACE2 expression. Notably, injection of SARS-CoV Spike into mice worsens acute lung failure in vivo that can be attenuated by blocking the renin-angiotensin pathway. These results provide a molecular explanation why SARS-CoV infections cause severe and often lethal lung failure and suggest a rational therapy for SARS and possibly other respiratory disease viruses.


Subject(s)
Carboxypeptidases/metabolism , Lung Diseases/enzymology , Membrane Glycoproteins/metabolism , Severe Acute Respiratory Syndrome/enzymology , Severe acute respiratory syndrome-related coronavirus/metabolism , Viral Envelope Proteins/metabolism , Analysis of Variance , Angiotensin-Converting Enzyme 2 , Animals , Immunohistochemistry , Lung Diseases/etiology , Lung Diseases/pathology , Membrane Glycoproteins/genetics , Mice , Peptidyl-Dipeptidase A , Severe acute respiratory syndrome-related coronavirus/genetics , Severe Acute Respiratory Syndrome/complications , Spike Glycoprotein, Coronavirus , Viral Envelope Proteins/genetics
15.
Immunobiology ; 227(6): 152296, 2022 11.
Article in English | MEDLINE | ID: mdl-36327543

ABSTRACT

Aerobic exercise is well recognized to be beneficial to physical and mental health. Many studies have shown that aerobic exercise can improve the human immune system, but whether it could affect lung regeneration and inflammation remained unclear. Bronchioloalveolar stem cells (BASCs) play a key role in lung regeneration and repair, but it is unclear whether aerobic exercise affects BASCs. Here, we randomly divided 8 weeks old male mice into three groups: the control group without any aerobic exercise; the rest group which received 2 weeks of aerobic exercise (running wheel training) plus 5 days' rest, and the exercise group which received 2 weeks of aerobic exercise without any rest. Our data indicated that mice in the exercise group had significantly increased BASCs compared to the control group, such difference did not exist in the rest group. Furthermore, the immune profiling suggested that lung inflammation was slightly up-regulated in the exercise group, particularly the inflammatory monocytes and IL-17A+ T cells. In conclusion, we provide direct evidence showing that aerobic exercise can facilitate lung regeneration with mild inflammatory effect, this finding is of great importance in the current COVID-19 pandemic.


Subject(s)
COVID-19 , Pandemics , Mice , Humans , Male , Animals , Lung , Inflammation , Regeneration
16.
Cancer Med ; 11(19): 3700-3713, 2022 10.
Article in English | MEDLINE | ID: mdl-35608274

ABSTRACT

BACKGROUND: Esophageal carcinoma is one of the most fatal cancers worldwide. In China, over 90% of esophageal cancer patients are diagnosed with esophageal squamous cell carcinoma (ESCC). Currently, the survival and prognosis of ESCC patients are not satisfying because of insufficient early screening and lack of efficacious medication. Accumulating studies have suggested that antibody-drug conjugates (ADC) represent a promising antitumor strategy. METHOD: Here, we carried out a specific membrane proteome screening with ESCC patients' samples using a high-throughput antibody microarray to uncover potential targets for ADC development. Candidates were validated with expression and cytotoxicity evaluation both in vitro and in vivo. RESULTS: Our data have shown that the Piezo-Type Mechanosensitive Ion Channel Component 1 (PIEZO1) is particularly overexpressed in human ESCC tumors and can be efficiently internalized when bound with its monoclonal antibody. Furthermore, the PIEZO1 antibody combined with monomethyl auristatin E (MMAE) can specifically kill PIEZO1 high-expressed ESCC tumor cells by inducing cell cycle arrest and apoptosis. More importantly, the Anti-PIEZO1-MMAE can significantly suppress tumor progression in ESCC xenograft tumor models without any obvious side effects. CONCLUSION: Taken together, our work demonstrates that PIEZO1 is a promising target to develop ADCs for human ESCC treatment, providing a new strategy for ESCC patients' personalized therapy.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Immunoconjugates , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Apoptosis , Cell Line, Tumor , Cell Proliferation , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/drug therapy , Humans , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Ion Channels , Proteome/metabolism , Xenograft Model Antitumor Assays
17.
Nat Commun ; 13(1): 7335, 2022 11 28.
Article in English | MEDLINE | ID: mdl-36443325

ABSTRACT

Type 2 diabetes mellitus is one of the most prevalent metabolic diseases presenting with systemic pathologies, including reproductive disorders in male diabetic patients. However, the molecular mechanisms that contributing to spermatogenesis dysfunction in diabetic patients have not yet been fully elucidated. Here, we perform STRT-seq to examine the transcriptome of diabetic patients' testes at single-cell resolution including all major cell types of the testis. Intriguingly, whereas spermatogenesis appears largely preserved, the gene expression profiles of Sertoli cells and the blood-testis barrier (BTB) structure are dramatically impaired. Among these deregulate pathways, the Apelin (APLN) peptide/Apelin-receptor (APJ) axis is hyper-activated in diabetic patients' testes. Mechanistically, APLN is produced locally by Sertoli cells upon high glucose treatment, which subsequently suppress the production of carnitine and repress the expression of cell adhesion genes in Sertoli cells. Together, these effects culminate in BTB structural dysfunction. Finally, using the small molecule APLN receptor antagonist, ML221, we show that blocking APLN/APJ significantly ameliorate the BTB damage and, importantly, improve functional spermatogenesis in diabetic db/db mice. We also translate and validate these findings in cultured human testes. Our findings identify the APLN/APJ axis as a promising therapeutic target to improve reproduction capacity in male diabetic patients.


Subject(s)
Blood-Testis Barrier , Diabetes Mellitus, Type 2 , Animals , Humans , Male , Mice , Apelin , Apelin Receptors/genetics , Spermatogenesis , Testis
18.
Sci Transl Med ; 14(660): eabj1531, 2022 08 31.
Article in English | MEDLINE | ID: mdl-36044597

ABSTRACT

Increased tetrahydrobiopterin (BH4) generated in injured sensory neurons contributes to increased pain sensitivity and its persistence. GTP cyclohydrolase 1 (GCH1) is the rate-limiting enzyme in the de novo BH4 synthetic pathway, and human single-nucleotide polymorphism studies, together with mouse genetic modeling, have demonstrated that decreased GCH1 leads to both reduced BH4 and pain. However, little is known about the regulation of Gch1 expression upon nerve injury and whether this could be modulated as an analgesic therapeutic intervention. We performed a phenotypic screen using about 1000 bioactive compounds, many of which are target-annotated FDA-approved drugs, for their effect on regulating Gch1 expression in rodent injured dorsal root ganglion neurons. From this approach, we uncovered relevant pathways that regulate Gch1 expression in sensory neurons. We report that EGFR/KRAS signaling triggers increased Gch1 expression and contributes to neuropathic pain; conversely, inhibiting EGFR suppressed GCH1 and BH4 and exerted analgesic effects, suggesting a molecular link between EGFR/KRAS and pain perception. We also show that GCH1/BH4 acts downstream of KRAS to drive lung cancer, identifying a potentially druggable pathway. Our screen shows that pharmacologic modulation of GCH1 expression and BH4 could be used to develop pharmacological treatments to alleviate pain and identified a critical role for EGFR-regulated GCH1/BH4 expression in neuropathic pain and cancer in rodents.


Subject(s)
Lung Neoplasms , Neuralgia , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Biopterins/analogs & derivatives , ErbB Receptors/genetics , ErbB Receptors/metabolism , GTP Cyclohydrolase/genetics , GTP Cyclohydrolase/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mice , Neuralgia/drug therapy , Neuralgia/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism
19.
Nat Cell Biol ; 24(8): 1278-1290, 2022 08.
Article in English | MEDLINE | ID: mdl-35927451

ABSTRACT

METTL3 encodes the predominant catalytic enzyme to promote m6A methylation in nucleus. Recently, accumulating evidence has shown the expression of METTL3 in cytoplasm, but its function is not fully understood. Here we demonstrated an m6A-independent mechanism for METTL3 to promote tumour progression. In gastric cancer, METTL3 could not only facilitate cancer progression via m6A modification, but also bind to numerous non-m6A-modified mRNAs, suggesting an unexpected role of METTL3. Mechanistically, cytoplasm-anchored METTL3 interacted with PABPC1 to stabilize its association with cap-binding complex eIF4F, which preferentially promoted the translation of epigenetic factors without m6A modification. Clinical investigation showed that cytoplasmic distributed METTL3 was highly correlated with gastric cancer progression, and this finding could be expanded to prostate cancer. Therefore, the cytoplasmic METTL3 enhances the translation of epigenetic mRNAs, thus serving as an oncogenic driver in cancer progression, and METTL3 subcellular distribution can assist diagnosis and predict prognosis for patients with cancer.


Subject(s)
Methyltransferases , Stomach Neoplasms , Adenosine/metabolism , Carcinogenesis/genetics , Epigenesis, Genetic , Humans , Male , Methyltransferases/genetics , Methyltransferases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stomach Neoplasms/genetics
20.
Nature ; 436(7047): 112-6, 2005 Jul 07.
Article in English | MEDLINE | ID: mdl-16001071

ABSTRACT

Acute respiratory distress syndrome (ARDS), the most severe form of acute lung injury, is a devastating clinical syndrome with a high mortality rate (30-60%) (refs 1-3). Predisposing factors for ARDS are diverse and include sepsis, aspiration, pneumonias and infections with the severe acute respiratory syndrome (SARS) coronavirus. At present, there are no effective drugs for improving the clinical outcome of ARDS. Angiotensin-converting enzyme (ACE) and ACE2 are homologues with different key functions in the renin-angiotensin system. ACE cleaves angiotensin I to generate angiotensin II, whereas ACE2 inactivates angiotensin II and is a negative regulator of the system. ACE2 has also recently been identified as a potential SARS virus receptor and is expressed in lungs. Here we report that ACE2 and the angiotensin II type 2 receptor (AT2) protect mice from severe acute lung injury induced by acid aspiration or sepsis. However, other components of the renin-angiotensin system, including ACE, angiotensin II and the angiotensin II type 1a receptor (AT1a), promote disease pathogenesis, induce lung oedemas and impair lung function. We show that mice deficient for Ace show markedly improved disease, and also that recombinant ACE2 can protect mice from severe acute lung injury. Our data identify a critical function for ACE2 in acute lung injury, pointing to a possible therapy for a syndrome affecting millions of people worldwide every year.


Subject(s)
Carboxypeptidases/metabolism , Lung Diseases/enzymology , Lung Diseases/pathology , Acids/pharmacology , Acute Disease , Analysis of Variance , Angiotensin II/metabolism , Angiotensin-Converting Enzyme 2 , Animals , Capillary Permeability , Carboxypeptidases/deficiency , Carboxypeptidases/genetics , Disease Models, Animal , Gene Deletion , Humans , Lung/pathology , Lung/physiopathology , Lung Diseases/genetics , Lung Diseases/physiopathology , Mice , Mice, Knockout , Mice, Transgenic , Oxygen/blood , Peptidyl-Dipeptidase A , Pulmonary Edema/pathology , Pulmonary Edema/physiopathology , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/metabolism , Respiratory Distress Syndrome/enzymology , Respiratory Distress Syndrome/genetics , Respiratory Distress Syndrome/pathology , Sepsis/enzymology , Sepsis/genetics , Sepsis/pathology , Transgenes/genetics
SELECTION OF CITATIONS
SEARCH DETAIL