1
|
K HS, R G, Veeraraghavan VP, Ramani P. Netrin 1 as a biomarker in cancer: scoping diagnostic, prognostic, and therapeutic perspectives with a focus on oral squamous cell carcinoma. JOURNAL OF STOMATOLOGY, ORAL AND MAXILLOFACIAL SURGERY 2024; 125:101982. [PMID: 39067640 DOI: 10.1016/j.jormas.2024.101982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 07/19/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
Goal of the review: The utilization of biomarkers to predict cancer risk, prognosis, and treatment outcomes is paramount. Netrin-1 (NTN1), known for its role in commissural axon guidance during embryonic development, has emerged as a versatile molecule with significant implications in cancer and neurobiology. Structurally resembling laminin, Netrin-1 regulates neuronal connectivity and plasticity in adulthood, influencing axonal and dendritic growth, neurotransmission, and cell migration. In addition to its neurological functions, Netrin-1 is increasingly recognized for its involvement in maintaining epithelial tissue and its regulatory roles in fundamental cellular processes, including adhesion, proliferation, differentiation, apoptosis, and angiogenesis. In cancer biology, Netrin-1's interactions with its receptors, such as DCC [Deleted in Colorectal Cancer] and UNC5 (a homolog of DCC), have been implicated in tumor progression across various physiological systems. Elevated levels of Netrin-1 in colorectal cancer and head and neck squamous cell carcinoma are correlated with increased tumorigenic potential, mediated through pathways involving NFκB activation and anti-apoptotic mechanisms. Mechanically induced hypermethylation and downstream signaling cascades that inhibit apoptosis and promote cell survival are observed upon Netrin-1 binding to DCC. Furthermore, Netrin-1 shows promise as a biomarker for detecting inflammatory activity in diseases such as multiple sclerosis and as a potential diagnostic, prognostic, and therapeutic indicator in oral squamous cell carcinoma. Elevated levels of Netrin-1 in bodily fluids, alongside immunohistochemical evidence, support its potential as a valuable clinical marker in cancer management. This abstract emphasizes Netrin-1's diverse biological roles, underscoring its potential as a diagnostic tool and therapeutic target in cancer research. The need for further exploration of Netrin-1's molecular interactions and clinical applications is urgent and crucial to advance personalized medicine approaches and enhance patient outcomes in oncology and neurology.
Collapse
Affiliation(s)
- Hema Shree K
- Department of Oral and Maxillofacial Pathology, Saveetha Dental College and Hospitals, Saveetha Institute Medical and Technical Science, Saveetha University, Chennai, India
| | - Gayathri R
- Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute Medical and Technical Science, Saveetha University, Chennai, India.
| | - Vishnu Priya Veeraraghavan
- Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute Medical and Technical Science, Saveetha University, Chennai, India
| | - Pratibha Ramani
- Department of Oral and Maxillofacial Pathology, Saveetha Dental College and Hospitals, Saveetha Institute Medical and Technical Science, Saveetha University, Chennai, India
| |
Collapse
|
2
|
Jiang Y, Sachdeva K, Goulbourne CN, Berg MJ, Peddy J, Stavrides PH, Pensalfini A, Pawlik M, Whyte L, Balapal BS, Shivakumar S, Bleiwas C, Smiley JF, Mathews PM, Nixon RA. Increased neuronal expression of the early endosomal adaptor APPL1 leads to endosomal and synaptic dysfunction with cholinergic neurodegeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.19.613736. [PMID: 39345644 PMCID: PMC11430014 DOI: 10.1101/2024.09.19.613736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Dysfunction of the endolysosomal system within neurons is a prominent feature of Alzheimer's disease (AD) pathology. Multiple AD-risk factors are known to cause hyper-activity of the early-endosome small GTPase rab5, resulting in neuronal endosomal pathway disruption. APPL1, an important rab5 effector protein, is an interface between endosomal and neuronal function through a rab5-activating interaction with the BACE1-generated C-terminal fragment (βCTF or C99) of the amyloid precursor protein (APP), a pathogenic APP fragment generated within endolysosomal compartments. To better understand the role of APPL1 in the AD endosomal phenotype, we generated a transgenic mouse model over-expressing human APPL1 within neurons (Thy1-APPL1 mice). Consistent with the important endosomal regulatory role of APPL1, Thy1-APPL1 mice have enlarged neuronal early endosomes and increased synaptic endocytosis due to increased rab5 activation. We additionally demonstrate pathological consequences of APPL1 overexpression, including functional changes in hippocampal long-term potentiation (LTP) and long-term depression (LTD), as well as degeneration of the large projection cholinergic neurons of the basal forebrain and impairment of hippocampal-dependent memory. Our findings show that increased neuronal APPL1 levels lead to a cascade of pathological effects within neurons, including early endosomal alterations, synaptic dysfunction, and neurodegeneration. Multiple risk factors and molecular regulators, including APPL1 activity, are known to contribute to the endosomal dysregulation seen in the early stages of AD, and these findings further highlight the shared pathobiology and consequences to a neuron of early endosomal pathway disruption. Significance Statement Dysfunction in the endolysosomal system within neurons is a key feature of Alzheimer's disease (AD). Multiple AD risk factors lead to hyperactivity of the early-endosome GTPase rab5, disrupting neuronal pathways including the cholinergic circuits involved early in memory decline. APPL1, a crucial rab5 effector, connects endosomal and neuronal functions through its interaction with a specific amyloid precursor protein (APP) fragment generated within endosomes. To understand APPL1's role, a transgenic mouse model over-expressing human APPL1 in neurons (Thy1-APPL1 mice) was developed. These mice show enlarged early endosomes and increased synaptic endocytosis due to rab5 activation, resulting in impaired hippocampal long-term potentiation and depression, the degeneration of basal forebrain cholinergic neurons, and memory deficits, highlighting a pathological cascade mediated through APPL1 at the early endosome.
Collapse
|
3
|
Shi P, Tian Y, Xu F, Liu LN, Wu WH, Shi YZ, Dai AQ, Fang HY, Li KX, Xu C. Assessment of pathogenicity and functional characterization of APPL1 gene mutations in diabetic patients. World J Diabetes 2024; 15:275-286. [PMID: 38464380 PMCID: PMC10921161 DOI: 10.4239/wjd.v15.i2.275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/22/2023] [Accepted: 01/09/2024] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND Adaptor protein, phosphotyrosine interacting with PH domain and leucine zipper 1 (APPL1) plays a crucial role in regulating insulin signaling and glucose metabolism. Mutations in the APPL1 gene have been associated with the development of maturity-onset diabetes of the young type 14 (MODY14). Currently, only two mutations [c.1655T>A (p.Leu552*) and c.281G>A p.(Asp94Asn)] have been identified in association with this disease. Given the limited understanding of MODY14, it is imperative to identify additional cases and carry out comprehensive research on MODY14 and APPL1 mutations. AIM To assess the pathogenicity of APPL1 gene mutations in diabetic patients and to characterize the functional role of the APPL1 domain. METHODS Patients exhibiting clinical signs and a medical history suggestive of MODY were screened for the study. Whole exome sequencing was performed on the patients as well as their family members. The pathogenicity of the identified APPL1 variants was predicted on the basis of bioinformatics analysis. In addition, the pathogenicity of the novel APPL1 variant was preliminarily evaluated through in vitro functional experiments. Finally, the impact of these variants on APPL1 protein expression and the insulin pathway were assessed, and the potential mechanism underlying the interaction between the APPL1 protein and the insulin receptor was further explored. RESULTS A total of five novel mutations were identified, including four missense mutations (Asp632Tyr, Arg633His, Arg532Gln, and Ile642Met) and one intronic mutation (1153-16A>T). Pathogenicity prediction analysis revealed that the Arg532Gln was pathogenic across all predictions. The Asp632Tyr and Arg633His variants also had pathogenicity based on MutationTaster. In addition, multiple alignment of amino acid sequences showed that the Arg532Gln, Asp632Tyr, and Arg633His variants were conserved across different species. Moreover, in in vitro functional experiments, both the c.1894G>T (at Asp632Tyr) and c.1595G>A (at Arg532Gln) mutations were found to downregulate the expression of APPL1 on both protein and mRNA levels, indicating their pathogenic nature. Therefore, based on the patient's clinical and family history, combined with the results from bioinformatics analysis and functional experiment, the c.1894G>T (at Asp632Tyr) and c.1595G>A (at Arg532Gln) mutations were classified as pathogenic mutations. Importantly, all these mutations were located within the phosphotyrosine-binding domain of APPL1, which plays a critical role in the insulin sensitization effect. CONCLUSION This study provided new insights into the pathogenicity of APPL1 gene mutations in diabetes and revealed a potential target for the diagnosis and treatment of the disease.
Collapse
Affiliation(s)
- Ping Shi
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong Province, China
| | - Yang Tian
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong Province, China
| | - Feng Xu
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong Province, China
| | - Lu-Na Liu
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong Province, China
| | - Wan-Hong Wu
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong Province, China
| | - Ying-Zhou Shi
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong Province, China
| | - An-Qi Dai
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong Province, China
| | - Hang-Yu Fang
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong Province, China
| | - Kun-Xia Li
- Department of Pediatric, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai 264099, Shandong Province, China
| | - Chao Xu
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong Province, China
| |
Collapse
|
4
|
Fauzi A, Thoe ES, Quan TY, Yin ACY. Insights from insulin resistance pathways: Therapeutic approaches against Alzheimer associated diabetes mellitus. J Diabetes Complications 2023; 37:108629. [PMID: 37866274 DOI: 10.1016/j.jdiacomp.2023.108629] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 09/03/2023] [Accepted: 10/15/2023] [Indexed: 10/24/2023]
Abstract
Alzheimer Associated Diabetes Mellitus, commonly known as Type 3 Diabetes Mellitus (T3DM) is a distinct subtype of diabetes with a pronounced association with Alzheimer's disease (AD). Insulin resistance serves as a pivotal link between these two conditions, leading to diminished insulin sensitivity, hyperglycemia, and impaired glucose uptake. The brain, a vital organ in AD context, is also significantly impacted by insulin resistance, resulting in energy deficits and neuronal damage, which are hallmark features of the neurodegenerative disorder. To pave the way for potential therapeutic interventions targeting the insulin resistance pathway, it is crucial to comprehend the intricate pathophysiology of T3DM and identify the overlapped features between diabetes and AD. This comprehensive review article aims to explore various pathway such as AMPK, PPARγ, cAMP and P13K/Akt pathway as potential target for management of T3DM. Through the analysis of these complex mechanisms, our goal is to reveal their interdependencies and support the discovery of innovative therapeutic strategies. The review extensively discusses several promising pharmaceutical candidates that have demonstrated dual drug action mechanisms, addressing both peripheral and cerebral insulin resistance observed in T3DM. These candidates hold significant promise for restoring insulin function and mitigating the detrimental effects of insulin resistance on the brain. The exploration of these therapeutic options contributes to the development of innovative interventions that alleviate the burden of T3DM and enhance patient care.
Collapse
Affiliation(s)
- Ayesha Fauzi
- School of Biosciences, Faculty of Health & Medical Sciences, Taylor's University Lakeside Campus, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Ewen Se Thoe
- School of Biosciences, Faculty of Health & Medical Sciences, Taylor's University Lakeside Campus, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Tang Yin Quan
- School of Biosciences, Faculty of Health & Medical Sciences, Taylor's University Lakeside Campus, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia; Medical Advancement for Better Quality of Life Impact Lab, Taylor's University Lakeside Campus, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Adeline Chia Yoke Yin
- School of Biosciences, Faculty of Health & Medical Sciences, Taylor's University Lakeside Campus, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia; Medical Advancement for Better Quality of Life Impact Lab, Taylor's University Lakeside Campus, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
5
|
Wen T, Thapa N, Cryns VL, Anderson RA. Regulation of Phosphoinositide Signaling by Scaffolds at Cytoplasmic Membranes. Biomolecules 2023; 13:1297. [PMID: 37759697 PMCID: PMC10526805 DOI: 10.3390/biom13091297] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
Cytoplasmic phosphoinositides (PI) are critical regulators of the membrane-cytosol interface that control a myriad of cellular functions despite their low abundance among phospholipids. The metabolic cycle that generates different PI species is crucial to their regulatory role, controlling membrane dynamics, vesicular trafficking, signal transduction, and other key cellular events. The synthesis of phosphatidylinositol (3,4,5)-triphosphate (PI3,4,5P3) in the cytoplamic PI3K/Akt pathway is central to the life and death of a cell. This review will focus on the emerging evidence that scaffold proteins regulate the PI3K/Akt pathway in distinct membrane structures in response to diverse stimuli, challenging the belief that the plasma membrane is the predominant site for PI3k/Akt signaling. In addition, we will discuss how PIs regulate the recruitment of specific scaffolding complexes to membrane structures to coordinate vesicle formation, fusion, and reformation during autophagy as well as a novel lysosome repair pathway.
Collapse
Affiliation(s)
- Tianmu Wen
- School of Medicine and Public Health, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI 53705, USA; (T.W.); (N.T.)
| | - Narendra Thapa
- School of Medicine and Public Health, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI 53705, USA; (T.W.); (N.T.)
| | - Vincent L. Cryns
- Department of Medicine, University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI 53705, USA
| | - Richard A. Anderson
- School of Medicine and Public Health, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI 53705, USA; (T.W.); (N.T.)
| |
Collapse
|
6
|
Chen G, Zhang K, Liang Z, Zhang S, Dai Y, Cong Y, Qiao G. Integrated transcriptome analysis identifies APPL1/RPS6KB2/GALK1 as immune-related metastasis factors in breast cancer. Open Med (Wars) 2023; 18:20230732. [PMID: 37273920 PMCID: PMC10238809 DOI: 10.1515/med-2023-0732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/04/2023] [Accepted: 05/14/2023] [Indexed: 06/06/2023] Open
Abstract
The aim of this study is to investigate the prognostic immune-related factors in breast cancer (BC) metastasis. The gene expression chip GSE159956 was downloaded from the gene expression omnibus database. Differentially expressed genes (DEGs) were selected using GEO2R online tools based on lymph node and metastasis status. The intersected survival-associated DEGs were screened from the Kaplan-Meier curve. Gene ontology (GO) and Kyoto Encyclopedia of Gene and Genome (KEGG) annotation analyses were performed to determine the survival-associated DEGs. Immune-related prognostic factors were screened based on immune infiltration. The screened prognostic factors were verified by the Cancer Genome Atlas (TCGA) database and single-sample gene set enrichment analysis (ssGSEA). As a result, twenty-eight upregulated and three downregulated genes were generated by the survival analysis. The enriched GO and KEGG pathways were mostly correlated with "regulation of cellular amino acid metabolic process," "proteasome complex," "endopeptidase activity," and "proteasome." Six of 19 (17 upregulated and 2 downregulated) immune-related prognostic factors were verified by the TCGA database. Four immune-related factors were obtained after ssGSEA, and three significant immune-related factors were selected after univariate and multivariate analyses. Based on the risk score receiver operating characteristic, the three immune-related prognosis factors could be potential biomarkers of BC metastasis. In conclusion, APPL1, RPS6KB2, and GALK1 may play a pivotal role as potential biomarkers for prediction of BC metastasis.
Collapse
Affiliation(s)
- Gang Chen
- Department of Breast Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264001, P.R. China
| | - Kun Zhang
- Department of Breast Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264001, P.R. China
| | - Zhi Liang
- Department of Breast Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264001, P.R. China
| | - Song Zhang
- Department of Breast Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264001, P.R. China
| | - Yuanping Dai
- Department of Medical Genetics, Liuzhou Maternal and Child Health Hospital, Liuzhou545001, P.R. China
| | - Yizi Cong
- Department of Breast Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, No. 20 Yudong Road, Yantai, Shandong 264001, P.R. China
| | - Guangdong Qiao
- Department of Breast Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, No. 20 Yudong Road, Yantai, Shandong 264001, P.R. China
| |
Collapse
|
7
|
Kiełbowski K, Bakinowska E, Ostrowski P, Pala B, Gromowska E, Gurazda K, Dec P, Modrzejewski A, Pawlik A. The Role of Adipokines in the Pathogenesis of Psoriasis. Int J Mol Sci 2023; 24:ijms24076390. [PMID: 37047363 PMCID: PMC10094354 DOI: 10.3390/ijms24076390] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 03/24/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023] Open
Abstract
Psoriasis is a chronic and immune-mediated skin condition characterized by pro-inflammatory cytokines and keratinocyte hyperproliferation. Dendritic cells, T lymphocytes, and keratinocytes represent the main cell subtypes involved in the pathogenesis of psoriasis, while the interleukin-23 (IL-23)/IL-17 pathway enhances the disease progression. Human adipose tissue is an endocrine organ, which secretes multiple proteins, known as adipokines, such as adiponectin, leptin, visfatin, or resistin. Current evidence highlights the immunomodulatory roles of adipokines, which may contribute to the progression or suppression of psoriasis. A better understanding of the complexity of psoriasis pathophysiology linked with adipokines could result in developing novel diagnostic or therapeutic strategies. This review aims to present the pathogenesis of psoriasis and the roles of adipokines in this process.
Collapse
|
8
|
Cabral-Dias R, Antonescu CN. Control of phosphatidylinositol-3-kinase signaling by nanoscale membrane compartmentalization. Bioessays 2023; 45:e2200196. [PMID: 36567275 DOI: 10.1002/bies.202200196] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 09/12/2022] [Accepted: 12/13/2022] [Indexed: 12/27/2022]
Abstract
Phosphatidylinositol-3-kinases (PI3Ks) are lipid kinases that produce 3-phosphorylated derivatives of phosphatidylinositol upon activation by various cues. These 3-phosphorylated lipids bind to various protein effectors to control many cellular functions. Lipid phosphatases such as phosphatase and tensin homolog (PTEN) terminate PI3K-derived signals and are critical to ensure appropriate signaling outcomes. Many lines of evidence indicate that PI3Ks and PTEN, as well as some specific lipid effectors are highly compartmentalized, either in plasma membrane nanodomains or in endosomal compartments. We examine the evidence for specific recruitment of PI3Ks, PTEN, and other related enzymes to membrane nanodomains and endocytic compartments. We then examine the hypothesis that scaffolding of the sources (PI3Ks), terminators (PTEN), and effectors of these lipid signals with a common plasma membrane nanodomain may achieve highly localized lipid signaling and ensure selective activation of specific effectors. This highlights the importance of spatial regulation of PI3K signaling in various physiological and disease contexts.
Collapse
Affiliation(s)
- Rebecca Cabral-Dias
- Department of Chemistry and Biology and Graduate Program in Molecular Science, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Costin N Antonescu
- Department of Chemistry and Biology and Graduate Program in Molecular Science, Toronto Metropolitan University, Toronto, Ontario, Canada
| |
Collapse
|
9
|
Abstract
The global prevalences of obesity and type 2 diabetes mellitus have reached epidemic status, presenting a heavy burden on society. It is therefore essential to find novel mechanisms and targets that could be utilized in potential treatment strategies and, as such, intracellular membrane trafficking has re-emerged as a regulatory tool for controlling metabolic homeostasis. Membrane trafficking is an essential physiological process that is responsible for the sorting and distribution of signalling receptors, membrane transporters and hormones or other ligands between different intracellular compartments and the plasma membrane. Dysregulation of intracellular transport is associated with many human diseases, including cancer, neurodegeneration, immune deficiencies and metabolic diseases, such as type 2 diabetes mellitus and its associated complications. This Review focuses on the latest advances on the role of endosomal membrane trafficking in metabolic physiology and pathology in vivo, highlighting the importance of this research field in targeting metabolic diseases.
Collapse
Affiliation(s)
- Jerome Gilleron
- Université Côte d'Azur, Institut National de la Santé et de la Recherche Médicale (Inserm), UMR1065 C3M, Team Cellular and Molecular Pathophysiology of Obesity, Nice, France.
| | - Anja Zeigerer
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| |
Collapse
|
10
|
Sandoval L, Fuentealba LM, Marzolo MP. Participation of OCRL1, and APPL1, in the expression, proteolysis, phosphorylation and endosomal trafficking of megalin: Implications for Lowe Syndrome. Front Cell Dev Biol 2022; 10:911664. [PMID: 36340038 PMCID: PMC9630597 DOI: 10.3389/fcell.2022.911664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 10/10/2022] [Indexed: 11/13/2022] Open
Abstract
Megalin/LRP2 is the primary multiligand receptor for the re-absorption of low molecular weight proteins in the proximal renal tubule. Its function is significantly dependent on its endosomal trafficking. Megalin recycling from endosomal compartments is altered in an X-linked disease called Lowe Syndrome (LS), caused by mutations in the gene encoding for the phosphatidylinositol 5-phosphatase OCRL1. LS patients show increased low-molecular-weight proteins with reduced levels of megalin ectodomain in the urine and accumulation of the receptor in endosomal compartments of the proximal tubule cells. To gain insight into the deregulation of megalin in the LS condition, we silenced OCRL1 in different cell lines to evaluate megalin expression finding that it is post-transcriptionally regulated. As an indication of megalin proteolysis, we detect the ectodomain of the receptor in the culture media. Remarkably, in OCRL1 silenced cells, megalin ectodomain secretion appeared significantly reduced, according to the observation in the urine of LS patients. Besides, the silencing of APPL1, a Rab5 effector associated with OCRL1 in endocytic vesicles, also reduced the presence of megalin’s ectodomain in the culture media. In both silencing conditions, megalin cell surface levels were significantly decreased. Considering that GSK3ß-mediated megalin phosphorylation reduces receptor recycling, we determined that the endosomal distribution of megalin depends on its phosphorylation status and OCRL1 function. As a physiologic regulator of GSK3ß, we focused on insulin signaling that reduces kinase activity. Accordingly, megalin phosphorylation was significantly reduced by insulin in wild-type cells. Moreover, even though in cells with low activity of OCRL1 the insulin response was reduced, the phosphorylation of megalin was significantly decreased and the receptor at the cell surface increased, suggesting a protective role of insulin in a LS cellular model.
Collapse
Affiliation(s)
- Lisette Sandoval
- Laboratorio de Tráfico Intracelular y Señalización, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Luz M. Fuentealba
- Laboratorio de Tráfico Intracelular y Señalización, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - María-Paz Marzolo
- Laboratorio de Tráfico Intracelular y Señalización, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- *Correspondence: María-Paz Marzolo,
| |
Collapse
|
11
|
A novel partial duplication in OPHN1, associated with vermis cerebellar hypoplasia, seizures and developmental delay. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
12
|
Bao F, Hao P, An S, Yang Y, Liu Y, Hao Q, Ejaz M, Guo XX, Xu TR. Akt scaffold proteins: the key to controlling specificity of Akt signaling. Am J Physiol Cell Physiol 2021; 321:C429-C442. [PMID: 34161152 DOI: 10.1152/ajpcell.00146.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The phosphatidylinositol 3-kinase-Akt signaling pathway plays an essential role in regulating cell proliferation and apoptosis. Akt kinase is at the center of this signaling pathway and interacts with a variety of proteins. Akt is overexpressed in almost 80% of tumors. However, inhibiting Akt has serious clinical side effects so is not a suitable treatment for cancer. During recent years, Akt scaffold proteins have received increasing attention for their ability to regulate Akt signaling and have emerged as potential targets for cancer therapy. In this paper, we categorize Akt kinase scaffold proteins into four groups based on their cellular location: membrane-bound activator and inhibitor, cytoplasm, and endosome. We describe how these scaffolds interact with Akt kinase, how they affect Akt activity, and how they regulate the specificity of Akt signaling. We also discuss the clinical application of Akt scaffold proteins as targets for cancer therapy.
Collapse
Affiliation(s)
- Fan Bao
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China.,Center of Stomatology, The First People's Hospital of Yunnan Province, Kunming, China.,The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Peiqi Hao
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Su An
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Yang Yang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Ying Liu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Qian Hao
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Mubashir Ejaz
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Xiao-Xi Guo
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Tian-Rui Xu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| |
Collapse
|
13
|
Schmidt-Arras D, Rose-John S. Endosomes as Signaling Platforms for IL-6 Family Cytokine Receptors. Front Cell Dev Biol 2021; 9:688314. [PMID: 34141712 PMCID: PMC8204807 DOI: 10.3389/fcell.2021.688314] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 04/28/2021] [Indexed: 12/12/2022] Open
Abstract
Interleukin-6 (IL-6) is the name-giving cytokine of a family of eleven members, including IL-6, CNTF, LIF, and IL-27. IL-6 was first recognized as a B-cell stimulating factor but we now know that the cytokine plays a pivotal role in the orchestration of inflammatory processes as well as in inflammation associated cancer. Moreover, IL-6 is involved in metabolic regulation and it has been shown to be involved in major neural activities such as neuroprotection, which can help to repair and to reduce brain damage. Receptor complexes of all members formed at the plasma membrane contain one or two molecules of the signaling receptor subunit GP130 and the mechanisms of signal transduction are well understood. IL-6 type cytokines can also signal from endomembranes, in particular the endosome, and situations have been reported in which endocytosis of receptor complexes are a prerequisite of intracellular signaling. Moreover, pathogenic GP130 variants were shown to interfere with spatial activation of downstream signals. We here summarize the molecular mechanisms underlying spatial regulation of IL-6 family cytokine signaling and discuss its relevance for pathogenic processes.
Collapse
Affiliation(s)
- Dirk Schmidt-Arras
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| |
Collapse
|
14
|
Tanaka M, Komikawa T, Yanai K, Okochi M. Proteomic Exploration of Membrane Curvature Sensors Using a Series of Spherical Supported Lipid Bilayers. Anal Chem 2020; 92:16197-16203. [DOI: 10.1021/acs.analchem.0c04039] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Masayoshi Tanaka
- Department of Chemical Science and Engineering, Tokyo Institute of Technology, 2-12-1 O-okayama, Meguro-ku, Tokyo 152-8552, Japan
| | - Takumi Komikawa
- Department of Chemical Science and Engineering, Tokyo Institute of Technology, 2-12-1 O-okayama, Meguro-ku, Tokyo 152-8552, Japan
| | - Kentaro Yanai
- Department of Chemical Science and Engineering, Tokyo Institute of Technology, 2-12-1 O-okayama, Meguro-ku, Tokyo 152-8552, Japan
| | - Mina Okochi
- Department of Chemical Science and Engineering, Tokyo Institute of Technology, 2-12-1 O-okayama, Meguro-ku, Tokyo 152-8552, Japan
| |
Collapse
|
15
|
Nader N, Dib M, Hodeify R, Courjaret R, Elmi A, Hammad AS, Dey R, Huang XY, Machaca K. Membrane progesterone receptor induces meiosis in Xenopus oocytes through endocytosis into signaling endosomes and interaction with APPL1 and Akt2. PLoS Biol 2020; 18:e3000901. [PMID: 33137110 PMCID: PMC7660923 DOI: 10.1371/journal.pbio.3000901] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 11/12/2020] [Accepted: 09/18/2020] [Indexed: 12/13/2022] Open
Abstract
The steroid hormone progesterone (P4) mediates many physiological processes through either nuclear receptors that modulate gene expression or membrane P4 receptors (mPRs) that mediate nongenomic signaling. mPR signaling remains poorly understood. Here we show that the topology of mPRβ is similar to adiponectin receptors and opposite to that of G-protein-coupled receptors (GPCRs). Using Xenopus oocyte meiosis as a well-established physiological readout of nongenomic P4 signaling, we demonstrate that mPRβ signaling requires the adaptor protein APPL1 and the kinase Akt2. We further show that P4 induces clathrin-dependent endocytosis of mPRβ into signaling endosome, where mPR interacts transiently with APPL1 and Akt2 to induce meiosis. Our findings outline the early steps involved in mPR signaling and expand the spectrum of mPR signaling through the multitude of pathways involving APPL1. The steroid hormone progesterone mediates many physiological processes through either nuclear receptors that modulate gene expression, or membrane progesterone receptors (mPRs) that mediate non-genomic signaling. This study shows that non-genomic mPRβ signaling progresses through clathrin-dependent endocytosis into signaling endosomes where it interacts with and activates APPL1 and Akt2 to induce oocyte meiosis.
Collapse
Affiliation(s)
- Nancy Nader
- Department of Physiology and Biophysics, Weill Cornell Medicine Qatar, Education City, Qatar Foundation, Doha, Qatar
- Calcium Signaling Group, Weill Cornell Medicine Qatar
| | - Maya Dib
- Department of Physiology and Biophysics, Weill Cornell Medicine Qatar, Education City, Qatar Foundation, Doha, Qatar
- Calcium Signaling Group, Weill Cornell Medicine Qatar
| | - Rawad Hodeify
- Department of Physiology and Biophysics, Weill Cornell Medicine Qatar, Education City, Qatar Foundation, Doha, Qatar
- Calcium Signaling Group, Weill Cornell Medicine Qatar
| | - Raphael Courjaret
- Department of Physiology and Biophysics, Weill Cornell Medicine Qatar, Education City, Qatar Foundation, Doha, Qatar
- Calcium Signaling Group, Weill Cornell Medicine Qatar
| | - Asha Elmi
- Department of Physiology and Biophysics, Weill Cornell Medicine Qatar, Education City, Qatar Foundation, Doha, Qatar
- Calcium Signaling Group, Weill Cornell Medicine Qatar
- College of Health and Life Science, Hamad bin Khalifa University, Doha, Qatar
| | - Ayat S. Hammad
- Department of Physiology and Biophysics, Weill Cornell Medicine Qatar, Education City, Qatar Foundation, Doha, Qatar
- Calcium Signaling Group, Weill Cornell Medicine Qatar
- College of Health and Life Science, Hamad bin Khalifa University, Doha, Qatar
| | - Raja Dey
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, United States of America
| | - Xin-Yun Huang
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, United States of America
| | - Khaled Machaca
- Department of Physiology and Biophysics, Weill Cornell Medicine Qatar, Education City, Qatar Foundation, Doha, Qatar
- Calcium Signaling Group, Weill Cornell Medicine Qatar
- * E-mail:
| |
Collapse
|
16
|
Lee MF, Trotman LC. PTEN: Bridging Endocytosis and Signaling. Cold Spring Harb Perspect Med 2020; 10:cshperspect.a036103. [PMID: 31818848 DOI: 10.1101/cshperspect.a036103] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The transduction of signals in the PTEN/PI3-kinase (PI3K) pathway is built around a phosphoinositide (PIP) lipid messenger, phosphatidylinositol trisphosphate, PI(3,4,5)P3 or PIP3 Another, more ancient role of this family of messengers is the control of endocytosis, where a handful of separate PIPs act like postal codes. Prominent among them is PI(3)P, which helps to ensure that endocytic vesicles, their cargo, and membranes themselves reach their correct destinations. Traditionally, the cancer and the endocytic functions of the PI3K signaling pathway have been studied by cancer and membrane biologists, respectively, with some notable but overall minimal overlap. Modern microscopy has enabled monitoring of the PTEN/PI3K pathway in action. Here, we explore the flurry of groundbreaking concepts emerging from those efforts. The discovery that PTEN contains an autonomous PI(3)P reader domain, fused to the catalytic PIP3 eraser domain has prompted us to explore the relationship between PI3K signaling and endocytosis. This revealed how PTEN can achieve signal termination in a precisely controlled fashion, because endocytosis can package the PIP3 signal into discrete units that PTEN will erase. We explore how PTEN can bridge the worlds of endocytosis and PI3K signaling and discuss progress on how PI3K/AKT signaling can be acting from internal membranes. We discuss how the PTEN/PI3K system for growth control may have emerged from principles of endocytosis, and how this development could have affected the evolution of multicellular organisms.
Collapse
Affiliation(s)
- Matthew F Lee
- Watson School of Biological Sciences, Cold Spring Harbor, New York 11724, USA
| | - Lloyd C Trotman
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| |
Collapse
|
17
|
Schmidt-Arras D, Böhmer FD. Mislocalisation of Activated Receptor Tyrosine Kinases - Challenges for Cancer Therapy. Trends Mol Med 2020; 26:833-847. [PMID: 32593582 DOI: 10.1016/j.molmed.2020.06.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/28/2020] [Accepted: 06/01/2020] [Indexed: 12/20/2022]
Abstract
Activating mutations in genes encoding receptor tyrosine kinases (RTKs) mediate proliferation, cell migration, and cell survival, and are therefore important drivers of oncogenesis. Numerous targeted cancer therapies are directed against activated RTKs, including small compound inhibitors, and immunotherapies. It has recently been discovered that not only certain RTK fusion proteins, but also many full-length RTKs harbouring activating mutations, notably RTKs of the class III family, are to a large extent mislocalised in intracellular membranes. Active kinases in these locations cause aberrant activation of signalling pathways. Moreover, low levels of activated RTKs at the cell surface present an obstacle for immunotherapy. We outline here why understanding of the mechanisms underlying mislocalisation will help in improving existing and developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Dirk Schmidt-Arras
- Christian-Albrechts-University Kiel, Institute of Biochemistry, 24118 Kiel, Germany.
| | - Frank-D Böhmer
- Institute of Molecular Cell Biology, CMB, Jena University Hospital, Jena, Germany
| |
Collapse
|
18
|
Luan X, Yan Y, Zheng Q, Wang M, Chen W, Yu J, Fang J. Excessive reactive oxygen species induce apoptosis via the APPL1-Nrf2/HO-1 antioxidant signalling pathway in trophoblasts with missed abortion. Life Sci 2020; 254:117781. [PMID: 32407842 DOI: 10.1016/j.lfs.2020.117781] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 05/09/2020] [Accepted: 05/10/2020] [Indexed: 02/07/2023]
Abstract
AIMS Previous evidence has demonstrated that oxidative stress is related to the pathogenesis of missed abortion (MA), but the specific mechanism remains obscure. The adaptor protein APPL1 is one of the differential proteins in chorionic trophoblasts. Thus, this study aimed to assess the potential influence of APPL1 on oxidative stress responses as well the possible molecular mechanisms involving in MA. MAIN METHODS In the present study, the chorionic trophoblasts and the HTR-8/SVneo cell line were researched in vitro. Small interfering RNA (siRNA) was used to suppress the expression of APPL1. The fluorescent probes DHE and DCFH-DA were used to assess the intracellular reactive oxidative species (ROS). The activity of superoxide dismutase (SOD) was determined. Apoptosis was detected by TUNEL and flow cytometry. Cell viability was determined using Cell Counting Kit-8. Protein expression was detected by immunohistochemistry, western blotting, and reverse transcription-quantitative PCR. KEY FINDINGS The application of oxidant in normal chorionic trophoblasts induced cell death and overproduction of ROS, which was consistent with MA. In addition, knockdown of APPL1 in HTR-8/SVneo cells resulted in increased ROS and apoptosis, which could be rescued by pretreatment with antioxidants. Mechanistically, we report that overproduction of ROS in trophoblasts and disturbed SOD, APPL1 and Nrf2/HO-1 antioxidant responses constitute important contributors to apoptosis. SIGNIFICANCE Our results suggest that APPL1 has antioxidant properties that suppress oxidative stress and apoptosis via the Nrf2/HO-1 pathway. Moreover, antioxidant N-acetylcysteine (NAC) effectively restored the impaired antioxidative defense system elicited by excess ROS, as a potential therapeutic reagent for MA.
Collapse
Affiliation(s)
- Xiaojin Luan
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - Yidan Yan
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - Qianwen Zheng
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - Min Wang
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - Wanyin Chen
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - Jun Yu
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu 212001, China.
| | - Jie Fang
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu 212001, China.
| |
Collapse
|
19
|
Heng J, Lv P, Zhang Y, Cheng X, Wang L, Ma D, Liu F. Rab5c-mediated endocytic trafficking regulates hematopoietic stem and progenitor cell development via Notch and AKT signaling. PLoS Biol 2020; 18:e3000696. [PMID: 32275659 PMCID: PMC7176290 DOI: 10.1371/journal.pbio.3000696] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 04/22/2020] [Accepted: 03/24/2020] [Indexed: 12/12/2022] Open
Abstract
It is well known that various developmental signals play diverse roles in hematopoietic stem and progenitor cell (HSPC) production; however, how these signaling pathways are orchestrated remains incompletely understood. Here, we report that Rab5c is essential for HSPC specification by endocytic trafficking of Notch and AKT signaling in zebrafish embryos. Rab5c deficiency leads to defects in HSPC production. Mechanistically, Rab5c regulates hemogenic endothelium (HE) specification by endocytic trafficking of Notch ligands and receptor. We further show that the interaction between Rab5c and Appl1 in the endosome is required for the survival of HE in the ventral wall of the dorsal aorta through AKT signaling. Interestingly, Rab5c overactivation can also lead to defects in HSPC production, which is attributed to excessive endolysosomal trafficking inducing Notch signaling defect. Taken together, our findings establish a previously unrecognized role of Rab5c-mediated endocytic trafficking in HSPC development and provide new insights into how spatiotemporal signals are orchestrated to accurately execute cell fate transition. Cell-autonomous Notch signaling regulated by the membrane trafficking protein Rab5c plays an instructive role in hematopoietic stem and progenitor cell specification, while the AKT signaling seems to provide a permissive signal to maintain hemogenic endothelium survival.
Collapse
Affiliation(s)
- Jian Heng
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Peng Lv
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yifan Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xinjie Cheng
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Lu Wang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Dongyuan Ma
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Feng Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- * E-mail:
| |
Collapse
|
20
|
O'Loughlin T, Kendrick-Jones J, Buss F. Approaches to Identify and Characterise MYO6-Cargo Interactions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1239:355-380. [PMID: 32451866 DOI: 10.1007/978-3-030-38062-5_15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Given the prevalence and importance of the actin cytoskeleton and the host of associated myosin motors, it comes as no surprise to find that they are linked to a plethora of cellular functions and pathologies. Although our understanding of the biophysical properties of myosin motors has been aided by the high levels of conservation in their motor domains and the extensive work on myosin in skeletal muscle contraction, our understanding of how the nonmuscle myosins participate in such a wide variety of cellular processes is less clear. It is now well established that the highly variable myosin tails are responsible for targeting these myosins to distinct cellular sites for specific functions, and although a number of adaptor proteins have been identified, our current understanding of the cellular processes involved is rather limited. Furthermore, as more adaptor proteins, cargoes and complexes are identified, the importance of elucidating the regulatory mechanisms involved is essential. Ca2+, and now phosphorylation and ubiquitination, are emerging as important regulators of cargo binding, and it is likely that other post-translational modifications are also involved. In the case of myosin VI (MYO6), a number of immediate binding partners have been identified using traditional approaches such as yeast two-hybrid screens and affinity-based pull-downs. However, these methods have only been successful in identifying the cargo adaptors, but not the cargoes themselves, which may often comprise multi-protein complexes. Furthermore, motor-adaptor-cargo interactions are dynamic by nature and often weak, transient and highly regulated and therefore difficult to capture using traditional affinity-based methods. In this chapter we will discuss the various approaches including functional proteomics that have been used to uncover and characterise novel MYO6-associated proteins and complexes and how this work contributes to a fuller understanding of the targeting and function(s) of this unique myosin motor.
Collapse
Affiliation(s)
- Thomas O'Loughlin
- Cambridge Institute for Medical Research, University of Cambridge, The Keith Peters Building, Cambridge, UK
| | | | - Folma Buss
- Cambridge Institute for Medical Research, University of Cambridge, The Keith Peters Building, Cambridge, UK.
| |
Collapse
|
21
|
Fan L, Ye H, Wan Y, Qin L, Zhu L, Su J, Zhu X, Zhang L, Miao Q, Zhang Q, Zhang Z, Xu A, Li Y, Li X, Wang Y. Adaptor protein APPL1 coordinates HDAC3 to modulate brown adipose tissue thermogenesis in mice. Metabolism 2019; 100:153955. [PMID: 31390528 DOI: 10.1016/j.metabol.2019.153955] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/12/2019] [Accepted: 07/31/2019] [Indexed: 01/17/2023]
Abstract
OBJECTIVES The activation of brown adipose tissue (BAT) is considered as a promising therapeutic target for obesity. APPL1 (Adaptor protein containing the Pleckstrin homology domain, Phosphotyrosine binding domain and Leucine zipper motif) is an intracellular adaptor protein and its genetic variation is correlated with BMI and body fat distribution in diabetic patients. However, little is known about the roles of APPL1 in BAT thermogenesis. MATERIALS/METHODS In this study, adipose tissue specific knockout (ASKO) mice were generated to evaluate APPL1's role in BAT thermogenesis in vivo, and possible signaling pathways were further explored in cultured brown adipocytes. RESULTS After high fat diet challenge, APPL1 ASKO mice developed more severe obesity, glucose intolerance and insulin resistance compared with control mice. Metabolic cage study showed that APPL1 deficiency impaired energy expenditure and adaptive thermogenesis in ASKO mice. PET-CT analysis showed decreased standardized uptake value (SUV) in the inter-scapular region which indicated impaired BAT activity in ASKO mice. Further study showed deletion of APPL1 attenuated brown fat specific gene expression, such as UCP1 and PGC1α in both BAT and brown adipocytes. In cultured brown adipocytes, upon cAMP stimulation, APPL1 shuttled from cytosol to nuclei. Co-IP and ChIP study showed that APPL1 could directly interact with histone deacetylase 3 (HDAC3) to mediate chromatin remodeling and UCP1 gene expression. CONCLUSIONS Our data demonstrated the essential role of APPL1 in regulating brown adipocytes thermogenesis via interaction with HDAC3, which may have potential therapeutic implications for treatment of obesity.
Collapse
Affiliation(s)
- Linling Fan
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Hongying Ye
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Yun Wan
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Lang Qin
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Lu Zhu
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Jing Su
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaoming Zhu
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Lv Zhang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Qing Miao
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiongyue Zhang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhaoyun Zhang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Aimin Xu
- Department of Medicine, the University of Hong Kong, Hong Kong
| | - Yiming Li
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Xi Li
- Institute of Life Sciences, Chongqing Medical University, China.
| | - Yi Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
22
|
Wang HY, Capuano AW, Khan A, Pei Z, Lee KC, Bennett DA, Ahima RS, Arnold SE, Arvanitakis Z. Insulin and adipokine signaling and their cross-regulation in postmortem human brain. Neurobiol Aging 2019; 84:119-130. [PMID: 31539648 DOI: 10.1016/j.neurobiolaging.2019.08.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 08/09/2019] [Accepted: 08/13/2019] [Indexed: 12/14/2022]
Abstract
Aberrant insulin and adipokine signaling has been implicated in cognitive decline associated with both type 2 diabetes mellitus and neurodegenerative diseases. We established methods that reliably measure insulin, adiponectin and leptin signaling, and their crosstalk, in thawed postmortem mid-frontal cortical tissue from cognitively normal older subjects with a short postmortem interval. Insulin-evoked insulin receptor (IR) activation increases activated, tyrosine-phosphorylated IRβ on tyrosine residues 960, 1150, and 1151, insulin receptor substrate-1 recruitment to IRβ and phosphorylated RAC-α-serine/threonine-protein kinase. Adiponectin augments, but leptin inhibits, insulin signaling. Adiponectin activates adiponectin receptors to induce APPL1 binding to adiponectin receptor 1 and 2 and T-cadherin and downstream adenosine monophosphate-dependent protein kinase phosphorylation. Insulin inhibited adiponectin-induced signaling. In addition, leptin-induced leptin receptor (OB-R) signaling promotes Janus kinase 2 recruitment to OB-R and Janus kinase 2 and downstream signal transducer and activator of transcription 3 phosphorylation. Insulin enhanced leptin signaling. These data demonstrate insulin and adipokine signaling interactions in human brain. Future studies can use these methods to examine insulin, adiponectin, and leptin metabolic dysregulation in aging and disease states, such as type 2 diabetes and Alzheimer's disease-related dementias.
Collapse
Affiliation(s)
- Hoau-Yan Wang
- Department of Molecular, Cellular and Biomedical Sciences, City University of New York School of Medicine, New York, NY, USA; Department of Biology, Neuroscience Program, Graduate School of The City University of New York, New York, NY, USA.
| | - Ana W Capuano
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Amber Khan
- Department of Molecular, Cellular and Biomedical Sciences, City University of New York School of Medicine, New York, NY, USA; Department of Biology, Neuroscience Program, Graduate School of The City University of New York, New York, NY, USA
| | - Zhe Pei
- Department of Molecular, Cellular and Biomedical Sciences, City University of New York School of Medicine, New York, NY, USA
| | - Kuo-Chieh Lee
- Department of Molecular, Cellular and Biomedical Sciences, City University of New York School of Medicine, New York, NY, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Rexford S Ahima
- Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Steven E Arnold
- Department of Neurology and the Massachusetts Alzheimer's Disease Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Zoe Arvanitakis
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
23
|
Lysosome Positioning Influences mTORC2 and AKT Signaling. Mol Cell 2019; 75:26-38.e3. [PMID: 31130364 DOI: 10.1016/j.molcel.2019.05.009] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 01/18/2019] [Accepted: 04/16/2019] [Indexed: 12/20/2022]
Abstract
Growth factor signaling is initiated at the plasma membrane and propagated through the cytoplasm for eventual relay to intracellular organelles such as lysosomes. The serine/threonine kinase mTOR participates in growth factor signaling as a component of two multi-subunit complexes, mTORC1 and mTORC2. mTORC1 associates with lysosomes, and its activity depends on the positioning of lysosomes within the cytoplasm, although there is no consensus regarding the exact effect of perinuclear versus peripheral distribution. mTORC2 and its substrate kinase AKT have a widespread distribution, but they are thought to act mainly at the plasma membrane. Using cell lines with knockout of components of the lysosome-positioning machinery, we show that perinuclear clustering of lysosomes delays reactivation of not only mTORC1, but also mTORC2 and AKT upon serum replenishment. These experiments demonstrate the existence of pools of mTORC2 and AKT that are sensitive to lysosome positioning.
Collapse
|
24
|
Sugiyama MG, Fairn GD, Antonescu CN. Akt-ing Up Just About Everywhere: Compartment-Specific Akt Activation and Function in Receptor Tyrosine Kinase Signaling. Front Cell Dev Biol 2019; 7:70. [PMID: 31131274 PMCID: PMC6509475 DOI: 10.3389/fcell.2019.00070] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 04/09/2019] [Indexed: 12/12/2022] Open
Abstract
The serine/threonine kinase Akt is a master regulator of many diverse cellular functions, including survival, growth, metabolism, migration, and differentiation. Receptor tyrosine kinases are critical regulators of Akt, as a result of activation of phosphatidylinositol-3-kinase (PI3K) signaling leading to Akt activation upon receptor stimulation. The signaling axis formed by receptor tyrosine kinases, PI3K and Akt, as well as the vast range of downstream substrates is thus central to control of cell physiology in many different contexts and tissues. This axis must be tightly regulated, as disruption of PI3K-Akt signaling underlies the pathology of many diseases such as cancer and diabetes. This sophisticated regulation of PI3K-Akt signaling is due in part to the spatial and temporal compartmentalization of Akt activation and function, including in specific nanoscale domains of the plasma membrane as well as in specific intracellular membrane compartments. Here, we review the evidence for localized activation of PI3K-Akt signaling by receptor tyrosine kinases in various specific cellular compartments, as well as that of compartment-specific functions of Akt leading to control of several fundamental cellular processes. This spatial and temporal control of Akt activation and function occurs by a large number of parallel molecular mechanisms that are central to regulation of cell physiology.
Collapse
Affiliation(s)
- Michael G. Sugiyama
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON, Canada
| | - Gregory D. Fairn
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Costin N. Antonescu
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON, Canada
| |
Collapse
|
25
|
Lakoduk AM, Roudot P, Mettlen M, Grossman HM, Schmid SL, Chen PH. Mutant p53 amplifies a dynamin-1/APPL1 endosome feedback loop that regulates recycling and migration. J Cell Biol 2019; 218:1928-1942. [PMID: 31043431 PMCID: PMC6548126 DOI: 10.1083/jcb.201810183] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 03/15/2019] [Accepted: 04/12/2019] [Indexed: 12/31/2022] Open
Abstract
Feedback loops arising from crosstalk between early endocytic trafficking and receptor signaling can be co-opted or amplified in cancer cells to enhance their metastatic abilities. Lakoduk et al. reveal that mutant p53 upregulates dynamin-1 expression and recruitment of the APPL1 signaling scaffold to a spatially localized subpopulation of endosomes to increase receptor recycling and cell migration. Multiple mechanisms contribute to cancer cell progression and metastatic activity, including changes in endocytic trafficking and signaling of cell surface receptors downstream of gain-of-function (GOF) mutant p53. We report that dynamin-1 (Dyn1) is up-regulated at both the mRNA and protein levels in a manner dependent on expression of GOF mutant p53. Dyn1 is required for the recruitment and accumulation of the signaling scaffold, APPL1, to a spatially localized subpopulation of endosomes at the cell perimeter. We developed new tools to quantify peripherally localized early endosomes and measure the rapid recycling of integrins. We report that these perimeter APPL1 endosomes modulate Akt signaling and activate Dyn1 to create a positive feedback loop required for rapid recycling of EGFR and β1 integrins, increased focal adhesion turnover, and cell migration. Thus, Dyn1- and Akt-dependent perimeter APPL1 endosomes function as a nexus that integrates signaling and receptor trafficking, which can be co-opted and amplified in mutant p53–driven cancer cells to increase migration and invasion.
Collapse
Affiliation(s)
- Ashley M Lakoduk
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas TX
| | - Philippe Roudot
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas TX
| | - Marcel Mettlen
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas TX
| | - Heather M Grossman
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas TX.,Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas TX
| | - Sandra L Schmid
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas TX
| | - Ping-Hung Chen
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas TX
| |
Collapse
|
26
|
Abstract
Adiponectin is one of the most widely studied adipokines to date. First described in the mid-1990's, studying its regulation, biogenesis and physiological effects has proven to be extremely insightful and improved our understanding of the mechanisms that ensure systemic metabolic homeostasis. Here, we provide a brief overview of the current state of the field with respect to adiponectin, its history, sites and mechanisms of action, and the critical questions that will need to be addressed in the future.
Collapse
Affiliation(s)
- Leon G Straub
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Philipp E Scherer
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
27
|
Goto-Silva L, McShane MP, Salinas S, Kalaidzidis Y, Schiavo G, Zerial M. Retrograde transport of Akt by a neuronal Rab5-APPL1 endosome. Sci Rep 2019; 9:2433. [PMID: 30792402 PMCID: PMC6385319 DOI: 10.1038/s41598-019-38637-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 12/27/2018] [Indexed: 01/19/2023] Open
Abstract
Long-distance axonal trafficking plays a critical role in neuronal function and transport defects have been linked to neurodegenerative disorders. Various lines of evidence suggest that the small GTPase Rab5 plays a role in neuronal signaling via early endosomal transport. Here, we characterized the motility of Rab5 endosomes in primary cultures of mouse hippocampal pyramidal cells by live-cell imaging and showed that they exhibit bi-directional long-range motility in axons, with a strong bias toward retrograde transport. Characterization of key Rab5 effectors revealed that endogenous Rabankyrin-5, Rabenosyn-5 and APPL1 are all present in axons. Further analysis of APPL1-positive endosomes showed that, similar to Rab5-endosomes, they display more frequent long-range retrograde than anterograde movement, with the endosomal levels of APPL1 correlated with faster retrograde movement. Interestingly, APPL1-endosomes transport the neurotrophin receptor TrkB and mediate retrograde axonal transport of the kinase Akt1. FRET analysis revealed that APPL1 and Akt1 interact in an endocytosis-dependent manner. We conclude that Rab5-APPL1 endosomes exhibit the hallmarks of axonal signaling endosomes to transport Akt1 in hippocampal pyramidal cells.
Collapse
Affiliation(s)
- Livia Goto-Silva
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307, Dresden, Germany.,D'Or Institute for Research and Education (IDOR), Rua Diniz Cordeiro, 30, 22281-100, Rio de Janeiro, Brazil
| | - Marisa P McShane
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307, Dresden, Germany
| | - Sara Salinas
- Faculty of Bioengineering and Bioinformatics, Moscow State University, Moscow, Russia.,UMR1058. INSERM/Université de Montpellier/Etablissement Français du Sang Pathogenesis and Control of Chronic Infections, Montpellier, France
| | - Yannis Kalaidzidis
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307, Dresden, Germany.,Faculty of Bioengineering and Bioinformatics, Moscow State University, Moscow, Russia
| | - Giampietro Schiavo
- Molecular NeuroPathobiology Laboratory, Sobell Department of Motor Neuroscience & Movement Disorders, UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Marino Zerial
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307, Dresden, Germany.
| |
Collapse
|
28
|
Abstract
In addition to the common types of diabetes mellitus, two major monogenic diabetes forms exist. Maturity-onset diabetes of the young (MODY) represents a heterogenous group of monogenic, autosomal dominant diseases. MODY accounts for 1-2% of all diabetes cases, and it is not just underdiagnosed but often misdiagnosed to type 1 or type 2 diabetes. More than a dozen MODY genes have been identified to date, and their molecular classification is of great importance in the correct treatment decision and in the judgment of the prognosis. The most prevalent subtypes are HNF1A, GCK, and HNF4A. Genetic testing for MODY has changed recently due to the technological advancements, as contrary to the sequential testing performed in the past, nowadays all MODY genes can be tested simultaneously by next-generation sequencing. The other major group of monogenic diabetes is neonatal diabetes mellitus which can be transient or permanent, and often the diabetes is a part of a syndrome. It is a severe monogenic disease appearing in the first 6 months of life. The hyperglycemia usually requires insulin. There are two forms, permanent neonatal diabetes mellitus (PNDM) and transient neonatal diabetes mellitus (TNDM). In TNDM, the diabetes usually reverts within several months but might relapse later in life. The incidence of NDM is 1:100,000-1:400,000 live births, and PNDM accounts for half of the cases. Most commonly, neonatal diabetes is caused by mutations in KCNJ11 and ABCC8 genes encoding the ATP-dependent potassium channel of the β cell. Neonatal diabetes has experienced a quick and successful transition into the clinical practice since the discovery of the molecular background. In case of both genetic diabetes groups, recent guidelines recommend genetic testing.
Collapse
Affiliation(s)
- Zsolt Gaál
- 4th Department of Medicine, Jósa András Teaching Hospital, Nyíregyháza, Hungary
| | - István Balogh
- Division of Clinical Genetics, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
29
|
Muthiah A, Angulo MS, Walker NN, Keller SR, Lee JK. Biologically anchored knowledge expansion approach uncovers KLF4 as a novel insulin signaling regulator. PLoS One 2018; 13:e0204100. [PMID: 30240435 PMCID: PMC6150497 DOI: 10.1371/journal.pone.0204100] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 09/04/2018] [Indexed: 12/12/2022] Open
Abstract
One of the biggest challenges in analyzing high throughput omics data in biological studies is extracting information that is relevant to specific biological mechanisms of interest while simultaneously restricting the number of false positive findings. Due to random chances with numerous candidate targets and mechanisms, computational approaches often yield a large number of false positives that cannot easily be discerned from relevant biological findings without costly, and often infeasible, biological experiments. We here introduce and apply an integrative bioinformatics approach, Biologically Anchored Knowledge Expansion (BAKE), which uses sequential statistical analysis and literature mining to identify highly relevant network genes and effectively removes false positive findings. Applying BAKE to genomic expression data collected from mouse (Mus musculus) adipocytes during insulin resistance progression, we uncovered the transcription factor Krueppel-like Factor 4 (KLF4) as a regulator of early insulin signaling. We experimentally confirmed that KLF4 controls the expression of two key insulin signaling molecules, the Insulin Receptor Substrate 2 (IRS2) and Tuberous Sclerosis Complex 2 (TSC2).
Collapse
Affiliation(s)
- Annamalai Muthiah
- Department of Systems and Information Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Morgan S. Angulo
- Department of Surgery, University of Virginia Medical Center, University of Virginia, Charlottesville, Virginia, United States of America
| | - Natalie N. Walker
- Department of Medicine, Division of Endocrinology and Metabolism, University of Virginia, Charlottesville, Virginia, United States of America
| | - Susanna R. Keller
- Department of Medicine, Division of Endocrinology and Metabolism, University of Virginia, Charlottesville, Virginia, United States of America
| | - Jae K. Lee
- Department of Systems and Information Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, Florida, United States of America
- Department of Public Health Sciences, University of Virginia School of Medicine, University of Virginia, Charlottesville, Virginia, United States of America
| |
Collapse
|
30
|
Luo R, Chen PW, Kuo JC, Jenkins L, Jian X, Waterman CM, Randazzo PA. ARAP2 inhibits Akt independently of its effects on focal adhesions. Biol Cell 2018; 110:257-270. [PMID: 30144359 DOI: 10.1111/boc.201800044] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 08/17/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND INFORMATION ARAP2, an Arf GTPase-activating protein (Arf GAP) that binds to adaptor protein with PH domain, PTB domain and leucine zipper motifs 1 (APPL1), regulates focal adhesions (FAs). APPL1 affects FA dynamics by regulating Akt. Here, we tested the hypothesis that ARAP2 affects FAs in part by regulating Akt through APPL1. RESULTS We found that ARAP2 controlled FA dynamics dependent on its enzymatic Arf GAP activity. In some cells, ARAP2 also regulated phosphoAkt (pAkt) levels. However, ARAP2 control of FAs did not require Akt and conversely, the effects on pAkt were independent of FAs. Reducing ARAP2 expression reduced the size and number of FAs in U118, HeLa and MDA-MB-231 cells. Decreasing ARAP2 expression increased pAkt in U118 cells and HeLa cells and overexpressing ARAP2 decreased pAkt in U118 cells; in contrast, ARAP2 had no effect on pAkt in MDA-MB-231 cells. An Akt inhibitor did not block the effect of reduced ARAP2 on FAs in U118. Furthermore, the effect of ARAP2 on Akt did not require Arf GAP activity, which is necessary for effects on FAs and integrin traffic. Altering FAs by other means did not induce the same changes in pAkt as those seen by reducing ARAP2 in U118 cells. In addition, we discovered that ARAP2 and APPL1 had co-ordinated effects on pAkt in U118 cells. Reduced APPL1 expression, as for ARAP2, increased pAkt in U118 and the effect of reduced APPL1 expression was reversed by overexpressing ARAP2. Conversely, the effect of reduced ARAP2 expression was reversed by overexpressing APPL1. ARAP2 is an Arf GAP that has previously been reported to affect FAs by regulating Arf6 and integrin trafficking and to bind to the adaptor proteins APPL1. Here, we report that ARAP2 suppresses pAkt levels in cells co-ordinately with APPL1 and independently of GAP activity and its effect on the dynamic behaviour of FAs. CONCLUSIONS We conclude that ARAP2 affects Akt signalling in some cells by a mechanism independent of FAs or membrane traffic. SIGNIFICANCE Our results highlight an Arf GAP-independent function of ARAP2 in regulating Akt activity and distinguish the effect of ARAP2 on Akt from that on FAs and integrin trafficking, which requires regulation of Arf6.
Collapse
Affiliation(s)
- Ruibai Luo
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Pei-Wen Chen
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, Bethesda, MD, 20892, USA.,Department of Biology, Williams College, Williamstown, MA, 01267, USA
| | - Jean-Cheng Kuo
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institutes, Bethesda, MD, 20892, USA.,Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, 112, Taiwan
| | - Lisa Jenkins
- Laboratory of Cell Biology, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Xiaoying Jian
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Clare M Waterman
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institutes, Bethesda, MD, 20892, USA
| | - Paul A Randazzo
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, Bethesda, MD, 20892, USA
| |
Collapse
|
31
|
Ulloa-Aguirre A, Reiter E, Crépieux P. FSH Receptor Signaling: Complexity of Interactions and Signal Diversity. Endocrinology 2018; 159:3020-3035. [PMID: 29982321 DOI: 10.1210/en.2018-00452] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 06/27/2018] [Indexed: 12/20/2022]
Abstract
FSH is synthesized in the pituitary by gonadotrope cells. By binding to and interacting with its cognate receptor [FSH receptor (FSHR)] in the gonads, this gonadotropin plays a key role in the control of gonadal function and reproduction. Upon activation, the FSHR undergoes conformational changes leading to transduction of intracellular signals, including dissociation of G protein complexes into components and activation of several associated interacting partners, which concertedly regulate downstream effectors. The canonical Gs/cAMP/protein kinase A pathway, considered for a long time as the sole effector of FSHR-mediated signaling, is now viewed as one of several mechanisms employed by this receptor to transduce intracellular signals in response to the FSH stimulus. This complex network of signaling pathways allows for a fine-tuning regulation of the gonadotropic stimulus, where activation/inhibition of its multiple components vary depending on the cell context, cell developmental stage, and concentration of associated receptors and corresponding ligands. Activation of these multiple signaling modules eventually converge to the hormone-integrated biological response, including survival, proliferation and differentiation of target cells, synthesis and secretion of paracrine/autocrine regulators, and, at the molecular level, functional selectivity and differential gene expression. In this mini-review, we discuss the complexity of FSHR-mediated intracellular signals activated in response to ligand stimulation. A better understanding of the signaling pathways involved in FSH action might potentially influence the development of new therapeutic strategies for reproductive disorders.
Collapse
Affiliation(s)
- Alfredo Ulloa-Aguirre
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México-Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Eric Reiter
- Biology and Bioinformatics of Signaling Systems Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique, Nouzilly, France
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7247, Nouzilly, France
- Université François Rabelais, Nouzilly, France
| | - Pascale Crépieux
- Biology and Bioinformatics of Signaling Systems Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique, Nouzilly, France
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7247, Nouzilly, France
- Université François Rabelais, Nouzilly, France
| |
Collapse
|
32
|
Kido K, Ato S, Yokokawa T, Sato K, Fujita S. Resistance training recovers attenuated APPL1 expression and improves insulin-induced Akt signal activation in skeletal muscle of type 2 diabetic rats. Am J Physiol Endocrinol Metab 2018; 314:E564-E571. [PMID: 29406784 DOI: 10.1152/ajpendo.00362.2017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Adapter protein containing Pleckstrin homology (PH) domain, phosphotyrosine-binding (PTB) domain, and leucine zipper motif 1 (APPL1) has been reported as a positive regulator of insulin-stimulated Akt activation. The expression of APPL1 is reduced in skeletal muscles of type 2 diabetic (T2D) animals, implying that APPL1 may be an important factor affecting insulin sensitivity. However, the regulation of APPL1 expression and the physiological interventions modulating these effects are unclear. Accordingly, we first confirmed that APPL1 expression and insulin-induced Akt phosphorylation were significantly attenuated in skeletal muscles of T2D rats. Additionally, we found that APPL1 expression levels were significantly correlated with fasting blood glucose levels. Next, we identified important signals involved in the expression of APPL1. APPL1 mRNA expression increased upon AMP-activated protein kinase, calcium, p38 mitogen-activated protein kinase, and insulin-like growth factor-1 signal activation. Moreover, acute resistance exercise in vivo significantly activated these signaling pathways. Finally, through in vivo experiments, we found that chronic resistance training (RT) increased APPL1 expression and activated insulin-induced Akt signaling in skeletal muscles of rats with T2D. Furthermore, variations in APPL1 expression (i.e., the difference between control and RT muscles) significantly correlated with variations in insulin-stimulated Akt phosphorylation under the same conditions. Therefore, chronic RT recovered attenuated APPL1 expression and improved insulin-stimulated Akt phosphorylation in skeletal muscles of T2D rats. Accordingly, APPL1 may be a key regulator of insulin resistance in skeletal muscle, and RT may be an important physiological treatment increasing APPL1 expression, which is attenuated in T2D.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Cells, Cultured
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Down-Regulation/genetics
- Insulin/metabolism
- Insulin/pharmacology
- Insulin Resistance/genetics
- Mice
- Muscle, Skeletal/metabolism
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Physical Conditioning, Animal/physiology
- Proto-Oncogene Proteins c-akt/metabolism
- Rats
- Rats, Inbred OLETF
- Rats, Long-Evans
- Rats, Sprague-Dawley
- Resistance Training
- Signal Transduction/drug effects
- Signal Transduction/physiology
Collapse
Affiliation(s)
- Kohei Kido
- Faculty of Sport and Health Science, Ritsumeikan University , Kusatsu , Japan
| | - Satoru Ato
- Faculty of Sport and Health Science, Ritsumeikan University , Kusatsu , Japan
| | - Takumi Yokokawa
- Laboratory of Sports and Exercise Medicine, Graduate School of Human and Environmental Studies, Kyoto University , Kyoto , Japan
| | - Koji Sato
- Graduate School of Human Development and Environment, Kobe University , Kobe , Japan
| | - Satoshi Fujita
- Faculty of Sport and Health Science, Ritsumeikan University , Kusatsu , Japan
| |
Collapse
|
33
|
Masters TA, Tumbarello DA, Chibalina MV, Buss F. MYO6 Regulates Spatial Organization of Signaling Endosomes Driving AKT Activation and Actin Dynamics. Cell Rep 2018; 19:2088-2101. [PMID: 28591580 PMCID: PMC5469940 DOI: 10.1016/j.celrep.2017.05.048] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 04/05/2017] [Accepted: 05/12/2017] [Indexed: 02/06/2023] Open
Abstract
APPL1- and RAB5-positive signaling endosomes play a crucial role in the activation of AKT in response to extracellular stimuli. Myosin VI (MYO6) and two of its cargo adaptor proteins, GIPC and TOM1/TOM1L2, localize to these peripheral endosomes and mediate endosome association with cortical actin filaments. Loss of MYO6 leads to the displacement of these endosomes from the cell cortex and accumulation in the perinuclear space. Depletion of this myosin not only affects endosome positioning, but also induces actin and lipid remodeling consistent with endosome maturation, including accumulation of F-actin and the endosomal lipid PI(3)P. These processes acutely perturb endosome function, as both AKT phosphorylation and RAC-dependent membrane ruffling were markedly reduced by depletion of either APPL1 or MYO6. These results place MYO6 and its binding partners at a central nexus in cellular signaling linking actin dynamics at the cell surface and endosomal signaling in the cell cortex.
Collapse
Affiliation(s)
- Thomas A Masters
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Building, Hills Road, Cambridge CB2 0XY, UK
| | - David A Tumbarello
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Building, Hills Road, Cambridge CB2 0XY, UK
| | - Margarita V Chibalina
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Building, Hills Road, Cambridge CB2 0XY, UK
| | - Folma Buss
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Building, Hills Road, Cambridge CB2 0XY, UK.
| |
Collapse
|
34
|
Diggins NL, Kang H, Weaver A, Webb DJ. α5β1 integrin trafficking and Rac activation are regulated by APPL1 in a Rab5-dependent manner to inhibit cell migration. J Cell Sci 2018; 131:jcs.207019. [PMID: 29361527 DOI: 10.1242/jcs.207019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 01/09/2018] [Indexed: 01/04/2023] Open
Abstract
Cell migration is a tightly coordinated process that requires the spatiotemporal regulation of many molecular components. Because adaptor proteins can serve as integrators of cellular events, they are being increasingly studied as regulators of cell migration. The adaptor protein containing a pleckstrin-homology (PH) domain, phosphotyrosine binding (PTB) domain, and leucine zipper motif 1 (APPL1) is a 709 amino acid endosomal protein that plays a role in cell proliferation and survival as well as endosomal trafficking and signaling. However, its function in regulating cell migration is poorly understood. Here, we show that APPL1 hinders cell migration by modulating both trafficking and signaling events controlled by Rab5 in cancer cells. APPL1 decreases internalization and increases recycling of α5β1 integrin, leading to higher levels of α5β1 integrin at the cell surface that hinder adhesion dynamics. Furthermore, APPL1 decreases the activity of the GTPase Rac and its effector PAK, which in turn regulate cell migration. Thus, we demonstrate a novel role for the interaction between APPL1 and Rab5 in governing crosstalk between signaling and trafficking pathways on endosomes to affect cancer cell migration.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Nicole L Diggins
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232, USA
| | - Hakmook Kang
- Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Alissa Weaver
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.,Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.,Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Donna J Webb
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232, USA.,Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.,Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
35
|
Abstract
Heart diseases are major causes of mortality. Cardiac hypertrophy, myocardial infarction (MI), viral cardiomyopathy, ischemic and reperfusion (I/R) heart injury finally lead to heart failure and death. Insulin and IGF1 signal pathways play key roles in normal cardiomyocyte growth and physiological cardiac hypertrophy while inflammatory signal pathway is associated with pathological cardiac hypertrophy, MI, viral cardiomyopathy, I/R heart injury, and heart failure. Adapter proteins are the major family proteins, which transduce signals from insulin, IGF1, or cytokine receptors to the downstream pathways and have been shown to regulate variety of heart diseases. Here, we summarized the recent advances in understanding the physiological and pathological roles of adapter proteins in heart failure.
Collapse
Affiliation(s)
- Li Tao
- Cardiovascular Center, 305 Hospital of People's Liberation Army, Beijing, 100017, China
| | - Linna Jia
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, 130024, Jilin, China
| | - Yuntian Li
- Cardiovascular Center, 305 Hospital of People's Liberation Army, Beijing, 100017, China
| | - Chengyun Song
- Cardiovascular Center, 305 Hospital of People's Liberation Army, Beijing, 100017, China.
| | - Zheng Chen
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, 130024, Jilin, China.
| |
Collapse
|
36
|
Galan-Davila AK, Ryu J, Dong K, Xiao Y, Dai Z, Zhang D, Li Z, Dick AM, Liu KD, Kamat A, Lu M, Dong Q, Liu F, Dong LQ. Alternative splicing variant of the scaffold protein APPL1 suppresses hepatic adiponectin signaling and function. J Biol Chem 2018; 293:6064-6074. [PMID: 29483192 DOI: 10.1074/jbc.ra118.002162] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 02/09/2018] [Indexed: 11/06/2022] Open
Abstract
Adiponectin is an adipocyte-derived hormone with antidiabetic activities that include increasing the sensitivity of cells to insulin. Adaptor protein containing pleckstrin homology domain, phosphotyrosine-binding domain, and leucine zipper motif (APPL1) stimulates adiponectin signaling and promotes adiponectin's insulin-sensitizing effects by binding to two adiponectin receptors, AdipoR1 and AdipoR2, and the insulin receptor. In this study, we report an alternative splicing variant of APPL1 (APPL1sv) that is highly expressed in mouse liver, pancreas, and spleen tissues. The expression levels of APPL1sv in liver tissues were enhanced in a mouse model of obesity and diabetic dyslipidemia (i.e. db/db mice) and reduced in calorie-restricted mice compared with ad libitum-fed mice. APPL1sv overexpression or suppression inhibited or enhanced, respectively, adiponectin-stimulated phosphorylation of AMP protein kinase (AMPK) in mouse hepatocytes. We also found that APPL1sv binds to AdipoR1 and AdipoR2 under basal conditions and that adiponectin treatment reduces this binding. Overexpression of APPL1sv blocked adiponectin-induced interactions of APPL1 with the adiponectin receptors. Moreover, adenovirus-mediated and short hairpin RNA-based suppression of APPL1sv greatly reduced high fat diet-induced insulin resistance and hepatic glucose production in mice. Our study identifies a key suppressor of hepatic adiponectin signaling and insulin sensitivity, a finding that may shed light on identifying effective therapeutic targets for treating insulin resistance and type 2 diabetes.
Collapse
Affiliation(s)
- Amanda K Galan-Davila
- From the Departments of Cell Systems and Anatomy.,the Department of Clinical and Applied Science Education, University of the Incarnate Word School of Osteopathic Medicine, San Antonio, Texas 78253
| | - Jiyoon Ryu
- From the Departments of Cell Systems and Anatomy
| | - Kun Dong
- From the Departments of Cell Systems and Anatomy
| | - Yang Xiao
- From the Departments of Cell Systems and Anatomy
| | - Zhe Dai
- From the Departments of Cell Systems and Anatomy
| | - Deling Zhang
- From the Departments of Cell Systems and Anatomy
| | - Zhi Li
- From the Departments of Cell Systems and Anatomy
| | | | - Kevin D Liu
- From the Departments of Cell Systems and Anatomy
| | - Amrita Kamat
- Medicine and.,The Barshop Center for Longevity and Aging Studies, University of Texas Health Science Center, San Antonio, Texas 78229-3900.,the Geriatric Research Education and Clinical Center (GRECC), South Texas Veterans Health Care System, San Antonio, Texas 78229
| | - Min Lu
- the Department of Medicine, University of California, San Diego, La Jolla, California 92093.,the Merck Research Laboratory, Diabetes Early Discovery, Boston, Massachusetts 02115-5727
| | - Qunfeng Dong
- the Center for Biomedical Informatics, Department of Public Health Sciences, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois 60153, and
| | - Feng Liu
- The Barshop Center for Longevity and Aging Studies, University of Texas Health Science Center, San Antonio, Texas 78229-3900.,Pharmacology, and
| | - Lily Q Dong
- From the Departments of Cell Systems and Anatomy, .,The Barshop Center for Longevity and Aging Studies, University of Texas Health Science Center, San Antonio, Texas 78229-3900
| |
Collapse
|
37
|
Liu Y, Zhang C, Zhao L, Du N, Hou N, Song T, Huang C. APPL1 promotes the migration of gastric cancer cells by regulating Akt2 phosphorylation. Int J Oncol 2017; 51:1343-1351. [PMID: 28902365 DOI: 10.3892/ijo.2017.4121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 08/25/2017] [Indexed: 11/06/2022] Open
Abstract
As a multifunctional adaptor protein, APPL1 (adaptor protein containing pleckstrin homology domain, phosphotyrosine binding domain and a leucine zipper motif 1) is overexpressed in many cancers, and has been implicated in tumorigenesis and tumor progression. The present study investigated the expression of APPL1 in gastric carcinoma and the function in regulating cell migration. We investigated the expression of APPL1 in gastric carcinoma based upon The Cancer Genome Atlas (TCGA) database. The expression of APPL1 in collected gastric carcinoma tissues and cultured cells was measured by qRT-PCR and western blot analysis. Transwell assay and wound healing assay were used to analyze the effects of APPL1 on tumor cell migration. The statistical results based upon TCGA database showed significantly higher expression of APPL1 in gastric carcinoma compared to adjacent normal tissues, and we confirmed these findings by measuring APPL1 expression in collected gastric carcinoma tissues and cultured cells. The results of transwell assay and wound healing assay showed that when APPL1 was silenced by siRNA, cell migration was inhibited and overexpression of APPL1 promoted migration. Western blot results demonstrated that changes in several mesenchymal markers were consistent with the observed reduction or enhancement of cell migration. Importantly, the expression of APPL1 significantly affected the phosphorylation of Akt2. In addition, MMP2 and MMP9, downstream effectors of Akt2 changed accordingly, which is a critical requirement for Akt2-mediated cell migration. The results demonstrate an important new function of APPL1 in regulating cell migration through a mechanism that depends on Akt2 phosphorylation.
Collapse
Affiliation(s)
- Yingxun Liu
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Chunli Zhang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Lingyu Zhao
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Ning Du
- Department of Oncology Surgery, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Ni Hou
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Tusheng Song
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Chen Huang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
38
|
APPL1 is a multifunctional endosomal signaling adaptor protein. Biochem Soc Trans 2017; 45:771-779. [PMID: 28620038 DOI: 10.1042/bst20160191] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 03/17/2017] [Accepted: 03/22/2017] [Indexed: 11/17/2022]
Abstract
Endosomal adaptor proteins are important regulators of signaling pathways underlying many biological processes. These adaptors can integrate signals from multiple pathways via localization to specific endosomal compartments, as well as through multiple protein-protein interactions. One such adaptor protein that has been implicated in regulating signaling pathways is the adaptor protein containing a pleckstrin homology (PH) domain, phosphotyrosine-binding (PTB) domain, and leucine zipper motif 1 (APPL1). APPL1 localizes to a subset of Rab5-positive endosomes through its Bin-Amphiphysin-Rvs and PH domains, and it coordinates signaling pathways through its interaction with many signaling receptors and proteins through its PTB domain. This review discusses our current understanding of the role of APPL1 in signaling and trafficking, as well as highlights recent work into the function of APPL1 in cell migration and adhesion.
Collapse
|
39
|
Chen PH, Bendris N, Hsiao YJ, Reis CR, Mettlen M, Chen HY, Yu SL, Schmid SL. Crosstalk between CLCb/Dyn1-Mediated Adaptive Clathrin-Mediated Endocytosis and Epidermal Growth Factor Receptor Signaling Increases Metastasis. Dev Cell 2017; 40:278-288.e5. [PMID: 28171750 PMCID: PMC5740869 DOI: 10.1016/j.devcel.2017.01.007] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 11/15/2016] [Accepted: 01/09/2017] [Indexed: 01/18/2023]
Abstract
Signaling receptors are internalized and regulated by clathrin-mediated endocytosis (CME). Two clathrin light chain isoforms, CLCa and CLCb, are integral components of the endocytic machinery whose differential functions remain unknown. We report that CLCb is specifically upregulated in non-small-cell lung cancer (NSCLC) cells and is associated with poor patient prognosis. Engineered single CLCb-expressing NSCLC cells, as well as "switched" cells that predominantly express CLCb, exhibit increased rates of CME and altered clathrin-coated pit dynamics. This "adaptive CME" resulted from upregulation of dynamin-1 (Dyn1) and its activation through a positive feedback loop involving enhanced epidermal growth factor (EGF)-dependent Akt/GSK3β phosphorylation. CLCb/Dyn1-dependent adaptive CME selectively altered EGF receptor trafficking, enhanced cell migration in vitro, and increased the metastatic efficiency of NSCLC cells in vivo. We define molecular mechanisms for adaptive CME in cancer cells and a role for the reciprocal crosstalk between signaling and CME in cancer progression.
Collapse
Affiliation(s)
- Ping-Hung Chen
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Nawal Bendris
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yi-Jing Hsiao
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei 10617, Taiwan
| | - Carlos R Reis
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Marcel Mettlen
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Hsuan-Yu Chen
- Institute of Statistical Science, Academia Sinica, Taipei 11529, Taiwan
| | - Sung-Liang Yu
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei 10617, Taiwan
| | - Sandra L Schmid
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
40
|
Liu Z, Xiao T, Peng X, Li G, Hu F. APPLs: More than just adiponectin receptor binding proteins. Cell Signal 2017; 32:76-84. [PMID: 28108259 DOI: 10.1016/j.cellsig.2017.01.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 01/13/2017] [Accepted: 01/13/2017] [Indexed: 12/31/2022]
Abstract
APPLs (adaptor proteins containing the pleckstrin homology domain, phosphotyrosine binding domain and leucine zipper motif) are multifunctional adaptor proteins that bind to various membrane receptors, nuclear factors and signaling proteins to regulate many biological activities and processes, such as cell proliferation, chromatin remodeling, endosomal trafficking, cell survival, cell metabolism and apoptosis. APPL1, one of the APPL isoforms, was the first identified protein and interacts directly with adiponectin receptors to mediate adiponectin signaling to enhance lipid oxidation and glucose uptake. APPLs also act on insulin signaling pathways and are important mediators of insulin sensitization. Based on recent findings, this review highlights the critical roles of APPLs, particularly APPL1 and its isoform partner APPL2, in mediating adiponectin, insulin, endosomal trafficking and other signaling pathways. A deep understanding of APPLs and their related signaling pathways may potentially lead to therapeutic and interventional treatments for obesity, diabetes, cancer and neurodegenerative diseases.
Collapse
Affiliation(s)
- Zhuoying Liu
- Department of Metabolism and Endocrinology, Metabolic Syndrome Research Center of Central South University, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Ting Xiao
- Department of Metabolism and Endocrinology, Metabolic Syndrome Research Center of Central South University, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Xiaoyu Peng
- Department of Metabolism and Endocrinology, Metabolic Syndrome Research Center of Central South University, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Guangdi Li
- Department of Metabolism and Endocrinology, Metabolic Syndrome Research Center of Central South University, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Fang Hu
- Department of Metabolism and Endocrinology, Metabolic Syndrome Research Center of Central South University, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
41
|
Song J, Mu Y, Li C, Bergh A, Miaczynska M, Heldin CH, Landström M. APPL proteins promote TGFβ-induced nuclear transport of the TGFβ type I receptor intracellular domain. Oncotarget 2016; 7:279-92. [PMID: 26583432 PMCID: PMC4807998 DOI: 10.18632/oncotarget.6346] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 11/09/2015] [Indexed: 01/05/2023] Open
Abstract
The multifunctional cytokine transforming growth factor-β (TGFβ) is produced by several types of cancers, including prostate cancer, and promote tumour progression in autocrine and paracrine manners. In response to ligand binding, the TGFβ type I receptor (TβRI) activates Smad and non-Smad signalling pathways. The ubiquitin-ligase tumour necrosis factor receptor-associated factor 6 (TRAF6) was recently linked to regulate intramembrane proteolytic cleavage of the TβRI in cancer cells. Subsequently, the intracellular domain (ICD) of TβRI enters in an unknown manner into the nucleus, where it promotes the transcription of pro-invasive genes, such as MMP2 and MMP9. Here we show that the endocytic adaptor molecules APPL1 and APPL2 are required for TGFβ-induced nuclear translocation of TβRI-ICD and for cancer cell invasiveness of human prostate and breast cancer cell lines. Moreover, APPL proteins were found to be expressed at high levels in aggressive prostate cancer tissues, and to be associated with TβRI in a TRAF6-dependent manner. Our results suggest that the APPL–TβRI complex promotes prostate tumour progression, and may serve as a prognostic marker.
Collapse
Affiliation(s)
- Jie Song
- Medical Biosciences, Umeå University, Umeå, Sweden
| | - Yabing Mu
- Medical Biosciences, Umeå University, Umeå, Sweden
| | - Chunyan Li
- Implant Center, Stomatological Hospital, Jilin University, Changchun, China
| | - Anders Bergh
- Medical Biosciences, Umeå University, Umeå, Sweden
| | - Marta Miaczynska
- International Institute of Molecular and Cell Biology, Laboratory of Cell Biology, Warsaw, Poland
| | - Carl-Henrik Heldin
- Ludwig Institute for Cancer Research Ltd, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | | |
Collapse
|
42
|
|
43
|
Chen Z. Adapter proteins regulate insulin resistance and lipid metabolism in obesity. Sci Bull (Beijing) 2016. [DOI: 10.1007/s11434-016-1058-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
44
|
Saleh AJ, Soltani BM, Dokanehiifard S, Medlej A, Tavalaei M, Mowla SJ. Experimental verification of a predicted novel microRNA located in human PIK3CA gene with a potential oncogenic function in colorectal cancer. Tumour Biol 2016; 37:14089-14101. [PMID: 27511117 DOI: 10.1007/s13277-016-5264-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 07/15/2016] [Indexed: 12/21/2022] Open
Abstract
PI3K/AKT signaling is involved in cell survival, proliferation, and migration. In this pathway, PI3Kα enzyme is composed of a regulatory protein encoded by p85 gene and a catalytic protein encoded by PIK3CA gene. Human PIK3CA locus is amplified in several cancers including lung and colorectal cancer (CRC). Therefore, microRNAs (miRNAs) that are encoded within the PIK3CA gene might have a role in cancer development. Here, we report a novel microRNA named PIK3CA-miR1 (EBI accession no. LN626315), which is located within PIK3CA gene. A DNA segment corresponding to PIK3CA-premir1 sequence was transfected in human cell lines that resulted in generation of mature exogenous PIK3CA-miR1. Following the overexpression of PIK3CA-miR1, its predicted target genes (APPL1 and TrkC) were significantly downregulated in the CRC-originated HCT116 and SW480 cell lines, detected by qRT-PCR. Then, dual luciferase assay supported the interaction of PIK3CA-miR1 with APPL1 and TrkC transcripts. Endogenous PIK3CA-miR1 expression was also detected in several cell lines (highly in HCT116 and SW480) and highly in CRC specimens. Consistently, overexpression of PIK3CA-premir1 in HCT116 and SW480 cells resulted in significant reduction of the sub-G1 cell distribution and apoptotic cell rate, as detected by flowcytometry, and resulted in increased cell proliferation, as detected by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. PIK3CA-miR1 overexpression also resulted in Wnt signaling upregulation detected by Top/Fop assay. Overall, accumulative evidences indicated the presence of a bona fide novel onco-miRNA encoded within the PIK3CA oncogene, which is highly expressed in colorectal cancer and has a survival effect in CRC-originated cells.
Collapse
Affiliation(s)
- Ali Jason Saleh
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Bahram M Soltani
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Sadat Dokanehiifard
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Abdallah Medlej
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Seyed Javad Mowla
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
45
|
Wang J, Lu W, Chen L, Zhang P, Qian T, Cao W, Luo J. Serine 707 of APPL1 is Critical for the Synaptic NMDA Receptor-Mediated Akt Phosphorylation Signaling Pathway. Neurosci Bull 2016; 32:323-30. [PMID: 27300007 DOI: 10.1007/s12264-016-0042-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 05/07/2016] [Indexed: 12/31/2022] Open
Abstract
Accumulating evidence indicates that the synaptic activation of N-methyl-D-aspartate receptors (NMDARs) has a neuroprotective effect on neurons. Our previous study demonstrated that APPL1 (adaptor protein containing pleckstrin homology domain, phosphotyrosine-binding domain, and leucine zipper motif) mediates the synaptic activity-dependent activation of PI3K-Akt signaling via coupling this pathway with NMDAR-PSD95 (postsynaptic density protein 95) complexes. However, the molecular mechanism underlying this process is still unknown. In the present study, we investigated the interaction of APPL1 with PSD95 using co-immunocytochemical staining and western blotting. We found that the PDZ2 domain of PSD95 is a binding partner of APPL1. Furthermore, we identified serine 707 of APPL1, a predicted phosphorylation site within the PDZ-binding motif at the C-terminus, as critical for the binding of APPL1 to PSD95, as well as for activation of the Akt signaling pathway during synaptic activity. This suggests that serine 707 of APPL1 is a potential phosphorylation site and may be involved in regulating the neuroprotective Akt signaling pathway that depends on synaptic NMDAR activity.
Collapse
Affiliation(s)
- Jiejie Wang
- Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Wen Lu
- Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Lin Chen
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Ping Zhang
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Tingting Qian
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Wei Cao
- Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Jianhong Luo
- Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| |
Collapse
|
46
|
Naguib A. Following the trail of lipids: Signals initiated by PI3K function at multiple cellular membranes. Sci Signal 2016; 9:re4. [DOI: 10.1126/scisignal.aad7885] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
47
|
Ruan H, Dong LQ. Adiponectin signaling and function in insulin target tissues. J Mol Cell Biol 2016; 8:101-9. [PMID: 26993044 PMCID: PMC4816150 DOI: 10.1093/jmcb/mjw014] [Citation(s) in RCA: 266] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 01/08/2016] [Indexed: 12/13/2022] Open
Abstract
Obesity-linked type 2 diabetes is one of the paramount causes of morbidity and mortality worldwide, posing a major threat on human health, productivity, and quality of life. Despite great progress made towards a better understanding of the molecular basis of diabetes, the available clinical counter-measures against insulin resistance, a defect that is central to obesity-linked type 2 diabetes, remain inadequate. Adiponectin, an abundant adipocyte-secreted factor with a wide-range of biological activities, improves insulin sensitivity in major insulin target tissues, modulates inflammatory responses, and plays a crucial role in the regulation of energy metabolism. However, adiponectin as a promising therapeutic approach has not been thoroughly explored in the context of pharmacological intervention, and extensive efforts are being devoted to gain mechanistic understanding of adiponectin signaling and its regulation, and reveal therapeutic targets. Here, we discuss tissue- and cell-specific functions of adiponectin, with an emphasis on the regulation of adiponectin signaling pathways, and the potential crosstalk between the adiponectin and other signaling pathways involved in metabolic regulation. Understanding better just why and how adiponectin and its downstream effector molecules work will be essential, together with empirical trials, to guide us to therapies that target the root cause(s) of type 2 diabetes and insulin resistance.
Collapse
Affiliation(s)
- Hong Ruan
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Lily Q Dong
- Department of Cell and Structural Biology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| |
Collapse
|
48
|
Imbalanced insulin action in chronic over nutrition: Clinical harm, molecular mechanisms, and a way forward. Atherosclerosis 2016; 247:225-82. [PMID: 26967715 DOI: 10.1016/j.atherosclerosis.2016.02.004] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 12/31/2015] [Accepted: 02/02/2016] [Indexed: 02/08/2023]
Abstract
The growing worldwide prevalence of overnutrition and underexertion threatens the gains that we have made against atherosclerotic cardiovascular disease and other maladies. Chronic overnutrition causes the atherometabolic syndrome, which is a cluster of seemingly unrelated health problems characterized by increased abdominal girth and body-mass index, high fasting and postprandial concentrations of cholesterol- and triglyceride-rich apoB-lipoproteins (C-TRLs), low plasma HDL levels, impaired regulation of plasma glucose concentrations, hypertension, and a significant risk of developing overt type 2 diabetes mellitus (T2DM). In addition, individuals with this syndrome exhibit fatty liver, hypercoagulability, sympathetic overactivity, a gradually rising set-point for body adiposity, a substantially increased risk of atherosclerotic cardiovascular morbidity and mortality, and--crucially--hyperinsulinemia. Many lines of evidence indicate that each component of the atherometabolic syndrome arises, or is worsened by, pathway-selective insulin resistance and responsiveness (SEIRR). Individuals with SEIRR require compensatory hyperinsulinemia to control plasma glucose levels. The result is overdrive of those pathways that remain insulin-responsive, particularly ERK activation and hepatic de-novo lipogenesis (DNL), while carbohydrate regulation deteriorates. The effects are easily summarized: if hyperinsulinemia does something bad in a tissue or organ, that effect remains responsive in the atherometabolic syndrome and T2DM; and if hyperinsulinemia might do something good, that effect becomes resistant. It is a deadly imbalance in insulin action. From the standpoint of human health, it is the worst possible combination of effects. In this review, we discuss the origins of the atherometabolic syndrome in our historically unprecedented environment that only recently has become full of poorly satiating calories and incessant enticements to sit. Data are examined that indicate the magnitude of daily caloric imbalance that causes obesity. We also cover key aspects of healthy, balanced insulin action in liver, endothelium, brain, and elsewhere. Recent insights into the molecular basis and pathophysiologic harm from SEIRR in these organs are discussed. Importantly, a newly discovered oxide transport chain functions as the master regulator of the balance amongst different limbs of the insulin signaling cascade. This oxide transport chain--abbreviated 'NSAPP' after its five major proteins--fails to function properly during chronic overnutrition, resulting in this harmful pattern of SEIRR. We also review the origins of widespread, chronic overnutrition. Despite its apparent complexity, one factor stands out. A sophisticated junk food industry, aided by subsidies from willing governments, has devoted years of careful effort to promote overeating through the creation of a new class of food and drink that is low- or no-cost to the consumer, convenient, savory, calorically dense, yet weakly satiating. It is past time for the rest of us to overcome these foes of good health and solve this man-made epidemic.
Collapse
|
49
|
Kalaidzidis I, Miaczynska M, Brewińska-Olchowik M, Hupalowska A, Ferguson C, Parton RG, Kalaidzidis Y, Zerial M. APPL endosomes are not obligatory endocytic intermediates but act as stable cargo-sorting compartments. J Cell Biol 2016; 211:123-44. [PMID: 26459602 PMCID: PMC4602042 DOI: 10.1083/jcb.201311117] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Endocytosis allows cargo to enter a series of specialized endosomal compartments, beginning with early endosomes harboring Rab5 and its effector EEA1. There are, however, additional structures labeled by the Rab5 effector APPL1 whose role in endocytic transport remains unclear. It has been proposed that APPL1 vesicles are transport intermediates that convert into EEA1 endosomes. Here, we tested this model by analyzing the ultrastructural morphology, kinetics of cargo transport, and stability of the APPL1 compartment over time. We found that APPL1 resides on a tubulo-vesicular compartment that is capable of sorting cargo for recycling or degradation and that displays long lifetimes, all features typical of early endosomes. Fitting mathematical models to experimental data rules out maturation of APPL1 vesicles into EEA1 endosomes as a primary mechanism for cargo transport. Our data suggest instead that APPL1 endosomes represent a distinct population of Rab5-positive sorting endosomes, thus providing important insights into the compartmental organization of the early endocytic pathway.
Collapse
Affiliation(s)
- Inna Kalaidzidis
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Marta Miaczynska
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany International Institute of Molecular and Cell Biology, 02-109 Warsaw, Poland
| | - Marta Brewińska-Olchowik
- International Institute of Molecular and Cell Biology, 02-109 Warsaw, Poland Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland
| | - Anna Hupalowska
- International Institute of Molecular and Cell Biology, 02-109 Warsaw, Poland
| | - Charles Ferguson
- Institute for Molecular Bioscience and Centre for Microscopy and Microanalysis, University of Queensland St. Lucia, Brisbane, Australia 4072
| | - Robert G Parton
- Institute for Molecular Bioscience and Centre for Microscopy and Microanalysis, University of Queensland St. Lucia, Brisbane, Australia 4072
| | - Yannis Kalaidzidis
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Marino Zerial
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| |
Collapse
|
50
|
Ding Y, Cao Y, Wang B, Wang L, Zhang Y, Zhang D, Chen X, Li M, Wang C. APPL1-Mediating Leptin Signaling Contributes to Proliferation and Migration of Cancer Cells. PLoS One 2016; 11:e0166172. [PMID: 27820851 PMCID: PMC5098739 DOI: 10.1371/journal.pone.0166172] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 10/23/2016] [Indexed: 11/24/2022] Open
Abstract
Leptin has been implicated in tumorigenesis and tumor progression, particularly in obese patients. As a multifunctional adaptor protein, APPL1 (containing pleckstrin homology domain, phosphotyrosine binding domain, and a leucine zipper motif 1) plays a critical role in regulating adiponectin and insulin signaling pathways. Currently, high APPL1 level has been suggested to be related to metastases and progression of some types of cancer. However, the intercourse between leptin signaling pathway and APPL1 remains poorly understood. Here, we show that the protein levels and phosphorylation statues of APPL1were highly expressed in tissues from human hepatocellular carcinoma and triple-positive breast cancer. Leptin stimulated APPL1 phosphorylation in a time-dependent manner in both human hepatocellular carcinoma HepG2 cell and breast cancer MCF-7 cell. Overexpression or suppression of APPL1 promoted or attenuated, respectively, leptin-induced phosphorylation of STAT3, ERK1/2, and Akt in the cancer cells, accompanied with enhanced or mitigated cell proliferation and migration. In addition, we identified that APPL1 directly bound to both leptin receptor and STAT3. This interaction was significantly enhanced by leptin stimulation. Our results suggested that APPL1 positively mediated leptin signaling and promoted leptin-induced proliferation and migration of cancer cells. This finding reveals a novel mechanism by which leptin promotes the motility and growth of cancer cells.
Collapse
Affiliation(s)
- Youming Ding
- Department of Hepatobiliary & Laparascopic Surgery, Wuhan University Renmin Hospital, Wuhan, 430060, China
| | - Yingkang Cao
- Department of Pathology & Pathophysiology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Bin Wang
- Department of Hepatobiliary & Laparascopic Surgery, Wuhan University Renmin Hospital, Wuhan, 430060, China
| | - Lei Wang
- Department of Hepatobiliary & Laparascopic Surgery, Wuhan University Renmin Hospital, Wuhan, 430060, China
| | - Yemin Zhang
- Department of Pathology & Pathophysiology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Deling Zhang
- Department of Pathology & Pathophysiology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Xiaoyan Chen
- Department of Hepatobiliary & Laparascopic Surgery, Wuhan University Renmin Hospital, Wuhan, 430060, China
| | - Mingxin Li
- Department of Pathology & Pathophysiology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Changhua Wang
- Department of Pathology & Pathophysiology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
- * E-mail:
| |
Collapse
|