704 results on '"ADP-Ribosylation Factor 6"'
Search Results
2. DDR1 promotes hepatocellular carcinoma metastasis through recruiting PSD4 to ARF6
- Author
-
Xiaochao Zhang, Yabing Hu, Yonglong Pan, Yixiao Xiong, Yuxin Zhang, Mengzhen Han, Keshuai Dong, Jia Song, Huifang Liang, Zeyang Ding, Xuewu Zhang, He Zhu, Qiumeng Liu, Xun Lu, Yongdong Feng, Xiaoping Chen, Zhanguo Zhang, and Bixiang Zhang
- Subjects
Cancer Research ,Carcinoma, Hepatocellular ,Discoidin Domain Receptor 1 ,ADP-Ribosylation Factor 6 ,MAP Kinase Signaling System ,Liver Neoplasms ,Genetics ,Guanine Nucleotide Exchange Factors ,Humans ,Receptor Protein-Tyrosine Kinases ,neoplasms ,Molecular Biology ,digestive system diseases - Abstract
Discoidin domain receptor 1 (DDR1) is a member of the receptor tyrosine kinase family, and its ligand is collagen. Previous studies demonstrated that DDR1 is highly expressed in many tumors. However, its role in hepatocellular carcinoma (HCC) remains obscure. In this study, we found that DDR1 was upregulated in HCC tissues, and the expression of DDR1 in TNM stage II-IV was higher than that in TNM stage I in HCC tissues, and high DDR1 expression was associated with poor prognosis. Gene expression analysis showed that DDR1 target genes were functionally involved in HCC metastasis. DDR1 positively regulated the migration and invasion of HCC cells and promoted lung metastasis. Human Phospho-Kinase Array showed that DDR1 activated ERK/MAPK signaling pathway. Mechanically, DDR1 interacted with ARF6 and activated ARF6 through recruiting PSD4. The kinase activity of DDR1 was required for ARF6 activation and its role in metastasis. High expression of PSD4 was associated with poor prognosis in HCC. In summary, our findings indicate that DDR1 promotes HCC metastasis through collagen induced DDR1 signaling mediated PSD4/ARF6 signaling, suggesting that DDR1 and ARF6 may serve as novel prognostic biomarkers and therapeutic targets for metastatic HCC.
- Published
- 2022
3. ARNO is recruited by the neuronal adaptor FE65 to potentiate ARF6-mediated neurite outgrowth
- Author
-
Yuqi Zhai, Wai Wa Ray Chan, Wen Li, and Kwok-Fai Lau
- Subjects
Neurons ,ADP-Ribosylation Factor 6 ,ADP-Ribosylation Factors ,Nucleotides ,General Neuroscience ,Immunology ,GTPase-Activating Proteins ,Neuronal Outgrowth ,Guanine Nucleotide Exchange Factors ,General Biochemistry, Genetics and Molecular Biology - Abstract
ADP-ribosylation factor 6 (ARF6) is a small GTPase that has a variety of neuronal functions including stimulating neurite outgrowth, a crucial process for the establishment and maintenance of neural connectivity. As impaired and atrophic neurites are often observed in various brain injuries and neurological diseases, understanding the intrinsic pathways that stimulate neurite outgrowth may provide insights into developing strategies to trigger the reconnection of injured neurons. The neuronal adaptor FE65 has been shown to interact with ARF6 and potentiate ARF6-mediated neurite outgrowth. However, the precise mechanism that FE65 activates ARF6 remains unclear, as FE65 does not possess a guanine nucleotide exchange factor (GEF) domain/function. Here, we show that FE65 interacts with the ARF6 GEF, namely the ARF nucleotide-binding site opener (ARNO). Moreover, a complex consisting of ARNO, ARF6 and FE65 is detected. Notably, FE65 potentiates the stimulatory effect of ARNO on ARF6-mediated neurite outgrowth, and the effect of FE65 is abrogated by an FE65 mutation that disrupts FE65–ARNO interaction. Additionally, the intramolecular interaction for mediating the autoinhibited conformation of ARNO is attenuated by FE65. Moreover, FE65 potentiates the effects of wild-type ARNO, but not the monomeric mutant, suggesting an association between FE65 and ARNO dimerization. Collectively, we demonstrate that FE65 binds to and activates ARNO and, consequently, potentiates ARF6-mediated neurite outgrowth.
- Published
- 2022
4. AMPK promotes Arf6 activation in a kinase-independent manner upon glucose starvation
- Author
-
Kuan-Jung Chen, Jia-Wei Hsu, and Fang-Jen S. Lee
- Subjects
Glucose ,ADP-Ribosylation Factor 6 ,ADP-Ribosylation Factors ,Guanine Nucleotide Exchange Factors ,Cell Biology ,AMP-Activated Protein Kinases - Abstract
AMP-activated protein kinase (AMPK) is a crucial cellular nutrient and energy sensor that maintains energy homeostasis. AMPK also governs cancer cell invasion and migration by regulating gene expression and activating multiple cellular signaling pathways. ADP-ribosylation factor 6 (Arf6) can be activated via nucleotide exchange by guanine-nucleotide-exchange factors (GEFs), and its activation also regulates tumor invasion and migration. By studying GEF-mediated Arf6 activation, we have elucidated that AMPK functions as a noncanonical GEF for Arf6 in a kinase-independent manner. Moreover, by examining the physiological role of the AMPK–Arf6 axis, we have determined that AMPK activates Arf6 upon glucose starvation and 5-aminoimidazole-4-carboxamide-1-β-D-ribofuranoside (AICAR) treatment. We have further identified the binding motif in the C-terminal regulatory domain of AMPK that is responsible for promoting Arf6 activation and, thus, inducing cell migration and invasion. These findings reveal a noncanonical role of AMPK in which its C-terminal regulatory domain serves as a GEF for Arf6 during glucose deprivation.
- Published
- 2022
5. MiR-1299 functions as a tumor suppressor to inhibit the proliferation and metastasis of gastric cancer by targeting ARF6
- Author
-
Yang Qiu, Ping Luo, and Yonggang Yuan
- Subjects
Cell ,Apoptosis ,Biology ,Biochemistry ,law.invention ,Metastasis ,Stomach Neoplasms ,law ,microRNA ,Genetics ,medicine ,Humans ,3' Untranslated Regions ,Molecular Biology ,Cell Proliferation ,Cell growth ,Cancer ,medicine.disease ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,medicine.anatomical_structure ,ADP-Ribosylation Factor 6 ,Cell culture ,Cancer research ,Suppressor - Abstract
MiRNAs belong to non-coding RNAs that are involved in cancer development. Acting as a mediator, they could regulate the expression level of numerous gens. However, the expression and function of miR-1299 in gastric cancer (GC) are not clear. To explore the role of miR-1299 in the process of GC. We detected the levels of miR-1299 in clinical samples of GC and investigated the role of miR-1299 in the regulation of the GC cells proliferation, apoptosis and metastasis. Luciferase reporter assay was employed to verify the target of miR-1299. Additionally, the proliferation, apoptosis and metastasis of AGS and SGC7901 cells were analyzed after the overexpression of miR-1299. Our study showed the expression of miR-1299 was decreased in GC tissues and cell lines. It indicated that the cell proliferation, migration and invasion was inhibited, while the cell apoptosis was promoted by the over-expressed miR-1299. Also, we found that miR-1299 could directly target the 3′-untranslated region (3′UTR) of ARF6 genes. In addition, rescue assay demonstrated that miR-1299 overexpression promoted the cell apoptosis and inhibited cell growth, which could be attenuated by the overexpression of ARF6. These findings indicate that miR-1299 regulates cell progression in GC by targeting ARF6 genes, and suggest that miR-1299 may be a tumor suppressor in the GC progression.
- Published
- 2021
6. Insulin stimulates atypical protein kinase C-mediated phosphorylation of the neuronal adaptor FE65 to potentiate neurite outgrowth by activating ARF6-Rac1 signaling
- Author
-
Dennis Dik‐Long Chau, Wen Li, Wai Wa Ray Chan, Jacquelyne Ka‐Li Sun, Yuqi Zhai, Hei‐Man Chow, and Kwok‐Fai Lau
- Subjects
Neurons ,rac1 GTP-Binding Protein ,Neuronal Outgrowth ,Neuropeptides ,Nuclear Proteins ,Nerve Tissue Proteins ,Biochemistry ,Mice ,Glucose ,ADP-Ribosylation Factor 6 ,Genetics ,Neurites ,Serine ,Animals ,Insulin ,Phosphorylation ,Molecular Biology ,Protein Kinase C ,Biotechnology - Abstract
Neurite outgrowth is a fundamental process in neurons that produces extensions and, consequently, neural connectivity. Neurite damage and atrophy are observed in various brain injuries and disorders. Understanding the intrinsic pathways of neurite outgrowth is essential for developing strategies to stimulate neurite regeneration. Insulin is a pivotal hormone in the regulation of glucose homeostasis. There is increasing evidence for the neurotrophic functions of insulin, including the induction of neurite outgrowth. However, the associated mechanism remains elusive. Here, we demonstrate that insulin potentiates neurite outgrowth mediated by the small GTPases ADP-ribosylation factor 6 (ARF6) and Ras-related C3 botulinum toxin substrate 1 (Rac1) through the neuronal adaptor FE65. Moreover, insulin enhances atypical protein kinase Cι/λ (PKCι/λ) activation and FE65 phosphorylation at serine 459 (S459) in neurons and mouse brains. In vitro and cellular assays show that PKCι/λ phosphorylated FE65 at S459. Consistently, insulin potentiates FE65 S459 phosphorylation only in the presence of PKCι/λ. Phosphomimetic studies show that an FE65 S459E mutant potently activates ARF6, Rac1, and neurite outgrowth. Notably, this phosphomimetic mutation enhances the FE65-ARF6 interaction, a process that promotes ARF6-Rac1-mediated neurite outgrowth. Likewise, insulin treatment and PKCι/λ overexpression potentiate the FE65-ARF6 interaction. Conversely, PKCι/λ knockdown suppresses the stimulatory effect of FE65 on ARF6-Rac1-mediated neurite outgrowth. The effect of insulin on neurite outgrowth is also markedly attenuated in PKCι/λ knockdown neurons, in the presence and absence of FE65. Our findings reveal a novel mechanism linking insulin with ARF6-Rac1-dependent neurite extension through the PKCι/λ-mediated phosphorylation of FE65.
- Published
- 2022
7. [Arf6 regulates endometriotic epithelial-mesenchymal transition and mitochondrial distribution]
- Author
-
Y C, Chen, Q M, Wang, L, Chen, M H, Zhu, and J, Zhang
- Subjects
Endometrium ,Epithelial-Mesenchymal Transition ,ADP-Ribosylation Factor 6 ,Cell Movement ,Proto-Oncogene Proteins ,Endometriosis ,Humans ,Vimentin ,Female ,Protein-Tyrosine Kinases ,RNA, Small Interfering ,Cadherins ,Child - Published
- 2022
8. Site-Specific
- Author
-
Jiarong, Wang, Bo, Wu, Youjia, Zhang, Liang, Ge, and Junfeng, Wang
- Subjects
Magnetic Resonance Spectroscopy ,ADP-Ribosylation Factor 6 ,Guanine Nucleotide Exchange Factors ,Monomeric GTP-Binding Proteins ,Signal Transduction - Abstract
Guanine nucleotide exchange factors (GEFs) of small GTPase (sGTPase) coordinate signal networks in normal cells and dysfunction in cancer. Therefore, effective monitoring of GEF activity is very important for studying the regulation of sGTPase signal transduction. In this study, we developed a 1D
- Published
- 2022
9. Pals1 prevents Rac1-dependent colorectal cancer cell metastasis by inhibiting Arf6
- Author
-
Albrecht Schwab, Christin Emming, Zoltán Pethő, Rita Schröter, Anja K. Wege, Pavel I. Nedvetsky, Julia Harms, Justine Guske, Simona Mareike Lüttgenau, Ute Neugebauer, Michael P. Krahn, Katrin Weber, Olga Panichkina, Thomas Wagner, and Florian Weber
- Subjects
0301 basic medicine ,rac1 GTP-Binding Protein ,Cancer Research ,Colorectal cancer ,Cell ,RAC1 ,Biology ,Metastasis ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Downregulation and upregulation ,Cell Movement ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,Cell migration ,Arf6 ,Letter to the Editor ,Cytoskeleton ,RC254-282 ,Cancer ,Membrane Proteins ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,HCT116 Cells ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Pals1 ,ADP-Ribosylation Factor 6 ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Molecular Medicine ,Heterografts ,Colorectal Neoplasms ,Nucleoside-Phosphate Kinase ,Rac1 - Abstract
Loss of apical-basal polarity and downregulation of cell-cell contacts is a critical step during the pathogenesis of cancer. Both processes are regulated by the scaffolding protein Pals1, however, it is unclear whether the expression of Pals1 is affected in cancer cells and whether Pals1 is implicated in the pathogenesis of the disease. Using mRNA expression data and immunostainings of cancer specimen, we show that Pals1 is frequently downregulated in colorectal cancer, correlating with poorer survival of patients. We further found that Pals1 prevents cancer cell metastasis by controlling Rac1-dependent cell migration through inhibition of Arf6, which is independent of the canonical binding partners of Pals1. Loss of Pals1 in colorectal cancer cells results in increased Arf6 and Rac1 activity, enhanced cell migration and invasion in vitro and increased metastasis of transplanted tumor cells in mice. Thus, our data reveal a new function of Pals1 as a key inhibitor of cell migration and metastasis of colorectal cancer cells. Notably, this new function is independent of the known role of Pals1 in tight junction formation and apical-basal polarity. Supplementary Information The online version contains supplementary material available at 10.1186/s12943-021-01354-2.
- Published
- 2021
10. microRNA-145-5p Inhibits Migration, Invasion, and Metastasis in Hepatocellular Carcinoma by Inhibiting ARF6
- Author
-
Wang S, Wang T, and Gu P
- Subjects
proliferation ,microrna-145-5p ,metastasis ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,adp-ribosylation factor 6 ,hepatocellular carcinoma ,invasion ,digestive system diseases ,RC254-282 - Abstract
Shuo Wang,1 Tianjiao Wang,2 Pengcheng Gu1 1Department of Orthopedics Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, People’s Republic of China; 2Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, Zhejiang, 310051, People’s Republic of ChinaCorrespondence: Pengcheng GuDepartment of Orthopedics Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, Zhejiang, 310006, People’s Republic of ChinaTel +86-571-87236848Email 1192014@zju.edu.cnPurpose: Hepatocellular carcinoma (HCC) has the fourth highest rate of mortality among the different types of cancer worldwide. This study aimed to investigate the functions of microRNA-145-5p and AFR6 on migration, invasion and metastasis in HCC.Methods: A total of 150 pairs of tumor and their matched adjacent nontumor liver tissues were collected from HCC patients. Expressions of microRNA-145-5p and AFR6 were measured by real-time PCR in HCC tissues and in HCC cell lines. The correlations between microRNA-145-5p and HCC prognosis were investigated. The proliferation, migration, invasion, cell cycle progression, and apoptosis of HCCLM3 cells were evaluated with CCK8, wound healing, transwell, and flow cytometric experiments.Results: The expression of miR-145-5p was confirmed to be downregulated not only in HCC tissues but also in several HCC cell lines compared with normal controls. A low expression level of miR-145-5p was notably associated with poor prognosis in patients with HCC and certain characteristics of metastatic tumors. In vitro, miR-145-5p negatively regulated cell proliferation, migration, and invasion and induced apoptosis in HCCLM3 cells. Subsequent experiments further verified that ARF6 is a novel target of miR-145-5p and is significantly overexpressed in HCC tissues. Overexpression of ARF6 circumvented the effects of miR-145-5p in HCCLM3 cells.Conclusion: miR-145-5p may play a pivotal role in HCC metastasis via regulating ARF6, and these findings may both provide further insights into the key factors of HCC metastasis and prove to be useful in the development of novel treatment options for HCC.Keywords: microRNA-145-5p, ADP-ribosylation factor 6, hepatocellular carcinoma, proliferation, invasion, metastasis
- Published
- 2021
11. Arf1 and Arf6 Synergistically Maintain Survival of T Cells during Activation
- Author
-
Yuki Ikuta, Yasunori Kanaho, Satoshi Matsuda, Madoka Ozawa, Kana Bando, Yui Kotani, Mami Sumiyoshi, Takaya Abe, Kazutomo Suzue, Shigeo Koyasu, Tomoya Katakai, Toshio Watanabe, and Taketo Yamada
- Subjects
Encephalomyelitis, Autoimmune, Experimental ,Cell Survival ,T-Lymphocytes ,T cell ,Immunology ,Receptors, Antigen, T-Cell ,Apoptosis ,Mice, Transgenic ,Biology ,Lymphocyte Activation ,Mice ,Immune system ,medicine ,Animals ,Immunology and Allergy ,Cells, Cultured ,ADP-Ribosylation Factors ,T-cell receptor ,Experimental autoimmune encephalomyelitis ,Colitis ,medicine.disease ,Cell biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Proto-Oncogene Proteins c-bcl-2 ,ADP-Ribosylation Factor 6 ,ADP-Ribosylation Factor 1 ,Cytokine secretion ,Immunotherapy ,Guanine nucleotide exchange factor ,Ras superfamily ,Signal Transduction - Abstract
ADP-ribosylation factor (Arf) family consisting of six family members, Arf1–Arf6, belongs to Ras superfamily and orchestrates vesicle trafficking under the control of guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins. It is well established that brefeldin A, a potent inhibitor of ArfGEFs, blocks cytokine secretion from activated T cells, suggesting that the Arf pathway plays important roles in T cell functions. In this study, because Arf1 and Arf6 are the best-characterized members among Arf family, we established T lineage–specific Arf1-deficient, Arf6-deficient, and Arf1/6 double-deficient mice to understand physiological roles of the Arf pathway in the immune system. Contrary to our expectation, Arf deficiency had little or no impact on cytokine secretion from the activated T cells. In contrast, the lack of both Arf1 and Arf6, but neither Arf1 nor Arf6 deficiency alone, rendered naive T cells susceptible to apoptosis upon TCR stimulation because of imbalanced expression of Bcl-2 family members. We further demonstrate that Arf1/6 deficiency in T cells alleviates autoimmune diseases like colitis and experimental autoimmune encephalomyelitis, whereas Ab response under Th2-polarizing conditions is seemingly normal. Our findings reveal an unexpected role for the Arf pathway in the survival of T cells during TCR-induced activation and its potential as a therapeutic target in the autoimmune diseases.
- Published
- 2021
12. The Rab11-family interacting proteins reveal selective interaction of mammalian recycling endosomes with the
- Author
-
Eric J, Hartman, Beejan, Asady, Julia D, Romano, and Isabelle, Coppens
- Subjects
Mammals ,ADP-Ribosylation Factor 6 ,rab GTP-Binding Proteins ,Vacuoles ,Animals ,Humans ,Endosomes ,Toxoplasma ,HeLa Cells ,Protein Binding - Abstract
After mammalian cell invasion, the parasite
- Published
- 2022
13. Evogliptin, a dipeptidyl peptidase-4 inhibitor, attenuates pathological retinal angiogenesis by suppressing vascular endothelial growth factor-induced Arf6 activation
- Author
-
Yeongju Yeo, Wonhee Suh, S. J. Seo, Koung Li Kim, Ryul-I Kim, and Mikyung Kim
- Subjects
Male ,Vascular Endothelial Growth Factor A ,0301 basic medicine ,Cell signaling ,Angiogenesis ,Clinical Biochemistry ,Gene Expression ,Angiogenesis Inhibitors ,QD415-436 ,Retinal Neovascularization ,030204 cardiovascular system & hematology ,Pharmacology ,Biochemistry ,Piperazines ,Article ,Pathogenesis ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Evogliptin ,Human Umbilical Vein Endothelial Cells ,medicine ,Animals ,Humans ,Phosphorylation ,Cell adhesion ,Vascular diseases ,Molecular Biology ,Dipeptidyl-Peptidase IV Inhibitors ,ADP-Ribosylation Factors ,Chemistry ,Growth factor signalling ,Adenosine ,Vascular endothelial growth factor ,Disease Models, Animal ,030104 developmental biology ,ADP-Ribosylation Factor 6 ,Medicine ,Molecular Medicine ,Signal transduction ,Signal Transduction ,medicine.drug - Abstract
Dipeptidyl peptidase-4 (DPP-4) inhibitors are used for the treatment of type 2 diabetes mellitus (DM). Recent studies have shown that beyond their effect in lowing glucose, DPP-4 inhibitors mitigate DM-related microvascular complications, such as diabetic retinopathy. However, the mechanism by which pathological retinal neovascularization, a major clinical manifestation of diabetic retinopathy, is inhibited is unclear. This study sought to examine the effects of evogliptin, a potent DPP-4 inhibitor, on pathological retinal neovascularization in mice and elucidate the mechanism by which evogliptin inhibits angiogenesis mediated by vascular endothelial growth factor (VEGF), a key factor in the vascular pathogenesis of proliferative diabetic retinopathy (PDR). In a murine model of PDR, an intravitreal injection of evogliptin significantly suppressed aberrant retinal neovascularization. In human endothelial cells, evogliptin reduced VEGF-induced angiogenesis. Western blot analysis showed that evogliptin inhibited the phosphorylation of signaling molecules associated with VEGF-induced cell adhesion and migration. Moreover, evogliptin substantially inhibited the VEGF-induced activation of adenosine 5′-diphosphate ribosylation factor 6 (Arf6), a small guanosine 5′-triphosphatase (GTPase) that regulates VEGF receptor 2 signal transduction. Direct activation of Arf6 using a chemical inhibitor of Arf-directed GTPase-activating protein completely abrogated the inhibitory effect of evogliptin on VEGF-induced activation of the angiogenic signaling pathway, which suggests that evogliptin suppresses VEGF-induced angiogenesis by blocking Arf6 activation. Our results provide insights into the molecular mechanism of the direct inhibitory effect of the DPP-4 inhibitor evogliptin on pathological retinal neovascularization. In addition to its glucose-lowering effect, the antiangiogenic effect of evogliptin could also render it beneficial for individuals with PDR., Retinal disease: How anti-diabetic drug can help eyes Pathological retinal angiogenesis, the damaging formation of new blood vessels in the retina, which is associated with various diseases including diabetes, could be reduced using the anti-diabetic drug evogliptin to inhibit the effects of a vascular growth factor. Researchers in South Korea led by Wonhee Suh and Koung Li Kim at Chung-Ang University in Seoul investigated the molecular mechanism underlying evogliptin’s effects. In studies using mice and cultured human cells they found that evogliptin inhibited the activation of signaling molecules that mediate the effects of vascular endothelial growth factor. They also identified an enzyme in the signaling pathway that is directly inhibited by evogliptin. The results offer molecular level insights into the additional benefit gained from using evogliptin to treat diabetes, distinct from the drug’s established effects in lowering blood glucose.
- Published
- 2020
14. High expression of AMAP1, an ARF6 effector, is associated with elevated levels of PD-L1 and fibrosis of pancreatic cancer
- Author
-
Ari Hashimoto, Shigeru Hashimoto, Shion Kachi, Satoshi Hirano, Hisataka Sabe, Akio Tsutaho, and Soichiro Hata
- Subjects
Adult ,Male ,0301 basic medicine ,PD-L1 ,endocrine system diseases ,lcsh:Medicine ,medicine.disease_cause ,Biochemistry ,B7-H1 Antigen ,Receptor tyrosine kinase ,Metastasis ,Focal adhesion ,03 medical and health sciences ,0302 clinical medicine ,Fibrosis ,Cell Line, Tumor ,Pancreatic cancer ,medicine ,Humans ,Gene silencing ,ARF6 ,lcsh:QH573-671 ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,Aged ,Aged, 80 and over ,biology ,FAK ,ADP-Ribosylation Factors ,lcsh:Cytology ,Research ,lcsh:R ,Cell Biology ,Middle Aged ,AMAP1 ,medicine.disease ,Pancreatic Neoplasms ,030104 developmental biology ,ADP-Ribosylation Factor 6 ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Female ,KRAS - Abstract
Background Not merely the onset of immune evasion, but other factors, such as acidosis and fibrosis, are also major barriers in cancer therapeutics. Dense fibrosis is a hallmark of pancreatic ductal carcinoma (PDAC), in which hyperactivation of focal adhesion kinase (FAK) in tumor cells was shown to be crucial. Double mutations of KRAS/ TP53 are characteristic to PDAC. We previously showed that high protein expression of ARF6 and its downstream effector AMAP1, as well as processes involved in the ARF6 activation by cell surface tyrosine kinase receptors, are major targets of the KRAS/TP53 mutations to promote PDAC invasion, metastasis, and immune evasion. This notion was recaptured by KPC mouse model of human PDAC (LSL-Kras(G12D/+); LSL-Trp53(R172H/+)); Pdx-1-Cre). Mechanistically, the ARF6-AMAP1 pathway is primarily involved in cellular dynamics of PD-L1, β1-integrins, and E-cadherin; and hence modulates cell-adhesion properties when ARF6 is activated. Here, with an aim to understand whether the ARF6-AMAP1 pathway is critically involved in the elevated levels of PD-L1 and fibrosis of PDAC, we analyzed relationship between AMAP1 and these malignant phenotypes. Moreover, because the ARF6 pathway may closely be related to focal adhesion dynamics and hence to FAK, we also investigated whether AMAP1 employs FAK in fibrosis. Methods Clinical specimens, as well as KPC cells/tumors and their shAMAP1 or shFAK derivatives were analyzed. Results Elevated levels of PD-L1 and fibrosis correlated with poor outcome of our patient cohort, to be consistent with previous reports; in which high AMAP1 expression statistically correlated with the elevated PD-L1 and fibrosis. To be consistent, silencing of AMAP1 (shAMAP1) in KPC cells resulted in reduced PD-L1 expression and fibrosis in their tumors. On the other hand, shAMAP1 only slightly affected FAK activation in KPC cells, and phosphorylated FAK did not correlate with enhanced fibrosis or with poor outcome of our patients. Conclusions Together with our previous data, our results collectively indicated that the ARF6-AMAP1 pathway, empowered by the KRAS/TP53 mutations, is closely associated with elevated PD-L1 expression and fibrosis of human PDACs, to be recaptured in the KPC mouse model. The ARF6 pathway may promote fibrosis independent of FAK.
- Published
- 2020
15. BRAG2a Mediates mGluR-Dependent AMPA Receptor Internalization at Excitatory Postsynapses through the Interaction with PSD-95 and Endophilin 3
- Author
-
Masahiro Fukaya, Makoto Itakura, Hiroyuki Sakagami, Yoshinobu Hara, Masahiko Watanabe, and Takeyuki Sugawara
- Subjects
Male ,0301 basic medicine ,Endosome ,Guinea Pigs ,Endocytic cycle ,AMPA receptor ,Receptors, Metabotropic Glutamate ,Endocytosis ,Hippocampus ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Animals ,Guanine Nucleotide Exchange Factors ,Humans ,Protein Isoforms ,Receptors, AMPA ,Cells, Cultured ,Research Articles ,Mice, Inbred ICR ,Binding Sites ,ADP-Ribosylation Factors ,Chemistry ,Long-Term Synaptic Depression ,musculoskeletal, neural, and ocular physiology ,General Neuroscience ,Post-Synaptic Density ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,nervous system ,ADP-Ribosylation Factor 6 ,Metabotropic glutamate receptor ,Synaptic plasticity ,Excitatory postsynaptic potential ,Female ,Rabbits ,Disks Large Homolog 4 Protein ,Postsynaptic density ,030217 neurology & neurosurgery ,HeLa Cells ,Protein Binding - Abstract
Brefeldin A-resistant ArfGEF 2 (BRAG2) [or Iqsec1 (IQ motif and Sec7 domain-containing protein 1)] is a guanine nucleotide exchange factor for ADP ribosylation factor 6 (Arf6), a small GTPase implicated in the membrane trafficking between the plasma membrane and endosomes. BRAG2 regulates Arf6-dependent endocytosis of AMPA receptors (AMPARs) through the direct interaction during the hippocampal long-term depression. However, the molecular mechanism by which the BRAG2–Arf6 pathway links AMPARs to the endocytic machinery remains elusive. Herein, using mouse brains of both sexes, we demonstrated that BRAG2a, an alternative isoform with a long C-terminal insert containing a proline-rich domain and type I PDZ-binding motif, was selectively localized to the excitatory postsynaptic density (PSD). Using yeast two-hybrid screening, we identified PSD-95 and endophilin 1/3 as BRAG2a-binding partners in the brain. The interaction with PSD-95 was required for synaptic targeting of BRAG2a. In cultured hippocampal neurons, stimulation of group I metabotropic glutamate receptors (mGluRs) increased the interaction of BRAG2a with endophilin 3 and concomitant Arf6 activation in a time-dependent manner. Knockdown of BRAG2 in cultured hippocampal neurons blocked the mGluR-dependent decrease in surface AMPAR levels, which was rescued by introducing wild-type BRAG2a, but not wild-type BRAG2b or BRAG2a mutants lacking the ability to activate Arf6 or to interact with endophilin 3 or PSD-95. Further postembedding immunoelectron microscopic analysis revealed the preorganized lateral distribution of BRAG2a, Arf6, and endophilin 3 for efficient endocytosis at the postsynaptic membrane. Together, the present findings unveiled a novel molecular mechanism by which BRAG2a links AMPARs to the clathrin-dependent endocytic pathway through its interaction with PSD-95 and endophilin 3.SIGNIFICANCE STATEMENTBRAG2/Iqsec1 is a GDP/GTP exchange factor for ADP ribosylation factor 6 (Arf6), a small GTPase implicated in the membrane trafficking between the plasma membrane and endosomes, and regulates Arf6-dependent endocytosis of AMPARs through direct interaction during hippocampal long-term depression, one of the mechanisms of synaptic plasticity related to learning and memory. However, the molecular mechanism by which the BRAG2–Arf6 pathway links AMPARs to the endocytic machinery remains elusive. Here, we identified isoform-specific mechanisms of BRAG2-mediated AMPAR internalization. We demonstrated that the interaction of BRAG2a isoform with PSD-95 and endophilin 3 was required for the mGluR-dependent decrease in surface AMPARs in hippocampal neurons. These results unveiled a novel molecular mechanism by which BRAG2 links AMPARs to the clathrin-mediated endocytic machinery at postsynaptic sites.
- Published
- 2020
16. MicroRNA‐145 suppresses cell migration and invasion in upper tract urothelial carcinoma by targeting ARF6
- Author
-
Lin-Li Chang, Yi-Chen Lee, Wei-Chi Hsu, A-Mei Huang, Hung-Lung Ke, Wen-Jeng Wu, Hui-Hui Lin, Ching-Chia Li, Wei-Ming Li, and Peir-In Liang
- Subjects
Male ,0301 basic medicine ,Urologic Neoplasms ,MMP2 ,Motility ,Apoptosis ,Biology ,Biochemistry ,MMP7 ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,law ,microRNA ,Biomarkers, Tumor ,Tumor Cells, Cultured ,Genetics ,medicine ,Humans ,Neoplasm Invasiveness ,Molecular Biology ,Aged ,Cell Proliferation ,Retrospective Studies ,ADP-Ribosylation Factors ,Cancer ,Cell migration ,Prognosis ,medicine.disease ,In vitro ,Gene Expression Regulation, Neoplastic ,Survival Rate ,MicroRNAs ,030104 developmental biology ,ADP-Ribosylation Factor 6 ,Case-Control Studies ,Cancer research ,Suppressor ,Female ,Urothelium ,030217 neurology & neurosurgery ,Biotechnology - Abstract
ADP-ribosylation factor 6 (ARF6) is a well-studied protein that is involved in multiple biological functions including cell migration and invasion. The mechanism by which ARF6 regulates the migration and invasion of upper tract urothelial carcinoma (UTUC) is still unknown. MiR-145-5p is a tumor suppressor microRNA, which is downregulated in several cancer types. We aimed to elucidate the molecular mechanism underlying the regulation of ARF6 by miR-145-5p in UTUC. ARF6 expression was observed to be higher in UTUC tissues than paired adjacent normal tissues. A reverse correlation between ARF6 and miR-145-5p was found in UTUC tissues. MiR-145-5p inhibited ARF6 expression by directly targeting its 3'-UTR. The functional studies indicated that ARF6 expression reversed the miR-145-5p-reduced tumor cell migration and invasion. Notably, miR-145-5p reduced MMP2, N-cadherin, FAK and MMP7, and elevated E-cadherin protein levels in vitro; however, the above effects were reversed by ARF6. Further, the expression of epithelial-to-mesenchymal transition (EMT) markers and cell invasion was suppressed by knocking down MMP7 in UTUC cells. These findings suggest that miR-145-5p may suppress UTUC cell motility and invasion by targeting ARF6/MMP7 through EMT.
- Published
- 2020
17. NMT1 and NMT2 are lysine myristoyltransferases regulating the ARF6 GTPase cycle
- Author
-
Hening Lin, Neil L. Kelleher, Shuai Zhang, J. Christopher Fromme, Kayla N. Johnson, Min Yang, Chengliang Zhu, Ian R. Price, Steve Halaby, Tatsiana Kosciuk, Garrison Paul Komaniecki, Xiaoyu Zhang, Caroline J. DeHart, and Paul M. Thomas
- Subjects
0301 basic medicine ,Cell biology ,Acylation ,Science ,Lysine ,General Physics and Astronomy ,GTPase ,SIRT2 ,Crystallography, X-Ray ,Biochemistry ,Myristic Acid ,complex mixtures ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cell Line ,03 medical and health sciences ,Sirtuin 2 ,Transferase ,Humans ,Amino Acid Sequence ,lcsh:Science ,Myristoylation ,Multidisciplinary ,030102 biochemistry & molecular biology ,Chemistry ,ADP-Ribosylation Factors ,General Chemistry ,Enzymes ,030104 developmental biology ,HEK293 Cells ,Structural biology ,ADP-Ribosylation Factor 6 ,Enzyme mechanisms ,bacteria ,lcsh:Q ,lipids (amino acids, peptides, and proteins) ,NAD+ kinase ,Fatty acylation ,Acyltransferases - Abstract
Lysine fatty acylation in mammalian cells was discovered nearly three decades ago, yet the enzymes catalyzing it remain unknown. Unexpectedly, we find that human N-terminal glycine myristoyltransferases (NMT) 1 and 2 can efficiently myristoylate specific lysine residues. They modify ADP-ribosylation factor 6 (ARF6) on lysine 3 allowing it to remain on membranes during the GTPase cycle. We demonstrate that the NAD+-dependent deacylase SIRT2 removes the myristoyl group, and our evidence suggests that NMT prefers the GTP-bound while SIRT2 prefers the GDP-bound ARF6. This allows the lysine myrisotylation-demyristoylation cycle to couple to and promote the GTPase cycle of ARF6. Our study provides an explanation for the puzzling dissimilarity of ARF6 to other ARFs and suggests the existence of other substrates regulated by this previously unknown function of NMT. Furthermore, we identified a NMT/SIRT2-ARF6 regulatory axis, which may offer new ways to treat human diseases., Lysine fatty acylation is an important protein posttranslational modification but mammalian lysine fatty acyl transferases have remained unknown so far. Here the authors report that the human N-terminal glycine myristoyltransferases 1 and 2 catalyze the addition of myristoyl chains to specific lysine residues and show that they myristoylate ARF6 lysine 3, which explains the unusual membrane binding properties of ARF6.
- Published
- 2020
18. Age-dependent autophagy induction after injury promotes axon regeneration by limiting NOTCH
- Author
-
Ellen C. Apple, Su Hyuk Ko, Zhijie Liu, and Lizhen Chen
- Subjects
0301 basic medicine ,p38 mitogen-activated protein kinases ,Notch signaling pathway ,Proto-Oncogene Mas ,03 medical and health sciences ,Autophagy ,medicine ,Animals ,Axon ,Caenorhabditis elegans ,Protein kinase A ,Molecular Biology ,Mechanistic target of rapamycin ,Receptors, Notch ,030102 biochemistry & molecular biology ,biology ,Age Factors ,Autophagosomes ,Cell Biology ,BECN1 ,MAP Kinase Kinase Kinases ,Axons ,Nerve Regeneration ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Notch proteins ,ADP-Ribosylation Factor 6 ,biology.protein ,Apoptosis Regulatory Proteins ,Lysosomes ,Signal Transduction ,Research Paper - Abstract
Macroautophagy/autophagy is essential for maintaining cellular homeostasis through the degradation of organelles and proteins. It also has a prominent role in modulating aging. However, the role of autophagy in the neuronal response to axon injury and axon regeneration, particularly in the context of aging, remains largely unknown. Our candidate genetic screen for axon regeneration regulators has identified genes in the autophagy pathway. Using a reporter that monitors autophagosomes and autolysosomes, we were able to monitor the dynamics of autophagy during axon regeneration. In response to axon injury, there was a significant increase in the number of autophagic vesicles. Injury-triggered autophagy activation and axon regeneration capacity undergo an age-dependent decline, and autophagy-activating agents partially rescued these declines. We found that DLK-1 was both required and sufficient for injury-induced autophagy activation. Autophagic vesicles co-localized with the NOTCH4 ortholog, LIN-12 receptor, a previously identified inhibitor of axon regeneration. Epistasis analyses indicate that LIN-12 might be a target of autophagy in axon regeneration. Together, our data suggest that DLK-mediated injury signaling can activate autophagy, which might limit the level of LIN-12 and NOTCH proteins to promote axon regeneration. Our findings reveal that autophagy activation can promote axon regeneration in neurons that lack maximal regrowth capacity, providing a promising therapeutic strategy for axon injury. Abbreviations: 3-MA: 3-methyladenine; ALs: autolysosomes; APs: autophagosomes; ARF-6: ADP-Ribosylation Factor related 6; ATG-9: AuTophaGy (yeast Atg homolog) 9; ATG9A: autophagy related 9A; BA1: bafilomycin A(1); BEC-1: BEClin (human autophagy) homolog; BECN1: beclin 1; C. elegans: Caenorhabditis elegans; CEBP-1: C/EBP (CCAAT/enhancer-binding protein) homolog; CNS: central nervous system; DLK-1: Dual-Leucine zipper Kinase; DMSO: dimethyl sulfoxide; DRG: dorsal root ganglion; FOS: Fos proto-oncogene, AP-1 transcription factor subunit; GABA: gamma-aminobutyric acid; GFP: green fluorescent protein; HDA-3: Histone DeAcetylase; IP3: inositol trisphosphate; ITR-1: Inositol Triphosphate Receptor; KLF-2: Kruppel-Like Factor (zinc finger protein) 2; LGG-1: LC3, GABARAP and GATE-16 family; MAK-2: MAP kinase Activated protein Kinase; MAP kinase: mitogen-activated protein kinase; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MKK-4: mitogen activated protein kinase kinase 4; MTOR: mechanistic target of rapamycin kinase; NGM: nematode growth medium; NICD: Notch intracellular domain; NOTCH: notch receptor; PLM: posterior lateral microtubule; PMK-3: P38 Map kinase family; PNS: peripheral nervous system; SCG10: superior cervical ganglion protein 10; SCI: spinal cord injury; UNC-51: UNCoordinated 51; ULK1: unc-51 like autophagy activating kinase 1; wnd: wallenda
- Published
- 2020
19. Cytohesin-2/ARNO: A Novel Bridge Between Cell Migration and Immunoregulation in Synovial Fibroblasts
- Author
-
Yilin Wang, Çağlar Çil, Margaret M. Harnett, and Miguel A. Pineda
- Subjects
musculoskeletal diseases ,STAT3 Transcription Factor ,Interleukin-1beta ,Immunology ,Gene Expression ,ARNO ,Immunomodulation ,Mice ,arthritis (including rheumatoid arthritis) ,Cell Movement ,Animals ,Immunology and Allergy ,Phosphorylation ,RNA, Small Interfering ,Original Research ,GTPase-Activating Proteins ,Synovial Membrane ,RC581-607 ,Fibroblasts ,synovial fibroblast (FLS) ,ADP-Ribosylation Factor 6 ,inflammation ,Cytokines ,cytohesin 2 ,Immunologic diseases. Allergy ,Biomarkers ,Signal Transduction - Abstract
The guanine nucleotide exchange factor cytohesin-2 (ARNO) is a major activator of the small GTPase ARF6 that has been shown to play an important role(s) in cell adhesion, migration and cytoskeleton reorganization in various cell types and models of disease. Interestingly, dysregulated cell migration, in tandem with hyper-inflammatory responses, is one of the hallmarks associated with activated synovial fibroblasts (SFs) during chronic inflammatory joint diseases, like rheumatoid arthritis. The role of ARNO in this process has previously been unexplored but we hypothesized that the pro-inflammatory milieu of inflamed joints locally induces activation of ARNO-mediated pathways in SFs, promoting an invasive cell phenotype that ultimately leads to bone and cartilage damage. Thus, we used small interference RNA to investigate the impact of ARNO on the pathological migration and inflammatory responses of murine SFs, revealing a fully functional ARNO-ARF6 pathway which can be rapidly activated by IL-1β. Such signalling promotes cell migration and formation of focal adhesions. Unexpectedly, ARNO was also shown to modulate SF-inflammatory responses, dictating their precise cytokine and chemokine expression profile. Our results uncover a novel role for ARNO in SF-dependent inflammation, that potentially links pathogenic migration with initiation of local joint inflammation, offering new approaches for targeting the fibroblast compartment in chronic arthritis and joint disease.
- Published
- 2022
20. ADP-ribosylation factor 6 expression increase in oesophageal adenocarcinoma suggests a potential biomarker role for it
- Author
-
Kate Murphy, Venkateswarlu Kanamarlapudi, and Salman Tamaddon-Jahromi
- Subjects
Gene Expression Regulation, Neoplastic ,Multidisciplinary ,Esophageal Neoplasms ,ADP-Ribosylation Factor 6 ,Tissue Array Analysis ,Case-Control Studies ,Biomarkers, Tumor ,Disease Progression ,Humans ,Adenocarcinoma ,Neoplasm Grading ,Up-Regulation - Abstract
ADP-ribosylation factor 6 small GTPase plays an important role in cell migration, invasion and angiogenesis, which are the hallmarks of cancer. Although alterations in ARF6 expression and activity have been linked to metastatic cancer in one or two tissues, the expression of ARF6 in cancers over a wide range of tissues has not been studied so far. In this report, we analysed the expression of ARF6 mRNA in cancers and corresponding healthy controls from 17 different tissues by real-time qualitative polymerase chain reaction (RT-qPCR). We further evaluated ARF6 protein expression in oesophageal adenocarcinoma (EAC) tissue microarray cores by immunohistochemistry. The ARF6 gene expression levels are highly variable between healthy and cancer tissues. Our findings suggest that the ARF6 gene expression is up-regulated highest in oesophageal cancer. In EAC TMAs, ARF6 protein expression increase correlated with EAC progression. This is the first study to investigate ARF6 gene expression in a wide array of cancer tissues and demonstrate that ARF6 expression, at both mRNA and protein levels, is significantly upregulated in higher grades of EAC, which may be useful in targeting ARF6 for cancer diagnostic and therapeutic purposes.
- Published
- 2022
21. Activation of the GTPase ARF6 regulates invasion of human vascular smooth muscle cells by stimulating MMP14 activity
- Author
-
Emilie Fiola-Masson, Julie Artigalas, Shirley Campbell, and Audrey Claing
- Subjects
Platelet-Derived Growth Factor ,Multidisciplinary ,ADP-Ribosylation Factor 6 ,Cell Movement ,Angiotensin II ,Myocytes, Smooth Muscle ,Matrix Metalloproteinase 14 ,Humans ,Atherosclerosis ,Cells, Cultured ,Muscle, Smooth, Vascular - Abstract
Hormones and growth factors stimulate vascular smooth muscle cells (VSMC) invasive capacities during the progression of atherosclerosis. The GTPase ARF6 is an important regulator of migration and proliferation of various cell types, but whether this small G protein can be activated by a variety of stimuli to promote invasion of VSMC remains unknown. Here, we aimed to define whether Platelet-derived growth factor (PDGF), a mitogenic stimulant of vascular tissues, and Angiotensin II (Ang II), a potent vasoactive peptide, can result in the activation of ARF6 in a human model of aortic SMC (HASMC). We demonstrate that these two stimuli can promote loading of GTP on this ARF isoform. Knockdown of ARF6 reduced the ability of both PDGF and Ang II to promote invasion suggesting that this GTPase regulates key molecular mechanisms mediating degradation of the extracellular matrix and migration. We report that PDGF-BB-mediated stimulation of ARF6 results in the activation of the MAPK/ERK1/2, PI3K/AKT and PAK pathways essential for invasion of HASMC. However, Ang II-mediated stimulation of ARF6 only promotes signaling through the MAPK/ERK1/2 and PAK pathways. These ARF6-mediated events lead to activation of MMP14, a membrane-bound collagenase upregulated in atherosclerosis. Moreover, ARF6 depletion decreases the release of MMP2 in the extracellular milieu. Altogether, our findings demonstrate that the GTPase ARF6 acts as a molecular switch to regulate specific signaling pathways that coordinate invasiveness of HASMC.
- Published
- 2021
22. LncRNA FOXD3-AS1 promotes breast cancer progression by mediating ARF6
- Author
-
Xingxing Zhang, Xiaojun Zhao, Lihua Chang, Fang Liu, Chunjuan Li, and Peng Ge
- Subjects
Mice, Nude ,Breast Neoplasms ,Forkhead Transcription Factors ,General Medicine ,Gene Expression Regulation, Neoplastic ,Mice ,MicroRNAs ,Oncology ,ADP-Ribosylation Factor 6 ,Cell Movement ,Cell Line, Tumor ,Animals ,Humans ,Pharmacology (medical) ,Radiology, Nuclear Medicine and imaging ,Female ,RNA, Long Noncoding ,Cell Proliferation - Abstract
Breast cancer is one of the most common malignant tumor in women. The high metastatic characteristics cause a high mortality rate of breast cancer. Increasing number of studies have indicated that long non-coding RNAs (lncRNAs) play key roles in the progression of human cancers including breast cancer. In this study, we studied the expression and molecular mechanisms of lncRNA FOXD3-AS1 in breast cancer.The expression of lncRNA FOXD3-AS1 was analyzed by TCGA database and RT-qPCR assay. CCK8 assay was used to measure cell proliferation ability. Cell migration and invasion capacities were detected by transwell assay. Potential targets of lncRNA and miRNA were predicted by bioinformatic tools. The targeting relationship between genes was verified by dual-luciferase reporter assay. The nude mice tumor model was performed to study the effect of FOXD3-AS1 on breast cancer in vivo. Protein expression was detected by western blot.In the present study, we found that the FOXD3-AS1 expression was significantly increased in breast cancer tissues compared with normal tissues and involved in the poor prognosis of patients. Functionally, knockdown of FOXD3-AS1 suppressed cell proliferation and metastasis abilities in vitro, and tumor growth in vivo. Mechanistically, FOXD3-AS1 functioned as a competing endogenous RNA (ceRNA) to upregulate ARF6 expression by targeting miR-127-3p. In addition, the roles of FOXD3-AS1 on cell proliferation and metastasis were achieved through miR-127-3p/ARF6 axis.In summary, our results reported the regulatory mechanism of FOXD3-AS1 in breast cancer progression by targeting miR-127-3p/ARF6 axis to affect cell proliferation, migration, invasion and tumor growth.
- Published
- 2021
23. IQSEC2 Deficiency Results in Abnormal Social Behaviors Relevant to Autism by Affecting Functions of Neural Circuits in the Medial Prefrontal Cortex
- Author
-
Anuradha Mehta, Emi Kouyama-Suzuki, Mengyun Zhou, Takahiro Yoshizawa, Takuma Mori, Katsuhiko Tabuchi, Yoshinori Shirai, and Toru Yanagawa
- Subjects
QH301-705.5 ,Prefrontal Cortex ,autism ,Stimulation ,Nerve Tissue Proteins ,AMPA receptor ,Biology ,Receptors, N-Methyl-D-Aspartate ,Synaptic Transmission ,Article ,social behavior ,Synapse ,Excitatory synapse ,Receptors, GABA ,synapse ,medicine ,IQSEC2 ,Animals ,Guanine Nucleotide Exchange Factors ,Protein Isoforms ,Receptors, AMPA ,Autistic Disorder ,Biology (General) ,Prefrontal cortex ,Mice, Knockout ,Pyramidal Cells ,General Medicine ,medicine.disease ,Grooming ,Up-Regulation ,Mice, Inbred C57BL ,ADP-Ribosylation Factor 6 ,Synapses ,NMDA receptor ,Autism ,Nerve Net ,Postsynaptic density ,Neuroscience ,Proto-Oncogene Proteins c-fos ,medial prefrontal cortex (mPFC) - Abstract
IQSEC2 is a guanine nucleotide exchange factor (GEF) for ADP-ribosylation factor 6 (Arf6), of which protein is exclusively localized to the postsynaptic density of the excitatory synapse. Human genome studies have revealed that the IQSEC2 gene is associated with X-linked neurodevelopmental disorders, such as intellectual disability (ID), epilepsy, and autism. In this study, we examined the behavior and synapse function in IQSEC2 knockout (KO) mice that we generated using CRIPSR/Cas9-mediated genome editing to solve the relevance between IQSEC2 deficiency and the pathophysiology of neurodevelopmental disorders. IQSEC2 KO mice exhibited autistic behaviors, such as overgrooming and social deficits. We identified that up-regulation of c-Fos expression in the medial prefrontal cortex (mPFC) induced by social stimulation was significantly attenuated in IQSEC2 KO mice. Whole cell electrophysiological recording identified that synaptic transmissions mediated by α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR), N-methyl-D-aspartate receptor (NMDAR), and γ-aminobutyric acid receptor (GABAR) were significantly decreased in pyramidal neurons in layer 5 of the mPFC in IQSEC2 KO mice. Reexpression of IQSEC2 isoform 1 in the mPFC of IQSEC2 KO mice using adeno-associated virus (AAV) rescued both synaptic and social deficits, suggesting that impaired synaptic function in the mPFC is responsible for social deficits in IQSEC2 KO mice.
- Published
- 2021
24. Arf6 anchors Cdr2 nodes at the cell cortex to control cell size at division
- Author
-
Kim H, Hannah E. Opalko, Archana Singh, Michael-Christopher Keogh, Vargas-Garcia Ca, Kristi E. Miller, and James B. Moseley
- Subjects
biology ,Chemistry ,Cell ,GTPase ,Cell Biology ,Protein Serine-Threonine Kinases ,Cell biology ,Wee1 ,medicine.anatomical_structure ,ADP-Ribosylation Factor 6 ,Node (computer science) ,Cell cortex ,Schizosaccharomyces ,medicine ,biology.protein ,Guanine nucleotide exchange factor ,Schizosaccharomyces pombe Proteins ,Protein kinase A ,Mitosis ,Cell Division ,Cell Size ,Cytokinesis - Abstract
Fission yeast cells prevent mitotic entry until a threshold cell surface area is reached. The protein kinase Cdr2 contributes to this size control system by forming multiprotein nodes that inhibit Wee1 at the medial cell cortex. Cdr2 node anchoring at the cell cortex is not fully understood. Through a genomic screen, we identified the conserved GTPase Arf6 as a component of Cdr2 signaling. Cells lacking Arf6 failed to divide at a threshold surface area and instead shifted to volume-based divisions at increased overall size. Arf6 stably localized to Cdr2 nodes in its GTP-bound but not GDP-bound state, and its GEF (guanine nucleotide exchange factor) Syt22 was required for both Arf6 node localization and proper size at division. In arf6Δ mutants, Cdr2 nodes detached from the membrane and exhibited increased dynamics. These defects were enhanced when arf6Δ was combined with other node mutants. Our work identifies a regulated anchor for Cdr2 nodes that is required for cells to sense surface area.
- Published
- 2021
25. Blocking the cytohesin-2/ARF1 axis by SecinH3 ameliorates osteoclast-induced bone loss via attenuating JNK-mediated IRE1 endoribonuclease activity
- Author
-
Yimin, Dong, Kehan, Song, Pengju, Wang, Jiachao, Guo, Honglei, Kang, Xi, Tan, Binxiang, Zhu, Renpeng, Peng, Meipeng, Zhu, Kaixu, Yu, Qian, Guo, Hanfeng, Guan, and Feng, Li
- Subjects
Pharmacology ,ADP-Ribosylation Factors ,ADP-Ribosylation Factor 6 ,Endoribonucleases ,Humans ,Osteoclasts ,Osteoporosis ,Protein Serine-Threonine Kinases - Abstract
cytohesin-2 is a guanine nucleotide exchange factor to activate ARF1 and ARF6, which are involved in various biological processes, including signal transduction, cell differentiation, cell structure organization, and survival. Nevertheless, there is a lack of evidence revealing the role of cytohesin-2 in osteoclast differentiation and in the development of osteoporosis. In this study, we find cytohesin-2 and ARF1 positively regulate osteoclast differentiation and function. Blocking the cytohesin-2 /ARF1 axis with SecinH3 or by genetic silencing of cytohesin-2 inhibits osteoclast formation and function in vitro. In vivo treatment with SecinH3 ameliorates ovariectomy-induced osteoporosis. Mechanistically, RNA-sequencing combined with molecular biological methodologies reveal that the regulatory function of cythohesin-2/ARF1 axis in osteoclast differentiation is mainly dependent on activating the JNK pathway. Further, in addition to the common viewpoint that JNK is activated by IRE1 via its kinase activity, we found that JNK can act upstream and regulate the endoribonuclease activity of IRE1 to promote XBP1 splicing. Both SecinH3 and silencing of cytohesin-2 inhibit JNK activation and IRE1 endoribonuclease activity, leading to the suppression of osteoclast differentiation. Taken together, our findings add new insights into the regulation between JNK and IRE1, and reveal that inhibiting the cytohesin-2/ARF1/JNK/IRE1 axis might represent a potential new strategy for the treatment of post-menopause osteoporosis.
- Published
- 2022
26. miR‑28‑3p inhibits prostate cancer cell proliferation, migration and invasion, and promotes apoptosis by targeting ARF6
- Author
-
Huizhang Li, Shihua Ye, Jiabin Zhang, and Yi Yao
- Subjects
Erk/Rac signaling pathway ,Cancer Research ,Gene knockdown ,microRNA-28-3p ,Oncogene ,Cell growth ,Chemistry ,Cell ,ADP-ribosylation factor 6 ,Articles ,General Medicine ,Cell cycle ,medicine.anatomical_structure ,Immunology and Microbiology (miscellaneous) ,DU145 ,prostate cancer cells ,microRNA ,LNCaP ,medicine ,Cancer research - Abstract
Previous studies have reported that the expression levels of microRNA (miR)-28-3p are downregulated in prostate cancer (PCa) compared with those in adjacent normal tissues. However, to the best of our knowledge, the function and underlying mechanisms of miR-28-3p in PCa have not been reported. The present study aimed to explore the role of miR-28-3p and its mechanism in the development of PCa. In the present study, miR-28-3p and ADP-ribosylation factor 6 (ARF6) expression levels were analyzed using reverse transcription-quantitative PCR (RT-qPCR). Cell proliferation, colony formation, apoptosis, migration and invasion were determined using Cell Counting Kit-8, colony forming, flow cytometry and Transwell assays, respectively. The association between miR-28-3p and ARF6 was investigated using a dual luciferase reporter assay. ARF6, Rac1, Erk1/2 and phosphorylated (p)-Erk1/2 protein expression levels were analyzed using western blotting. The results of the present study revealed that miR-28-3p expression levels were downregulated, whereas ARF6 expression levels were upregulated in PCa cell lines (LNCaP, 22Rv-1, PC-3 and DU145) compared with those in the normal prostate line RWPE-1. The overexpression of miR-28-3p promoted cell apoptosis, and inhibited cell proliferation, colony formation, migration and invasion. However, the knockdown of miR-28-3p exerted the opposite results. The results of the dual luciferase reporter assays, RT-qPCR and western blotting indicated that ARF6 was a target gene of miR-28-3p. Finally, rescue experiments demonstrated that ARF6 overexpression attenuated the effects of the miR-28-3p mimic by upregulating Rac1 and p-Erk1/2 expression in PCa cells. In conclusion, these findings indicated that miR-28-3p may inhibit the biological behaviors of PCa cells by targeting ARF6, and therefore may represent a novel therapeutic candidate for PCa.
- Published
- 2021
27. A Brucella effector modulates the Arf6‐Rab8a GTPase cascade to promote intravacuolar replication
- Author
-
Kelsey A. Binder, Elizabeth Borghesan, Erin P. Smith, Jean Celli, Sebenzile K. Myeni, and Leigh A. Knodler
- Subjects
Brucella abortus ,Endosomes ,GTPase ,Vacuole ,Biology ,Models, Biological ,Brucellosis ,Article ,General Biochemistry, Genetics and Molecular Biology ,Type IV Secretion Systems ,Mice ,Bacterial Proteins ,Animals ,Humans ,Secretion ,Molecular Biology ,General Immunology and Microbiology ,Effector ,pathogenesis ,General Neuroscience ,Endoplasmic reticulum ,GTPase-Activating Proteins ,Articles ,retrograde membrane transport ,Membrane transport ,Membranes & Trafficking ,Brucella ,type IV secretion ,Microbiology, Virology & Host Pathogen Interaction ,Cell biology ,Vesicular transport protein ,Protein Transport ,ADP-Ribosylation Factor 6 ,rab GTP-Binding Proteins ,Host-Pathogen Interactions ,Vacuoles ,ACAP1 ,Intracellular ,HeLa Cells ,Protein Binding ,trans-Golgi Network - Abstract
Remodeling of host cellular membrane transport pathways is a common pathogenic trait of many intracellular microbes that is essential to their intravacuolar life cycle and proliferation. The bacterium Brucella abortus generates a host endoplasmic reticulum‐derived vacuole (rBCV) that supports its intracellular growth, via VirB Type IV secretion system‐mediated delivery of effector proteins, whose functions and mode of action are mostly unknown. Here, we show that the effector BspF specifically promotes Brucella replication within rBCVs by interfering with vesicular transport between the trans‐Golgi network (TGN) and recycling endocytic compartment. BspF targeted the recycling endosome, inhibited retrograde traffic to the TGN, and interacted with the Arf6 GTPase‐activating Protein (GAP) ACAP1 to dysregulate Arf6‐/Rab8a‐dependent transport within the recycling endosome, which resulted in accretion of TGN‐associated vesicles by rBCVs and enhanced bacterial growth. Altogether, these findings provide mechanistic insight into bacterial modulation of membrane transport used to promote their own proliferation within intracellular vacuoles., Brucella abortus effector protein BspF binding to host GTPase‐activating protein ACAP1 facilitates intracellular bacterial growth by decreasing retrograde vesicular transport to the trans‐Golgi network.
- Published
- 2021
28. Arf6 exacerbates allergic asthma through cell-to-cell transmission of ASC inflammasomes
- Author
-
Kyosuke Nagata, Takahiro Kuroki, Tsunaki Hongu, Yu-Hsien Lin, Akari Ishitsuka, Atsushi Kawaguchi, Yuji Funakoshi, SangJoon Lee, Akira Shibuya, and Yasunori Kanaho
- Subjects
Inflammasomes ,Ovalbumin ,THP-1 Cells ,Interleukin-1beta ,Inflammation ,Cell Communication ,Allergic inflammation ,Proinflammatory cytokine ,Mice ,Paracrine signalling ,Th2 Cells ,Phagocytosis ,Macrophages, Alveolar ,Extracellular ,Animals ,Humans ,Medicine ,Autocrine signalling ,Lung ,Asthma ,Mice, Knockout ,business.industry ,General Medicine ,Triazoles ,Eosinophil ,Symptom Flare Up ,medicine.disease ,respiratory tract diseases ,CARD Signaling Adaptor Proteins ,Disease Models, Animal ,medicine.anatomical_structure ,ADP-Ribosylation Factor 6 ,Immunology ,medicine.symptom ,business ,Research Article - Abstract
Asthma is a chronic inflammatory disease of the airways associated with excess production of Th2 cytokines and lung eosinophil accumulation. This inflammatory response persists in spite of steroid administration that blocks autocrine/paracrine loops of inflammatory cytokines, and the detailed mechanisms underlying asthma exacerbation remain unclear. Here, we show that asthma exacerbation is triggered by airway macrophages through a prion-like cell-to-cell transmission of extracellular particulates, including ASC protein, that assemble inflammasomes and mediate IL-1β production. OVA-induced allergic asthma and associated IL-1β production were alleviated in mice with small GTPase Arf6-deficient macrophages. The extracellular ASC specks were slightly engulfed by Arf6–/– macrophages, and the IL-1β production was reduced in Arf6–/– macrophages compared with that in WT macrophages. Furthermore, pharmacological inhibition of the Arf6 guanine nucleotide exchange factor suppressed asthma-like allergic inflammation in OVA-challenged WT mice. Collectively, the Arf6-dependent intercellular transmission of extracellular ASC specks contributes to the amplification of allergic inflammation and subsequent asthma exacerbation.
- Published
- 2021
29. A polymer‑calcium phosphate nanocapsule for RNAi-induced oxidative stress and cascaded chemotherapy
- Author
-
Huiling Huang, Yuanyuan Yuan, Chujie Zheng, Hong Xiao, Jun Pang, Jinsheng Huang, Yiyao Wang, Minzhao Lin, Yong Wang, and Xintao Shuai
- Subjects
Calcium Phosphates ,Chemistry ,Polymers ,Pharmaceutical Science ,Oxidative phosphorylation ,Mitochondrion ,medicine.disease_cause ,Micelle ,Cell biology ,Mice ,Oxidative Stress ,Nanocapsules ,RNA interference ,ADP-Ribosylation Factor 6 ,Cell Line, Tumor ,Cancer cell ,medicine ,Animals ,Nanoparticles ,Prodrugs ,RNA Interference ,Linker ,Camptothecin ,Oxidative stress ,medicine.drug - Abstract
As most of intracellular reactive oxygen species (ROS) is produced in the mitochondria, mitochondrial modulation of cancer cell is a promising strategy for maximizing the in situ-activable combination therapy of oxidative catastrophe and cascaded chemotherapy. Herein, a serum-stable polymer‑calcium phosphate (CaP) hybrid nanocapsule carrying siRNA against ADP-ribosylation factor 6 (Arf6) overexpressed in cancer cells and parent drug camptothecin (CPT), designated as PTkCPT/siRNA, was developed for the RNAi-induced oxidative catastrophe and cascaded chemotherapy. A copolymer of mPEG-P(Asp-co-TkCPT), covalently tethered with chemotherapeutic CPT via a ROS-labile dithioketal (Tk) linker, was synthesized and self-assembled into a PTkCPT micelle as a nanotemplate for the CaP mineralization. The as-prepared PTkCPT/siRNA nanoparticle showed a core-shell-distinct nanocapsule which was consisted of a spherical polymeric core enclosed within a CaP shell capable of releasing siRNA in response to lysosomal acidity. Blocking Arf6 signal pathway of cancer cells led to their mitochondrial aggregation and subsequently induced a burst of ROS for oxidative catastrophe, which further triggered the cascaded CPT chemotherapy via the breakage of ROS-labile dithioketal linker. This strategy of RNAi-induced oxidative catastrophe and cascaded chemotherapy resulted in a significant combination effect on cancer cell killing and tumor growth inhibition in mice with low side effects, and provided a promising paradigm for precise cancer therapy.
- Published
- 2021
30. Discovery of a dual Ras and ARF6 inhibitor from a GPCR endocytosis screen
- Author
-
Jean Duchaine, Stéphane A. Laporte, Etienne Khoury, Alexandre Beautrait, Yoon Namkung, Michel Bouvier, Mónica Lara-Márquez, Tomasz Maciej Stepniewski, Doris A. Schuetz, Audrey Claing, Olivier Radresa, Sylvain Armando, Anne Marinier, Nathalie Lamarche-Vane, Jana Selent, Jenna Giubilaro, and Shirley Campbell
- Subjects
MAPK/ERK pathway ,Bioluminescence Resonance Energy Transfer Techniques ,Science ,media_common.quotation_subject ,General Physics and Astronomy ,Small G Protein ,Hormone receptors ,Molecular Dynamics Simulation ,Endocytosis ,General Biochemistry, Genetics and Molecular Biology ,Article ,Receptors, G-Protein-Coupled ,Cell Line, Tumor ,Drug Discovery ,Humans ,Enzyme Inhibitors ,Receptor ,Internalization ,Protein kinase B ,G protein-coupled receptor ,media_common ,Cell Proliferation ,Multidisciplinary ,Binding Sites ,Chemistry ,ADP-Ribosylation Factors ,Receptor Protein-Tyrosine Kinases ,General Chemistry ,Angiotensin II ,Cell biology ,HEK293 Cells ,ADP-Ribosylation Factor 6 ,Screening ,ras Proteins ,Signal Transduction - Abstract
Internalization and intracellular trafficking of G protein-coupled receptors (GPCRs) play pivotal roles in cell responsiveness. Dysregulation in receptor trafficking can lead to aberrant signaling and cell behavior. Here, using an endosomal BRET-based assay in a high-throughput screen with the prototypical GPCR angiotensin II type 1 receptor (AT1R), we sought to identify receptor trafficking inhibitors from a library of ~115,000 small molecules. We identified a novel dual Ras and ARF6 inhibitor, which we named Rasarfin, that blocks agonist-mediated internalization of AT1R and other GPCRs. Rasarfin also potently inhibits agonist-induced ERK1/2 signaling by GPCRs, and MAPK and Akt signaling by EGFR, as well as prevents cancer cell proliferation. In silico modeling and in vitro studies reveal a unique binding modality of Rasarfin within the SOS-binding domain of Ras. Our findings unveil a class of dual small G protein inhibitors for receptor trafficking and signaling, useful for the inhibition of oncogenic cellular responses., While Ras is a promising target for cancer therapy, development of inhibitors targeting Ras signaling has proven challenging. Here, the authors report the discovery of Rasarfin, a small molecule from a phenotypic screen on G protein-coupled receptor (GPCR) endocytosis that acts as a dual Ras and ARF6 inhibitor.
- Published
- 2021
31. Bone Marrow-Derived Cells Restore Functional Integrity of the Gut Epithelial and Vascular Barriers in a Model of Diabetes and ACE2 Deficiency
- Author
-
Baskaran Athmanathan, Prabhakara R Nagareddy, Jason L. Floyd, Yaqian Duan, Justin P. Wright, Dongni Feng, Mariana Dupont, Gavin Y. Oudit, Regina Lamendella, Eleni Beli, Maria B. Grant, Sunil K. Noothi, Gopalkrishna Sreejit, Amanda R. Jensen, Alexander G. Obukhov, Ram Prasad, Ana Leda F. Longhini, Troy A. Markel, and Sergio Li Calzi
- Subjects
Male ,0301 basic medicine ,Physiology ,Neovascularization, Physiologic ,Peptidoglycan ,Peptidyl-Dipeptidase A ,Article ,Capillary Permeability ,03 medical and health sciences ,Incomplete knowledge ,0302 clinical medicine ,Diabetes mellitus ,Intestine, Small ,Animals ,Humans ,Medicine ,Intestinal Mucosa ,Cells, Cultured ,Bone Marrow Transplantation ,Mice, Knockout ,Bacteria ,Gut barrier ,business.industry ,Adherens Junctions ,Recovery of Function ,medicine.disease ,Gut microbiome ,Gastrointestinal Microbiome ,Mice, Inbred C57BL ,Disease Models, Animal ,Functional integrity ,030104 developmental biology ,medicine.anatomical_structure ,Diabetes Mellitus, Type 2 ,ADP-Ribosylation Factor 6 ,Immunology ,Dysbiosis ,Angiotensin-Converting Enzyme 2 ,Bone marrow ,Inflammation Mediators ,Cardiology and Cardiovascular Medicine ,business ,hormones, hormone substitutes, and hormone antagonists ,030217 neurology & neurosurgery - Abstract
Rationale: There is incomplete knowledge of the impact of bone marrow cells on the gut microbiome and gut barrier function. Objective: We postulated that diabetes mellitus and systemic ACE2 (angiotensin-converting enzyme 2) deficiency would synergize to adversely impact both the microbiome and gut barrier function. Methods and Results: Bacterial 16S rRNA sequencing and metatranscriptomic analysis were performed on fecal samples from wild-type, ACE2 −/y , Akita (type 1 diabetes mellitus), and ACE2 −/y -Akita mice. Gut barrier integrity was assessed by immunofluorescence, and bone marrow cell extravasation into the small intestine was evaluated by flow cytometry. In the ACE2 −/y -Akita or Akita mice, the disrupted barrier was associated with reduced levels of myeloid angiogenic cells, but no increase in inflammatory monocytes was observed within the gut parenchyma. Genomic and metatranscriptomic analysis of the microbiome of ACE2 −/y -Akita mice demonstrated a marked increase in peptidoglycan-producing bacteria. When compared with control cohorts treated with saline, intraperitoneal administration of myeloid angiogenic cells significantly decreased the microbiome gene expression associated with peptidoglycan biosynthesis and restored epithelial and endothelial gut barrier integrity. Also indicative of diabetic gut barrier dysfunction, increased levels of peptidoglycan and FABP-2 (intestinal fatty acid-binding protein 2) were observed in plasma of human subjects with type 1 diabetes mellitus (n=21) and type 2 diabetes mellitus (n=23) compared with nondiabetic controls (n=23). Using human retinal endothelial cells, we determined that peptidoglycan activates a noncanonical TLR-2 (Toll-like receptor 2) associated MyD88 (myeloid differentiation primary response protein 88)-ARNO (ADP-ribosylation factor nucleotide-binding site opener)-ARF6 (ADP-ribosylation factor 6) signaling cascade, resulting in destabilization of p120-catenin and internalization of VE-cadherin as a mechanism of deleterious impact of peptidoglycan on the endothelium. Conclusions: We demonstrate for the first time that the defect in gut barrier function and dysbiosis in ACE2 −/y -Akita mice can be favorably impacted by exogenous administration of myeloid angiogenic cells.
- Published
- 2019
32. Arf6-driven endocytic recycling of CD147 determines HCC malignant phenotypes
- Author
-
Linjia Su, Zhe Ma, Chuan-Shan Zhang, Gui-qiu Liu, Jing Li, Sihe Zhang, Guhe Jia, Yongjun Piao, Shanshan Qi, and Qing Zhang
- Subjects
0301 basic medicine ,Cancer Research ,Carcinoma, Hepatocellular ,Endocytic recycling ,RAC1 ,lcsh:RC254-282 ,Flow cytometry ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Cell Adhesion ,Humans ,Arf6 ,Cell adhesion ,Cell Aggregation ,rab5 GTP-Binding Proteins ,medicine.diagnostic_test ,Chemistry ,Cell adhesion molecule ,ADP-Ribosylation Factors ,Research ,Liver Neoplasms ,Malignant phenotype ,Cancer ,Hep G2 Cells ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immunohistochemistry ,Cell aggregation ,Endocytosis ,Up-Regulation ,030104 developmental biology ,Phenotype ,Oncology ,ADP-Ribosylation Factor 6 ,rab GTP-Binding Proteins ,030220 oncology & carcinogenesis ,Cancer research ,Basigin ,CD147 ,Liver cancer - Abstract
BackgroundAdhesion molecules distributed on the cell-surface depends upon their dynamic trafficking that plays an important role during cancer progression. ADP-ribosylation factor 6 (Arf6) is a master regulator of membrane trafficking. CD147, a tumor-related adhesive protein, can promote the invasion of liver cancer. However, the role of Arf6 in CD147 trafficking and its contribution to liver cancer progression remain unclear.MethodsStable liver cancer cell lines with Arf6 silencing and over-expression were established. Confocal imaging, flow cytometry, biotinylation and endomembrane isolation were used to detect CD147 uptake and recycling. GST-pull down, gelatin zymography, immunofluorescence, cell adhesion, aggregation and tight junction formation, Transwell migration, and invasion assays were used to examine the cellular phenotypes. GEPIA bioinformatics, patient’s specimens and electronic records collection, and immunohistochemistry were performed to obtain the clinical relevance for Arf6-CD147 signaling.ResultsWe found that the endocytic recycling of CD147 in liver cancer cells was controlled by Arf6 through concurrent Rab5 and Rab22 activation. Disruption of Arf6-mediated CD147 trafficking reduced the cell-matrix and cell-cell adhesion, weakened cell aggregation and junction stability, attenuated MMPs secretion and cytoskeleton reorganization, impaired HGF-stimulated Rac1 activation, and markedly decreased the migration and invasion of liver cancer cells. Moreover, high-expression of the Arf6-CD147 signaling components in HCC (hepatocellular carcinoma) was closely correlated with poor clinical outcome of patients.ConclusionsOur results revealed that Arf6-mediated CD147 endocytic recycling is required for the malignant phenotypes of liver cancer. The Arf6-driven signaling machinery provides excellent biomarkers or therapeutic targets for the prevention of liver cancer.
- Published
- 2019
33. ApoE4 Alters ABCA1 Membrane Trafficking in Astrocytes
- Author
-
Shaowei Wang, Ori Liraz, Jan O. Johansson, Helena C. Chui, Trusha Parekh, Chongren Tang, Roni Bar, Daniel M. Michaelson, Hussein N. Yassine, Jian Sima, Jamie Chan, Usha Gundimeda, Varun Rawat, and Michael G. Harrington
- Subjects
Male ,0301 basic medicine ,Apolipoprotein E ,Apolipoprotein E4 ,Mice, Transgenic ,ATP-binding cassette transporter ,Lipid-anchored protein ,Protein aggregation ,Cell membrane ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,In vivo ,Cricetinae ,polycyclic compounds ,medicine ,Animals ,Humans ,Cells, Cultured ,Research Articles ,Aged ,Cell Line, Transformed ,Aged, 80 and over ,biology ,Chemistry ,Cholesterol ,General Neuroscience ,Cell Membrane ,Middle Aged ,Cell biology ,Mice, Inbred C57BL ,Protein Transport ,030104 developmental biology ,medicine.anatomical_structure ,ADP-Ribosylation Factor 6 ,Astrocytes ,ABCA1 ,biology.protein ,Female ,lipids (amino acids, peptides, and proteins) ,030217 neurology & neurosurgery ,ATP Binding Cassette Transporter 1 ,HeLa Cells ,Protein Binding - Abstract
TheAPOEε4 allele is the strongest genetic risk factor for late-onset Alzheimer's disease (AD). ApoE protein aggregation plays a central role in AD pathology, including the accumulation of β-amyloid (Aβ). Lipid-poor ApoE4 protein is prone to aggregate and lipidating ApoE4 protects it from aggregation. The mechanisms regulating ApoE4 aggregationin vivoare surprisingly not known. ApoE lipidation is controlled by the activity of the ATP binding cassette A1 (ABCA1). ABCA1 recycling and degradation is regulated by ADP-ribosylation factor 6 (ARF6). We found that ApoE4 promoted greater expression of ARF6 compared with ApoE3, trapping ABCA1 in late-endosomes and impairing its recycling to the cell membrane. This was associated with lower ABCA1-mediated cholesterol efflux activity, a greater percentage of lipid-free ApoE particles, and lower Aβ degradation capacity. Human CSF fromAPOEε4/ε4 carriers showed a lower ability to induce ABCA1-mediated cholesterol efflux activity and greater percentage of aggregated ApoE protein compared with CSF fromAPOEε3/ε3 carriers. Enhancing ABCA1 activity rescued impaired Aβ degradation in ApoE4-treated cells and reduced both ApoE and ABCA1 aggregation in the hippocampus of male ApoE4-targeted replacement mice. Together, our data demonstrate that aggregated and lipid-poor ApoE4 increases ABCA1 aggregation and decreases ABCA1 cell membrane recycling. Enhancing ABCA1 activity to reduce ApoE and ABCA1 aggregation is a potential therapeutic strategy for the prevention of ApoE4 aggregation-driven pathology.SIGNIFICANCE STATEMENTApoE protein plays a key role in the formation of amyloid plaques, a hallmark of Alzheimer's disease (AD). ApoE4 is more aggregated and hypolipidated compared with ApoE3, but whether enhancing ApoE lipidationin vivocan reverse ApoE aggregation is not known. ApoE lipidation is controlled by the activity of the ATP binding cassette A1 (ABCA1). In this study, we demonstrated that the greater propensity of lipid-poor ApoE4 to aggregate decreased ABCA1 membrane recycling and its ability to lipidate ApoE. Importantly, enhancing ABCA1 activity to lipidate ApoE reduced ApoE and ABCA1 aggregation. This work provides critical insights into the interactions among ABCA1, ApoE lipidation and aggregation, and underscores the promise of stabilizing ABCA1 activity to prevent ApoE-driven aggregation pathology.
- Published
- 2019
34. Arf6 regulates RhoB subcellular localization to control cancer cell invasion
- Author
-
Kossay Zaoui, Morag Park, Stephanie Duhamel, and Charles V. Rajadurai
- Subjects
Endosome ,RHOB ,Uterine Cervical Neoplasms ,Endocytic recycling ,Breast Neoplasms ,Endosomes ,Biology ,Article ,Focal adhesion ,03 medical and health sciences ,0302 clinical medicine ,Tumor Cells, Cultured ,Humans ,Small GTPase ,rhoB GTP-Binding Protein ,Cytoskeleton ,Research Articles ,Cell Proliferation ,030304 developmental biology ,0303 health sciences ,ADP-Ribosylation Factors ,Cell migration ,Cell Biology ,Subcellular localization ,Cell biology ,ADP-Ribosylation Factor 6 ,030220 oncology & carcinogenesis ,Female ,HeLa Cells - Abstract
Zaoui et al. provide the first mechanistic evidence supporting a regulatory mechanism for RhoB localization and stability by Arf6. These findings establish that Arf6 is essential for RhoB-specific subcellular targeting to endosomes and, thus, the regulation of membrane trafficking and cell motility following activation of the oncogenic receptor tyrosine kinase, Met., The ADP-ribosylation factor 6 (Arf6) is a small GTPase that regulates endocytic recycling processes in concert with various effectors. Arf6 controls cytoskeletal organization and membrane trafficking; however, the detailed mechanisms of regulation remain poorly understood. Here, we report that Arf6 forms a complex with RhoB. The interaction between RhoB and Arf6 is mediated by the GCI (glycine, cysteine, and isoleucine) residues (188–190) of RhoB. Specific targeting of Arf6 to plasma membrane or mitochondrial membranes promotes recruitment and colocalization of RhoB to these membrane microdomains. Arf6 depletion promotes the loss of RhoB from endosomal membranes and leads to RhoB degradation through an endolysosomal pathway. This results in defective actin and focal adhesion dynamics and increased 3D cell migration upon activation of the Met receptor tyrosine kinase. Our findings identify a novel regulatory mechanism for RhoB localization and stability by Arf6 and establish the strict requirement of Arf6 for RhoB-specific subcellular targeting to endosomes and biological functions.
- Published
- 2019
35. A NAV2729-sensitive mechanism promotes adrenergic smooth muscle contraction and growth of stromal cells in the human prostate
- Author
-
Annika Herlemann, Christian Gratzke, Christian G. Stief, Beata Rutz, Raphaela Waidelich, Paul Kuppermann, Martin Hennenberg, Yiming Wang, Alexander Tamalunas, Anna Ciotkowska, Bingsheng Li, Xiaolong Wang, Qingfeng Yu, Frank Strittmatter, and Ruixiao Wang
- Subjects
Male ,0301 basic medicine ,Stromal cell ,RHOA ,Prostatic Hyperplasia ,RAC1 ,Pyrimidinones ,Chlorobenzenes ,Biochemistry ,Norepinephrine ,03 medical and health sciences ,Thromboxane A2 ,chemistry.chemical_compound ,Prostate ,medicine ,Humans ,RNA, Small Interfering ,Molecular Biology ,Cell Proliferation ,030102 biochemistry & molecular biology ,biology ,ADP-Ribosylation Factors ,Chemistry ,Muscle, Smooth ,Molecular Bases of Disease ,Cell Biology ,Smooth muscle contraction ,Prostate-Specific Antigen ,Hyperplasia ,Nitro Compounds ,medicine.disease ,Actin Cytoskeleton ,030104 developmental biology ,medicine.anatomical_structure ,ADP-Ribosylation Factor 6 ,15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid ,Cancer research ,biology.protein ,Pyrazoles ,RNA Interference ,Benign prostatic hyperplasia (BPH) ,Stromal Cells ,Muscle Contraction - Abstract
Voiding symptoms in benign prostatic hyperplasia (BPH) are driven by prostate smooth muscle contraction and prostate growth. Smooth muscle contraction in the prostate and other organs critically depends on activation of the small monomeric GTPase RhoA and probably Rac1. A role of another GTPase, ADP-ribosylation factor 6 (ARF6), for smooth muscle contraction has been recently suggested by indirect evidence but remains to be proven for any organ. Here, we report effects of NAV2729, an inhibitor with assumed specificity for ARF6, in human prostate tissues and cultured prostate stromal cells (WPMY-1). NAV2729 (5 μm) inhibited neurogenic and α(1)-adrenergic contractions of human prostate tissues. Contractions induced by endothelin-1, by the thromboxane A(2) agonist U46619, or by high molar KCl were not inhibited. Correlation analyses suggested up-regulation of prostatic ARF6 expression with increasing degree of BPH, as ARF6 expression increased with the content of prostate-specific antigen (PSA) of prostate tissues. NAV2729 inhibited ARF6 activity but not other GTPases (ARF1, RhoA, Rac1) in prostate tissues and in WPMY-1 cells. Proliferation of WPMY-1 cells was inhibited concentration-dependently by NAV2726, as reflected by decreased viability, 5-ethynyl-2′-deoxyuridine (EdU) assay, colony formation assay, and expression of Ki-67. Silencing of ARF6 expression mimicked effects of NAV2729 on viability and in the EdU assay. Effects of NAV2729 on viability and proliferation were attenuated in cells with silenced ARF6 expression. Our findings suggest that a NAV2729-sensitive mechanism promotes adrenergic contraction and stromal cell growth in the human prostate, which is probably ARF6-mediated. Similar actions in other organs and urodynamic effects of NAV2729 appear possible.
- Published
- 2019
36. ACAP4 interacts with CrkII to promote the recycling of integrin β1
- Author
-
Shuai Kong, Xueyan Song, Jin Xiao, Xinwang Cao, Wenjuan Xu, Lu Ding, Fengsong Wang, and Guangsheng Xu
- Subjects
0301 basic medicine ,Integrin β1 ,Biophysics ,medicine.disease_cause ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,Cell polarity ,Cell Adhesion ,medicine ,Humans ,Protein Interaction Domains and Motifs ,Protein Interaction Maps ,Cell adhesion ,Molecular Biology ,Cell growth ,Chemistry ,Integrin beta1 ,Vesicle ,Cell Membrane ,GTPase-Activating Proteins ,Signal transducing adaptor protein ,Bacterial pathogenesis ,Cell Biology ,Proto-Oncogene Proteins c-crk ,Cell biology ,Protein Transport ,HEK293 Cells ,030104 developmental biology ,ADP-Ribosylation Factor 6 ,030220 oncology & carcinogenesis ,Carcinogenesis ,HeLa Cells - Abstract
ACAP4, a GTPase-activating protein (GAP) for the ADP-ribosylation factor 6 (ARF6), plays import roles in cell migration, cell polarity, vesicle trafficking and tumorigenesis. Similarly, the ubiquitously expressed adaptor protein CrkII functions in a wide range of cellular activities, including cell proliferation, T cell adhesion and activation, tumorigenesis, and bacterial pathogenesis. Here, we demonstrate that ACAP4 physically interacts with CrkII. Biochemical experiments revealed that ACAP4550−660 and the SH3N domain of CrkII are responsible for the interaction. Functional characterization showed that the interaction is required for the recruitment of ACAP4 to the plasma membrane where ACAP4 functions to regulate the recycling of the signal transducer integrin β1. Thus, we suggest that the CrkII-ACAP4 complex may be involved in regulation of cell adhesion.
- Published
- 2019
37. An ARF6–Exportin-5 axis delivers pre-miRNA cargo to tumour microvesicles
- Author
-
Ye Zhang, Colin Sheehan, Crislyn D'Souza-Schorey, and James W. Clancy
- Subjects
Cell ,Active Transport, Cell Nucleus ,Karyopherins ,Article ,03 medical and health sciences ,0302 clinical medicine ,Cell-Derived Microparticles ,Neoplasms ,microRNA ,Tumor Microenvironment ,medicine ,Humans ,Phosphorylation ,Nuclear export signal ,030304 developmental biology ,0303 health sciences ,biology ,ADP-Ribosylation Factors ,Chemistry ,GTPase-Activating Proteins ,Cell Biology ,Microvesicles ,Cell biology ,MicroRNAs ,medicine.anatomical_structure ,ADP-Ribosylation Factor 6 ,Cytoplasm ,030220 oncology & carcinogenesis ,biology.protein ,RNA Interference ,Casein kinase 1 ,Dicer - Abstract
Tumour-derived microvesicles (TMVs) comprise a class of extracellular vesicles released from tumour cells that are now understood to facilitate communication between the tumour and the surrounding microenvironment. Despite their significance, the regulatory mechanisms governing the trafficking of bioactive cargos to TMVs at the cell surface remain poorly defined. Here we describe a molecular pathway for the delivery of microRNA (miRNA) cargo to nascent TMVs involving the dissociation of a pre-miRNA/Exportin-5 complex from Ran-GTP following nuclear export and its subsequent transfer to a cytoplasmic shuttle comprised of ARF6-GTP and GRP1. As such, ARF6 activation increases the pre-miRNA cargo contained within TMVs through a process that requires the casein kinase 2-mediated phosphorylation of RanGAP1. Furthermore, TMVs were found to contain pre-miRNA processing machinery including Dicer and Argonaute-2, which allow for cell-free pre-miRNA processing within shed vesicles. These findings offer cellular targets to block the loading and processing of pre-miRNAs within TMVs.
- Published
- 2019
38. Small GTPase Arf6 regulates diabetes‐induced cholesterol accumulation in podocytes
- Author
-
Zhaowei Chen, Jun Feng, Guohua Ding, Jijia Hu, Qian Yang, and Wei Liang
- Subjects
Blood Glucose ,Male ,0301 basic medicine ,Physiology ,Clinical Biochemistry ,Cell Line ,Diabetes Mellitus, Experimental ,Podocyte ,Rats, Sprague-Dawley ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,In vivo ,Diabetes mellitus ,medicine ,Animals ,Humans ,Diabetic Nephropathies ,Small GTPase ,Gene knockdown ,biology ,ADP-Ribosylation Factors ,Podocytes ,Cholesterol ,Biological Transport ,Transporter ,Cell Biology ,medicine.disease ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,ADP-Ribosylation Factor 6 ,030220 oncology & carcinogenesis ,ABCA1 ,biology.protein ,lipids (amino acids, peptides, and proteins) ,ATP Binding Cassette Transporter 1 - Abstract
Podocyte injury is a critical factor for the initiation and progression of diabetic kidney disease (DKD). However, the underlying mechanisms of podocyte injury in DKD have not been completely elucidated. Studies suggested that intracellular cholesterol accumulation was correlated with podocyte injury, but the cause of podocyte cholesterol disorders in DKD are still unknown. ADP-ribosylation factor 6 (Arf6) is a small GTPase with pleiotropic effects and has previously been shown to regulate ATP-binding cassette transporter 1 (ABCA1) recycling, and thus, cholesterol homeostasis. However, Arf6 involvement in cholesterol metabolism in podocytes is scarce. To investigate the role of Arf6 in cholesterol modulation in podocytes, the effect of Arf6 on the regulation of the cholesterol transporter ABCA1 was studied in podocytes in vivo and in vitro. Intracellular cholesterol accumulation was significantly increased in podocytes from streptozotocin-induced diabetic rats and that hyperglycemia downregulated the expression of Arf6. Arf6 knockdown could cause ABCA1 recycling disorders, and thus, further aggravate cholesterol accumulation in podocytes under high-glucose (HG) conditions. Our results demonstrate that HG-induced cholesterol accumulation and cellular injury in podocytes may be related to the recycling disorder of ABCA1 caused by the downexpression of Arf6 in DKD.
- Published
- 2019
39. The Small GTPase ARF6 Activates PI3K in Melanoma to Induce a Prometastatic State
- Author
-
Jae Hyuk Yoo, Andrea H. Bild, Lehi Acosta-Alvarez, Dean Y. Li, Aaron Rogers, Samuel W. Brady, Roger K. Wolff, Sheri L. Holmen, Shannon J. Odelberg, David A. Kircher, Lise K. Sorensen, Jingfu Peng, Tara M. Mleynek, Donghan Shin, Allie H. Grossmann, and Coulson P. Rich
- Subjects
Proto-Oncogene Proteins B-raf ,0301 basic medicine ,Cancer Research ,Lung Neoplasms ,Skin Neoplasms ,Melanoma, Experimental ,Mice, SCID ,Biology ,Article ,Metastasis ,Phosphatidylinositol 3-Kinases ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,medicine ,Animals ,Humans ,PTEN ,Small GTPase ,Neoplasm Metastasis ,Melanoma ,Protein kinase B ,Cyclin-Dependent Kinase Inhibitor p16 ,PI3K/AKT/mTOR pathway ,ADP-Ribosylation Factors ,PTEN Phosphohydrolase ,medicine.disease ,Mice, Mutant Strains ,030104 developmental biology ,Oncology ,ADP-Ribosylation Factor 6 ,030220 oncology & carcinogenesis ,Invadopodia ,Cancer research ,biology.protein ,Guanosine Triphosphate ,Proto-Oncogene Proteins c-akt - Abstract
Melanoma has an unusual capacity to spread in early-stage disease, prompting aggressive clinical intervention in very thin primary tumors. Despite these proactive efforts, patients with low-risk, low-stage disease can still develop metastasis, indicating the presence of permissive cues for distant spread. Here, we show that constitutive activation of the small GTPase ARF6 (ARF6Q67L) is sufficient to accelerate metastasis in mice with BRAFV600E/Cdkn2aNULL melanoma at a similar incidence and severity to Pten loss, a major driver of PI3K activation and melanoma metastasis. ARF6Q67L promoted spontaneous metastasis from significantly smaller primary tumors than PTENNULL, implying an enhanced ability of ARF6-GTP to drive distant spread. ARF6 activation increased lung colonization from circulating melanoma cells, suggesting that the prometastatic function of ARF6 extends to late steps in metastasis. Unexpectedly, ARF6Q67L tumors showed upregulation of Pik3r1 expression, which encodes the p85 regulatory subunit of PI3K. Tumor cells expressing ARF6Q67L displayed increased PI3K protein levels and activity, enhanced PI3K distribution to cellular protrusions, and increased AKT activation in invadopodia. ARF6 is necessary and sufficient for activation of both PI3K and AKT, and PI3K and AKT are necessary for ARF6-mediated invasion. We provide evidence for aberrant ARF6 activation in human melanoma samples, which is associated with reduced survival. Our work reveals a previously unknown ARF6-PI3K-AKT proinvasive pathway, it demonstrates a critical role for ARF6 in multiple steps of the metastatic cascade, and it illuminates how melanoma cells can acquire an early metastatic phenotype in patients. Significance: These findings reveal a prometastatic role for ARF6 independent of tumor growth, which may help explain how melanoma spreads distantly from thin, early-stage primary tumors.
- Published
- 2019
40. GEP100/ARF6 regulates VEGFR2 signaling to facilitate high-glucose-induced epithelial-mesenchymal transition and cell permeability in retinal pigment epithelial cells
- Author
-
Shan-Shan Chen, Shu-Hua Fu, Zhi-Peng You, Fan Xiong, Ting Liu, Shu-Rong Li, and Zhong-Yi Yang
- Subjects
0301 basic medicine ,Cell Membrane Permeability ,Epithelial-Mesenchymal Transition ,Physiology ,Cell ,Retinal Pigment Epithelium ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Pigment ,0302 clinical medicine ,medicine ,Animals ,Guanine Nucleotide Exchange Factors ,Humans ,Epithelial–mesenchymal transition ,Cell permeability ,Cells, Cultured ,Retinal pigment epithelium ,Transition (genetics) ,ADP-Ribosylation Factors ,Chemistry ,Retinal ,Cell Biology ,Diabetic retinopathy ,medicine.disease ,Vascular Endothelial Growth Factor Receptor-2 ,Cell biology ,Mice, Inbred C57BL ,Glucose ,030104 developmental biology ,medicine.anatomical_structure ,ADP-Ribosylation Factor 6 ,030220 oncology & carcinogenesis ,visual_art ,visual_art.visual_art_medium ,Signal Transduction - Abstract
Cell permeability and epithelial-mesenchymal transition (EMT) were found to be enhanced in diabetic retinopathy, and the aim of this study was to investigate the underlying mechanism. ARPE-19 cell line or primary retinal pigment epithelial (RPE) cells were cultured under high or normal glucose conditions. Specific shRNAs were employed to knock down ADP-ribosylation factor 6 (ARF6), GEP100, or VEGF receptor 2 (VEGFR2) in ARPE-19 or primary RPE cells. Cell migration ability was measured using Transwell assay. Western blotting was used to measure indicated protein levels. RPE cells treated with high glucose showed increased cell migration, paracellular permeability, EMT, and expression of VEGF. Knockdown of VEGFR2 inhibited the high-glucose-induced effects on RPE cells via inactivation of ARF6 and MAPK pathways. Knockdown ARF6 or GEP100 led to inhibition of high-glucose-induced effects via inactivation of VEGFR2 pathway. Knockdown of ARF6, but not GEP100, decreased high-glucose-induced internalization of VEGFR2. High-glucose enhances EMT and cell permeability of RPE cells through activation of VEGFR2 and ARF6/GEP100 pathways, which form a positive feedback loop to maximize the activation of VEGF/VEGFR2 signaling.
- Published
- 2019
41. Syndecan 1 is a critical mediator of macropinocytosis in pancreatic cancer
- Author
-
Timothy P. Heffernan, Shuxing Zhang, I. Lin Ho, Anirban Maitra, Alessandro Carugo, Liang Yan, Jason B. Fleming, Pingna Deng, Vandhana Ramamoorthy, Zhaohui Xu, Qiuyun Wang, Shan Jiang, Jun Yao, Wantong Yao, Piergiorgio Pettazzoni, Pingping Hou, Sahil Seth, Haoqiang Ying, Luigi Nezi, Zhi Tan, Hong Jiang, Jintan Liu, Giulio Draetta, Ziheng Chen, Ayumu Taguchi, Andrea Viale, Ningping Feng, Huamin Wang, Wei Wang, Grace J. Ma, Baoli Hu, Avnish Kapoor, Angela K. Deem, Johnathon L. Rose, Peter Den, Samir M. Hanash, Y. Alan Wang, and Ronald A. DePinho
- Subjects
Male ,0301 basic medicine ,endocrine system diseases ,Cell ,Context (language use) ,Biology ,medicine.disease_cause ,Article ,Syndecan 1 ,Malignant transformation ,Proto-Oncogene Proteins p21(ras) ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Pancreatic cancer ,medicine ,Animals ,Guanine Nucleotide Exchange Factors ,Humans ,neoplasms ,Cell Proliferation ,Multidisciplinary ,ADP-Ribosylation Factors ,Cell growth ,medicine.disease ,digestive system diseases ,Pancreatic Neoplasms ,030104 developmental biology ,medicine.anatomical_structure ,ADP-Ribosylation Factor 6 ,030220 oncology & carcinogenesis ,Disease Progression ,Cancer research ,Pinocytosis ,Female ,Syndecan-1 ,KRAS ,Signal transduction ,Carcinoma, Pancreatic Ductal ,Signal Transduction - Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains recalcitrant to all forms of cancer treatment and carries a five-year survival rate of only 8%1. Inhibition of oncogenic KRAS (hereafter KRAS*), the earliest lesion in disease development that is present in more than 90% of PDACs, and its signalling surrogates has yielded encouraging preclinical results with experimental agents2–4. However, KRAS*-independent disease recurrence following genetic extinction of Kras* in mouse models anticipates the need for co-extinction strategies5,6. Multiple oncogenic processes are initiated at the cell surface, where KRAS* physically and functionally interacts to direct signalling that is essential for malignant transformation and tumour maintenance. Insights into the complexity of the functional cell-surface-protein repertoire (surfaceome) have been technologically limited until recently and—in the case of PDAC—the genetic control of the function and composition of the PDAC surfaceome in the context of KRAS* signalling remains largely unknown. Here we develop an unbiased, functional target-discovery platform to query KRAS*-dependent changes of the PDAC surfaceome, which reveals syndecan 1 (SDC1, also known as CD138) as a protein that is upregulated at the cell surface by KRAS*. Localization of SDC1 at the cell surface—where it regulates macropinocytosis, an essential metabolic pathway that fuels PDAC cell growth—is essential for disease maintenance and progression. Thus, our study forges a mechanistic link between KRAS* signalling and a targetable molecule driving nutrient salvage pathways in PDAC and validates oncogene-driven surfaceome annotation as a strategy to identify cancer-specific vulnerabilities. In an inducible mouse model of pancreatic ductal adenocarcinoma, the signalling defect that underlies 90% of these tumours causes increased cell-surface expression of syndecan 1, leading to misregulation of macropinocytosis, and linking the defective signalling with nutrient-salvage pathways.
- Published
- 2019
42. Cell-type–specific, multicolor labeling of endogenous proteins with split fluorescent protein tags in Drosophila
- Author
-
Rie Kamiyama, Fangchao Jiang, Daichi Kamiyama, Miyuki A Fitch, Abhijit Marar, Peiwei Liu, Ryo Tamura, Jin Xie, and Kota Banzai
- Subjects
Green Fluorescent Proteins ,ved/biology.organism_classification_rank.species ,Biology ,Protein Engineering ,Fluorescence ,Green fluorescent protein ,Animals, Genetically Modified ,03 medical and health sciences ,0302 clinical medicine ,Tandem repeat ,Fluorescence microscope ,Animals ,Drosophila Proteins ,Coding region ,Model organism ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,Staining and Labeling ,ved/biology ,Protein engineering ,Biological Sciences ,Protein subcellular localization prediction ,Cell biology ,Microscopy, Fluorescence ,ADP-Ribosylation Factor 6 ,Drosophila ,030217 neurology & neurosurgery ,Function (biology) ,Transcription Factors - Abstract
The impact of the Drosophila experimental system on studies of modern biology cannot be understated. The ability to tag endogenously expressed proteins is essential to maximize the use of this model organism. Here, we describe a method for labeling endogenous proteins with self-complementing split fluorescent proteins (split FPs) in a cell-type–specific manner in Drosophila. A short fragment of an FP coding sequence is inserted into a specific genomic locus while the remainder of the FP is expressed using an available GAL4 driver line. In consequence, complementation fluorescence allows examination of protein localization in particular cells. Besides, when inserting tandem repeats of the short FP fragment at the same genomic locus, we can substantially enhance the fluorescence signal. The enhanced signal is of great value in live-cell imaging at the subcellular level. We can also accomplish a multicolor labeling system with orthogonal split FPs. However, other orthogonal split FPs do not function for in vivo imaging besides split GFP. Through protein engineering and in vivo functional studies, we report a red split FP that we can use for duplexed visualization of endogenous proteins in intricate Drosophila tissues. Using the two orthogonal split FP systems, we have simultaneously imaged proteins that reside in distinct subsynaptic compartments. Our approach allows us to study the proximity between and localization of multiple proteins endogenously expressed in essentially any cell type in Drosophila.
- Published
- 2021
43. EFA6 in Axon Regeneration, as a Microtubule Regulator and as a Guanine Nucleotide Exchange Factor
- Author
-
Gilberto Gonzalez and Lizhen Chen
- Subjects
0301 basic medicine ,QH301-705.5 ,integrin ,Integrin ,Regulator ,Review ,Microtubules ,EFA6 ,03 medical and health sciences ,0302 clinical medicine ,Microtubule ,medicine ,Guanine Nucleotide Exchange Factors ,Humans ,ARF6 ,Biology (General) ,Axon ,Cytoskeleton ,biology ,ADP-Ribosylation Factors ,Chemistry ,Regeneration (biology) ,Cell Membrane ,axon regeneration ,General Medicine ,microtubule dynamics ,Axons ,Nerve Regeneration ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,nervous system ,ADP-Ribosylation Factor 6 ,axon injury ,biology.protein ,Guanine nucleotide exchange factor ,Reprogramming ,030217 neurology & neurosurgery - Abstract
Axon regeneration after injury is a conserved biological process that involves a large number of molecular pathways, including rapid calcium influx at injury sites, retrograde injury signaling, epigenetic transition, transcriptional reprogramming, polarized transport, and cytoskeleton reorganization. Despite the numerous efforts devoted to understanding the underlying cellular and molecular mechanisms of axon regeneration, the search continues for effective target molecules for improving axon regeneration. Although there have been significant historical efforts towards characterizing pro-regenerative factors involved in axon regeneration, the pursuit of intrinsic inhibitors is relatively recent. EFA6 (exchange factor for ARF6) has been demonstrated to inhibit axon regeneration in different organisms. EFA6 inhibition could be a promising therapeutic strategy to promote axon regeneration and functional recovery after axon injury. This review summarizes the inhibitory role on axon regeneration through regulating microtubule dynamics and through affecting ARF6 (ADP-ribosylation factor 6) GTPase-mediated integrin transport.
- Published
- 2021
44. microRNA-145-5p Inhibits Migration, Invasion, and Metastasis in Hepatocellular Carcinoma by Inhibiting ARF6
- Author
-
Shuo Wang, Tianjiao Wang, and Pengcheng Gu
- Subjects
0301 basic medicine ,proliferation ,ADP-ribosylation factor 6 ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,microRNA ,metastasis ,Medicine ,neoplasms ,Original Research ,business.industry ,Cell growth ,microRNA-145-5p ,Cancer ,hepatocellular carcinoma ,invasion ,medicine.disease ,digestive system diseases ,030104 developmental biology ,Oncology ,Apoptosis ,Cell culture ,Cancer Management and Research ,030220 oncology & carcinogenesis ,Hepatocellular carcinoma ,Cancer research ,business ,Wound healing - Abstract
Shuo Wang,1 Tianjiao Wang,2 Pengcheng Gu1 1Department of Orthopedics Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, People’s Republic of China; 2Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, Zhejiang, 310051, People’s Republic of ChinaCorrespondence: Pengcheng GuDepartment of Orthopedics Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, Zhejiang, 310006, People’s Republic of ChinaTel +86-571-87236848Email 1192014@zju.edu.cnPurpose: Hepatocellular carcinoma (HCC) has the fourth highest rate of mortality among the different types of cancer worldwide. This study aimed to investigate the functions of microRNA-145-5p and AFR6 on migration, invasion and metastasis in HCC.Methods: A total of 150 pairs of tumor and their matched adjacent nontumor liver tissues were collected from HCC patients. Expressions of microRNA-145-5p and AFR6 were measured by real-time PCR in HCC tissues and in HCC cell lines. The correlations between microRNA-145-5p and HCC prognosis were investigated. The proliferation, migration, invasion, cell cycle progression, and apoptosis of HCCLM3 cells were evaluated with CCK8, wound healing, transwell, and flow cytometric experiments.Results: The expression of miR-145-5p was confirmed to be downregulated not only in HCC tissues but also in several HCC cell lines compared with normal controls. A low expression level of miR-145-5p was notably associated with poor prognosis in patients with HCC and certain characteristics of metastatic tumors. In vitro, miR-145-5p negatively regulated cell proliferation, migration, and invasion and induced apoptosis in HCCLM3 cells. Subsequent experiments further verified that ARF6 is a novel target of miR-145-5p and is significantly overexpressed in HCC tissues. Overexpression of ARF6 circumvented the effects of miR-145-5p in HCCLM3 cells.Conclusion: miR-145-5p may play a pivotal role in HCC metastasis via regulating ARF6, and these findings may both provide further insights into the key factors of HCC metastasis and prove to be useful in the development of novel treatment options for HCC.Keywords: microRNA-145-5p, ADP-ribosylation factor 6, hepatocellular carcinoma, proliferation, invasion, metastasis
- Published
- 2021
45. Long non‐coding RNA KCNQ1OT1 promotes the progression of gastric cancer via the miR‐145‐5p/ARF6 axis
- Author
-
Xianchang Yu, Xiongdong Zhong, Ni Gu, Lei Chen, Xiaoyan Wen, and Kang Sui
- Subjects
Male ,0301 basic medicine ,Apoptosis ,Flow cytometry ,03 medical and health sciences ,0302 clinical medicine ,Stomach Neoplasms ,Cell Line, Tumor ,miR‐145‐5p ,Drug Discovery ,Genetics ,medicine ,Humans ,ARF6 ,Viability assay ,Molecular Biology ,Research Articles ,Genetics (clinical) ,Aged ,Cell Proliferation ,Gene knockdown ,medicine.diagnostic_test ,Chemistry ,gastric cancer ,Transfection ,Middle Aged ,Molecular biology ,KCNQ1OT1 ,Antisense RNA ,Gene Expression Regulation, Neoplastic ,Blot ,MicroRNAs ,030104 developmental biology ,ADP-Ribosylation Factor 6 ,Potassium Channels, Voltage-Gated ,Cell culture ,Gene Knockdown Techniques ,030220 oncology & carcinogenesis ,Disease Progression ,Molecular Medicine ,Female ,Signal Transduction ,Research Article - Abstract
Background Long non‐coding RNA KCNQ1 opposite strand/antisense transcript one gene (KCNQ1OT1) has been reported to be involved in the progression of many types of human cancer, whereas its role in gastric cancer (GC) remains unknown. The present study aimed to investigate the role of KCNQ1OT1 in GC. Methods In total, 25 GC tissues and adjacent normal tissues were collected. The expression of KCNQ1OT1, miR‐145‐5p and ARF6 in GC tissues and cell lines was detected by quantitative reverse transcriptase‐polymerase chain reaction or western blotting. Bioinformatics analysis and a dual luciferase reporter assay were performed to determine the relationship between KCNQ1OT1 and miR‐145‐5p or miR‐145‐5p and ARF6. Gain‐ and loss‐of function of KCNQ1OT1 and miR‐145‐5p were achieved to confirm their roles in GC cells. Cell counting kit‐8, colony formation and flow cytometry assays were used to evaluate cell viability, proliferation and apoptosis. A xenograft tumor model was established with BGC803 tumor cells transfected with sh‐KCNQ1OT1 or empty vector to determine the role of LINC01089 in vivo. Results The expression levels of KCNQ1OT1 were markedly elevated in GC tissues and cells. Knockdown of KCNQ1OT1 inhibited GC tumor growth, reduced GC cell viability and colony formation, and induced GC cell apoptosis. The expression levels of miR‐145‐5p were significantly decreased in GC cells and correlated with the expression of KCNQ1OT1 in GC tumors. Moreover, KCNQ1OT1 directly binds with miR‐145‐5p, which is targeting ARF6. Knockdown of KCNQ1OT1 increased the expression levels of miR‐145‐5p. Inhibition of miR‐145‐5p increased the expression levels of KCNQ1OT1 and also attenuated the effects of knockdown of KCNQ1OT1 on the viability, proliferation and apoptosis of GC cells. In addition, overexpression of miR‐145‐5p reduced GC cell viability and colony formation and induced GC cell apoptosis, whereas overexpression of ARF6 attenuated the effects of overexpression of miR‐145‐5p on GC cell viability, colony formation and apoptosis. Conclusions KCNQ1OT1 can promote GC progression through the miR‐145‐5p/ARF6 axis. KCNQ1OT1 may serve as a therapeutic target and a diagnostic biomarker of GC., KCNQ1OT1 promotes the progression of gastric cancer by regulating the miR‐145‐5p/ARF6 axis. KCNQ1OT1 may serve as a therapeutic target and a diagnostic biomarker of gastric cancer.
- Published
- 2021
46. ADP Ribosylation Factor 6 Promotes Contraction and Proliferation, Suppresses Apoptosis and Is Specifically Inhibited by NAV2729 in Prostate Stromal Cells
- Author
-
Martin Hennenberg, Beata Rutz, Bingsheng Li, Christian G. Stief, Stephanie Schneider, Oliver T. Keppler, Ruixiao Wang, and Anna Ciotkowska
- Subjects
Male ,Programmed cell death ,Stromal cell ,Cell Survival ,Apoptosis ,Pyrimidinones ,Chlorobenzenes ,Prostate ,medicine ,Humans ,Gene knockout ,Actin ,Cell Line, Transformed ,Cell Proliferation ,Pharmacology ,Chemistry ,ADP-Ribosylation Factors ,Smooth muscle contraction ,Hyperplasia ,medicine.disease ,Cell biology ,medicine.anatomical_structure ,ADP-Ribosylation Factor 6 ,Gene Knockdown Techniques ,Molecular Medicine ,Pyrazoles ,Stromal Cells - Abstract
The presumed ADP ribosylation factor (ARF) 6 inhibitor NAV2729 inhibits human prostate smooth muscle contraction and proliferation of stromal cells, which are driving factors of voiding symptoms in benign prostatic hyperplasia (BPH). However, its specificity and a confirmed role of ARF6 for smooth muscle contraction are still pending. Here, we generated monoclonal ARF6 knockouts in human prostate stromal cells (WPMY-1), and characterized phenotypes of contractility, growth-related functions, and susceptibility to NAV2729 in knockout and control clones. ARF6 knockout was verified by Western blot. Knockout clones showed impaired contraction and actin organization, reduced proliferation and viability, and increased apoptosis and cell death. In ARF6-expressing control clones, NAV2729 (5 µM) strongly inhibited contraction (67% inhibition across all three control clones), actin organization (72%), proliferation (97%), and viability (up to 82%), and increased apoptosis (5-fold) and cell death (6-fold). In ARF6 knockouts, effects of NAV2729 (5 µM) were widely reduced, including lacking or minor effects on contractions (0% inhibition across all three knockout clones), actin (18%) and proliferation (13%), and lacking increases of apoptosis and cell death. Viability was reduced by NAV2729 with an IC50 of 3.3 µM across all three ARF6 control clones, but of 4.5–8.2 µM in ARF6 knockouts. In conclusion, ARF6 promotes prostate smooth muscle contraction and proliferation of stromal cells. Both are inhibited by NAV2729, which showed high specificity for ARF6 up to 5 µM and represents an attractive compound in the context of BPH. Considering the relevance of smooth muscle-based diseases, shared roles of ARF6 in other smooth muscle types merit further investigation. SIGNIFICANCE STATEMENT By knockout of ARF6 in prostate stromal cells, this study demonstrates the involvement of ARF6 in promotion of prostate smooth muscle contraction and stromal growth, and defines concentration ranges for their ARF6-specific inhibition by NAV2729. Besides the context of benign prostatic hyperplasia and lower urinary tract symptoms, analog ARF6 functions in contraction and growth appear possible in other smooth muscle-rich organs, which merits further attention considering the high clinical relevance of smooth muscle-based diseases.
- Published
- 2021
47. FilGAP, a GAP protein for Rac, regulates front‐rear polarity and tumor cell migration through the ECM
- Author
-
Yasutaka Ohta, Norito Kambara, Mamiko Mori, and Koji Saito
- Subjects
0301 basic medicine ,Cell ,Motility ,Biochemistry ,Filamentous actin ,Extracellular matrix ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cell Movement ,Cell Line, Tumor ,Cell polarity ,Genetics ,medicine ,Humans ,Small GTPase ,Phosphatidylinositol ,Molecular Biology ,rho-Associated Kinases ,GTPase-Activating Proteins ,Cell Polarity ,Extracellular Matrix ,Cell biology ,Pleckstrin homology domain ,Actin Cytoskeleton ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,ADP-Ribosylation Factor 6 ,030217 neurology & neurosurgery ,Biotechnology - Abstract
Migrating tumor cells are characterized by a sustained front-rear asymmetry, with a front enriched in filamentous actin, which is induced by Rho small GTPase Rac. Regulation of Rac activity by its regulators should be required for effective motility. Here, we show that FilGAP, a GTPase-activating protein (GAP) for Rac, controls front-rear polarity and contributes to maintain effective tumor cell migration through the extracellular matrix (ECM). Overexpression of FilGAP in breast cancer cells induced polarized morphology and led to increased migration speed in collagen matrices, while depletion of FilGAP impaired the cell polarity and migration. FilGAP localizes to the cell front through its pleckstrin-homology (PH) domain in a phosphatidylinositol 3,4,5-trisphosphate (PIP3)-dependent manner and appears to inactivate Rac at its site. We found that the affinity of PH domain to PIP3 is critically involved in the maintenance of cell polarity. Moreover, small GTPase ADP-ribosylation factor 6 (Arf6), which binds to the FilGAP PH domain, also regulates FilGAP-mediated cell polarity and migration of breast cancer cells. We propose that FilGAP regulates front-rear polarity through its PIP3 and Arf6 binding in tumor cell migration through the ECM.
- Published
- 2021
48. An ARF GTPase module promoting invasion and metastasis through regulating phosphoinositide metabolism
- Author
-
Tamas Yelland, Jennifer P. Morton, Eva C Freckmann, Sara Zanivan, Lynn McGarry, Shehab Ismail, Hing Y. Leung, Konstantina Nikolatou, Laura C. A. Galbraith, Karen Blyth, Rachana Patel, David M. Bryant, Álvaro Román-Fernández, Marisa Nacke, Elke Markert, Sergio Lilla, Emma Shanks, Emma Sandilands, and Susan M. Mason
- Subjects
0301 basic medicine ,Male ,ADP ribosylation factor ,Carcinogenesis ,Science ,Regulator ,General Physics and Astronomy ,Mice, Nude ,Collective cell migration ,Biology ,Phosphatidylinositols ,General Biochemistry, Genetics and Molecular Biology ,Article ,Metastasis ,03 medical and health sciences ,Mice ,0302 clinical medicine ,In vivo ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Guanine Nucleotide Exchange Factors ,Humans ,Neoplasm Metastasis ,Multidisciplinary ,Science & Technology ,Cell growth ,ADP-Ribosylation Factors ,Cancer ,Growth factor signalling ,General Chemistry ,medicine.disease ,In vitro ,Cell biology ,Cell invasion ,Multidisciplinary Sciences ,030104 developmental biology ,Signalling ,ADP-Ribosylation Factor 6 ,030220 oncology & carcinogenesis ,Cell polarity ,Heterografts ,Science & Technology - Other Topics ,Proto-Oncogene Proteins c-akt ,Low Density Lipoprotein Receptor-Related Protein-1 ,Signal Transduction - Abstract
The signalling pathways underpinning cell growth and invasion use overlapping components, yet how mutually exclusive cellular responses occur is unclear. Here, we report development of 3-Dimensional culture analyses to separately quantify growth and invasion. We identify that alternate variants of IQSEC1, an ARF GTPase Exchange Factor, act as switches to promote invasion over growth by controlling phosphoinositide metabolism. All IQSEC1 variants activate ARF5- and ARF6-dependent PIP5-kinase to promote PI(3,4,5)P3-AKT signalling and growth. In contrast, select pro-invasive IQSEC1 variants promote PI(3,4,5)P3 production to form invasion-driving protrusions. Inhibition of IQSEC1 attenuates invasion in vitro and metastasis in vivo. Induction of pro-invasive IQSEC1 variants and elevated IQSEC1 expression occurs in a number of tumour types and is associated with higher-grade metastatic cancer, activation of PI(3,4,5)P3 signalling, and predicts long-term poor outcome across multiple cancers. IQSEC1-regulated phosphoinositide metabolism therefore is a switch to induce invasion over growth in response to the same external signal. Targeting IQSEC1 as the central regulator of this switch may represent a therapeutic vulnerability to stop metastasis., The signalling pathways underpinning cell growth and invasion use overlapping components, yet how mutually exclusive responses occur is unclear. Here, the authors show that alternate isoforms of the ARF GTPase exchange factor IQSEC1 direct phosphoinositide metabolism to control this switch.
- Published
- 2021
49. Chlortetracycline, a Novel Arf Inhibitor That Decreases the Arf6-Dependent Invasive Properties of Breast Cancer Cells
- Author
-
Sophie Abelanet, Eric Macia, Monserrat Vazquez-Rojas, Mohamed Mehiri, Isabelle Mus-Veteau, Alessia Robiolo, Racha Fayad, Frédéric Luton, Michel Franco, Fabien Fontaine-Vive, Institut de pharmacologie moléculaire et cellulaire (IPMC), Centre National de la Recherche Scientifique (CNRS)-Université Nice Sophia Antipolis (... - 2019) (UNS), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Université Côte d'Azur (UCA), Institut de Chimie de Nice (ICN), Université Nice Sophia Antipolis (... - 2019) (UNS), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Institut de Chimie du CNRS (INC)-Centre National de la Recherche Scientifique (CNRS)-Université Côte d'Azur (UCA), and Centre National de la Recherche Scientifique (CNRS)
- Subjects
Chlortetracycline ,[SDV]Life Sciences [q-bio] ,Cell ,Antibiotics ,Pharmaceutical Science ,Disease ,small G protein ,Analytical Chemistry ,0302 clinical medicine ,Drug Discovery ,Arf6 ,media_common ,0303 health sciences ,ADP-Ribosylation Factors ,cell invasion ,3. Good health ,Gene Expression Regulation, Neoplastic ,inhibitor ,medicine.anatomical_structure ,Chemistry (miscellaneous) ,030220 oncology & carcinogenesis ,Molecular Medicine ,Female ,medicine.drug ,Signal Transduction ,Drug ,medicine.drug_class ,media_common.quotation_subject ,High-throughput screening ,Breast Neoplasms ,Article ,lcsh:QD241-441 ,03 medical and health sciences ,Breast cancer ,breast cancer ,lcsh:Organic chemistry ,Cell Line, Tumor ,medicine ,Humans ,[CHIM]Chemical Sciences ,Neoplasm Invasiveness ,Physical and Theoretical Chemistry ,chlortetracycline ,030304 developmental biology ,Cell Proliferation ,business.industry ,Organic Chemistry ,medicine.disease ,In vitro ,ADP-Ribosylation Factor 6 ,Cancer research ,business - Abstract
International audience; Breast cancer is a major disease for women worldwide, where mortality is associated with tumour cell dissemination to distant organs. While the number of efficient anticancer therapies increased in the past 20 years, treatments targeting the invasive properties of metastatic tumour cells are still awaited. Various studies analysing invasive breast cancer cell lines have demonstrated that Arf6 is an important player of the migratory and invasive processes. These observations make Arf6 and its regulators potential therapeutic targets. As of today, no drug effective against Arf6 has been identified, with one explanation being that the activation of Arf6 is dependent on the presence of lipid membranes that are rarely included in drug screening. To overcome this issue we have set up a fluorescence-based high throughput screening that follows overtime the activation of Arf6 at the surface of lipid membranes. Using this unique screening assay, we isolated several compounds that affect Arf6 activation, among which the antibiotic chlortetracycline (CTC) appeared to be the most promising. In this report, we describe CTC in vitro biochemical characterization and show that it blocks both the Arf6-stimulated collective migration and cell invasion in a 3D collagen I gel of the invasive breast cancer cell line MDA-MB-231. Thus, CTC appears as a promising hit to target deadly metastatic dissemination and a powerful tool to unravel the molecular mechanisms of Arf6-mediated invasive processes.
- Published
- 2021
50. SNX-3 mediates retromer-independent tubular endosomal recycling by opposing EEA-1-facilitated trafficking
- Author
-
Rongying Zhang, Xuemeng Shi, Yuan Wang, Qiaoju Kang, Tao Xu, Yangli Tian, Qifeng Xu, Nali Zhang, and Huan Ye
- Subjects
Cancer Research ,Retromer ,Nematoda ,Endocytic cycle ,Cell Membranes ,Vesicular Transport Proteins ,QH426-470 ,Biochemistry ,0302 clinical medicine ,RNA interference ,Phosphatidylinositol Phosphates ,Cargo Proteins ,Medicine and Health Sciences ,Protein Isoforms ,Sorting Nexins ,Genetics (clinical) ,Caenorhabditis elegans ,0303 health sciences ,biology ,ADP-Ribosylation Factors ,Eukaryota ,Animal Models ,Cell biology ,ESCRT complex ,Intestines ,Nucleic acids ,medicine.anatomical_structure ,Experimental Organism Systems ,Genetic interference ,Caenorhabditis Elegans ,Epigenetics ,Cellular Structures and Organelles ,Anatomy ,Signal Transduction ,Research Article ,Endosome ,Endosomes ,Research and Analysis Methods ,EEA1 ,03 medical and health sciences ,Model Organisms ,Lysosome ,medicine ,Genetics ,Animals ,Humans ,Vesicles ,Caenorhabditis elegans Proteins ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,030304 developmental biology ,Endosomal Sorting Complexes Required for Transport ,Cell Membrane ,Organisms ,Biology and Life Sciences ,Proteins ,Membrane Proteins ,Biological Transport ,Epithelial Cells ,Cell Biology ,biology.organism_classification ,Invertebrates ,Gastrointestinal Tract ,Gene Expression Regulation ,ADP-Ribosylation Factor 6 ,Proteolysis ,Animal Studies ,Caenorhabditis ,RNA ,Gene expression ,Lysosomes ,Digestive System ,Zoology ,030217 neurology & neurosurgery ,HeLa Cells ,Cargo Receptors - Abstract
Early endosomes are the sorting hub on the endocytic pathway, wherein sorting nexins (SNXs) play important roles for formation of the distinct membranous microdomains with different sorting functions. Tubular endosomes mediate the recycling of clathrin-independent endocytic (CIE) cargoes back toward the plasma membrane. However, the molecular mechanism underlying the tubule formation is still poorly understood. Here we screened the effect on the ARF-6-associated CIE recycling endosomal tubules for all the SNX members in Caenorhabditis elegans (C. elegans). We identified SNX-3 as an essential factor for generation of the recycling tubules. The loss of SNX-3 abolishes the interconnected tubules in the intestine of C. elegans. Consequently, the surface and total protein levels of the recycling CIE protein hTAC are strongly decreased. Unexpectedly, depletion of the retromer components VPS-26/-29/-35 has no similar effect, implying that the retromer trimer is dispensable in this process. We determined that hTAC is captured by the ESCRT complex and transported into the lysosome for rapid degradation in snx-3 mutants. Interestingly, EEA-1 is increasingly recruited on early endosomes and localized to the hTAC-containing structures in snx-3 mutant intestines. We also showed that SNX3 and EEA1 compete with each other for binding to phosphatidylinositol-3-phosphate enriching early endosomes in Hela cells. Our data demonstrate for the first time that PX domain-only C. elegans SNX-3 organizes the tubular endosomes for efficient recycling and retrieves the CIE cargo away from the maturing sorting endosomes by competing with EEA-1 for binding to the early endosomes. However, our results call into question how SNX-3 couples the cargo capture and membrane remodeling in the absence of the retromer trimer complex., Author summary Trafficking of internalized materials through the endolysosomal system is essential for the maintenance of homeostasis and signaling regulation in all eukaryotic cells. Early endosomes are the sorting hub on the endocytic pathway. After internalization, the plasma membrane lipid, proteins, and invading pathogens are delivered to early endosomes for further degradation in lysosomes or for retrieval to the plasma membrane or the trans-Golgi network for reuse. However, when, where and by what mechanism various cargo proteins are sorted from each other and into the different pathways largely remain to be explored. Here, we identified SNX-3, a PX-domain only sorting nexin family member, as a novel regulator for the tubular endosomes underlying recycling of a subset of CIE cargoes. Compared with EEA-1, the superior recruitment of SNX-3 at the CIE-derived subpopulation of endosomes is critical for preventing these endosomes from converging to the classical sorting endosomes and subsequently into the multivesicular endosomal pathway. We speculate that through a spatio-temporal interplay with the retromer, SNX-3 is involved in different recycling transport carriers. Our finding of SNX-3’s role in modulating the formation of tubular endosomes provides insight into the sorting and trafficking of CIE pathways.
- Published
- 2021
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.