Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Genes (Basel) ; 15(6)2024 May 22.
Article in English | MEDLINE | ID: mdl-38927595

ABSTRACT

Ageing has been identified as an independent risk factor for various diseases; however, the physiological basis and molecular changes related to ageing are still largely unknown. Here, we show that the level of APPL2, an adaptor protein, is significantly reduced in the major organs of aged mice. Knocking down APPL2 causes premature ageing of human umbilical vein endothelial cells (HUVECs). We find that a lack of T04C9.1, the homologue of mammalian APPL2, leads to premature ageing, slow movements, lipid deposition, decreased resistance to stresses, and shortened lifespan in Caenorhabditis elegans (C. elegans), which are associated with decreased autophagy. Activating autophagy by rapamycin or inhibition of let-363 suppresses the age-related alternations, impaired motility, and shortened lifespan of C. elegans, which are reversed by knocking down autophagy-related genes. Our work provides evidence that APPL2 and its C. elegans homologue T04C9.1 decrease with age and reveals that a lack of T04C9.1 bridges autophagy decline and ageing in C. elegans.


Subject(s)
Adaptor Proteins, Signal Transducing , Autophagy , Caenorhabditis elegans Proteins , Caenorhabditis elegans , Longevity , Animals , Humans , Mice , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Aging/genetics , Aging, Premature/genetics , Autophagy/genetics , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Human Umbilical Vein Endothelial Cells , Longevity/genetics
2.
Toxicol Appl Pharmacol ; 484: 116877, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38431228

ABSTRACT

Breast cancer, the most common cancer, presents a significant challenge to the health and longevity of women. Aspongopus chinensis Dallas is an insect with known anti-breast cancer properties. However, the anti-breast cancer effects and underlying mechanisms have not been elucidated. Exogenous microRNAs (miRNAs), which are derived from plants and animals, have been revealed to have notable capacities for controlling the proliferation of cancerous cells. To elucidate the inhibitory effects of miRNAs derived from A. chinensis and the regulatory mechanism involved in the growth of breast cancer cells, miRNA sequencing was initially employed to screen for miRNAs both in A. chinensis hemolymph and decoction and in mouse serum and tumor tissue after decoction gavage. Subsequently, the experiments were performed to assess the suppressive effect of ach-miR-276a-3p, the miRNA screened out from a previous study, on the proliferation of MDA-MB-231 and MDA-MB-468 breast cancer cell lines in vitro and in vivo. Finally, the regulatory mechanism of ach-miR-276a-3p in MDA-MB-231 and MDA-MB-468 breast cancer cells was elucidated. The results demonstrated that ach-miR-276a-3p notably inhibited breast cancer cell proliferation, migration, colony formation, and invasion and induced cell cycle arrest at the G0/G1 phase. Moreover, the ach-miR-276a-3p mimics significantly reduced the tumor volume and weight in xenograft tumor mice. Furthermore, ach-miR-276a-3p could induce cell cycle arrest by targeting APPL2 and regulating the CDK2-Rb-E2F1 signaling pathway. In summary, ach-miR-276a-3p, derived from A. chinensis, has anti-breast cancer activity by targeting APPL2 and regulating the CDK2-Rb-E2F1 signaling pathway and can serve as a promising candidate anticancer agent.


Subject(s)
Breast Neoplasms , MicroRNAs , Humans , Female , Animals , Mice , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Cycle Checkpoints , Signal Transduction , Gene Expression Regulation, Neoplastic , Cyclin-Dependent Kinase 2/genetics , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Adaptor Proteins, Signal Transducing/metabolism
3.
Mol Biol Rep ; 49(5): 3377-3387, 2022 May.
Article in English | MEDLINE | ID: mdl-35397713

ABSTRACT

BACKGROUND: Aberrant activation of the Wnt signaling pathway is observed in most colorectal cancers (CRC). OCC-1D is a splice variant of OCC-1 gene which is considered as a long noncoding RNA (lncRNA) due to lacking the translational initiation codon of the gene. Here, we sought supporting evidence for the effects of OCC-1D on the Wnt pathway and cell cycle progression in CRC. METHODS AND RESULTS: TOP/FOPflash assay and qRT-PCR indicated that expression alterations of OCC-1D could change Wnt signaling activity in colon cancer cells. Consistently, immunocytochemistry results showed the effect of OCC-1D overexpression on nuclear localization of ß-catenin proteins in SW480 cells. Flow cytometry, wound healing and MTT assay confirmed the cell cycle stimulatory effects of OCC-1D in CRC-originated cell lines (SW480 and HCT116). qRT-PCR revealed a positive correlation between the expression level of OCC-1D and its neighboring gene, APPL2. Two distinct tests, downregulation of APPL2 mRNA by using shRNA and Wnt signaling inhibition by using small molecule, along with OCC-1D overexpression confirmed that OCC-1D lncRNA exerts its effect on Wnt signaling pathway through expression modulation of APPL2 gene. CONCLUSIONS: Collectively, we suggested the putative regulatory effects of OCC-1D lncRNA on cell cycle progression and Wnt signaling activation through enhancing the APPL2 gene transcription.


Subject(s)
Colorectal Neoplasms , Neoplasm Proteins/metabolism , RNA, Long Noncoding , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Humans , RNA, Long Noncoding/genetics , Wnt Signaling Pathway , beta Catenin/metabolism
4.
Biochem J ; 2020 Dec 01.
Article in English | MEDLINE | ID: mdl-33258922

ABSTRACT

The APPL (adaptor proteins containing pleckstrin homology domain, phosphotyrosine binding domain and a leucine zipper motif) family consists of two isoforms, APPL1 and APPL2. By binding to curved plasma membrane, these adaptor proteins associate with multiple transmembrane receptors and recruit various downstream signaling components. They are involved in the regulation of signaling pathways evoked by a variety of extracellular stimuli, such as adiponectin, insulin, FSH (follicle stimulating hormone), EGF (epidermal growth factor). And they play important roles in cell proliferation, apoptosis, glucose uptake, insulin secretion and sensitivity. However, emerging evidence suggests that APPL1 and APPL2 perform different or even opposite functions and the underlying mechanism remains unclear. As APPL proteins can either homodimerize or heterodimerize in vivo, we hypothesized that heterodimerization of APPL proteins might account for the mechanism. By solving the crystal structure of APPL1-APPL2 BAR-PH heterodimer, we find that the overall structure is crescent-shaped with a longer curvature radius of 76 Å, compared to 55 Å of the APPL1 BAR-PH homodimer. However, there is no significant difference of the curvature between APPL BAR-PH heterodimer and APPL2 homodimer. The data suggest that the APPL1 BAR-PH homodimer, APPL2 BAR-PH homodimer and APPL1/APPL2 BAR-PH heterodimer may bind to endosomes of different sizes.   Different positive charge distribution is observed on the concave surface of APPL BAR-PH heterodimer than the homodimers, which may change the affinity of membrane association and subcellular localization. Collectively, APPL2 may regulate APPL1 function through altering the preference of endosome binding by heterodimerization.

5.
Proc Natl Acad Sci U S A ; 117(45): 28307-28315, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33122440

ABSTRACT

Filamentous actin (F-actin) cytoskeletal remodeling is critical for glucose-stimulated insulin secretion (GSIS) in pancreatic ß-cells, and its dysregulation causes type 2 diabetes. The adaptor protein APPL1 promotes first-phase GSIS by up-regulating soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) protein expression. However, whether APPL2 (a close homology of APPL1 with the same domain organization) plays a role in ß-cell functions is unknown. Here, we show that APPL2 enhances GSIS by promoting F-actin remodeling via the small GTPase Rac1 in pancreatic ß-cells. ß-cell specific abrogation of APPL2 impaired GSIS, leading to glucose intolerance in mice. APPL2 deficiency largely abolished glucose-induced first- and second-phase insulin secretion in pancreatic islets. Real-time live-cell imaging and phalloidin staining revealed that APPL2 deficiency abolished glucose-induced F-actin depolymerization in pancreatic islets. Likewise, knockdown of APPL2 expression impaired glucose-stimulated F-actin depolymerization and subsequent insulin secretion in INS-1E cells, which were attributable to the impairment of Ras-related C3 botulinum toxin substrate 1 (Rac1) activation. Treatment with the F-actin depolymerization chemical compounds or overexpression of gelsolin (a F-actin remodeling protein) rescued APPL2 deficiency-induced defective GSIS. In addition, APPL2 interacted with Rac GTPase activating protein 1 (RacGAP1) in a glucose-dependent manner via the bin/amphiphysin/rvs-pleckstrin homology (BAR-PH) domain of APPL2 in INS-1E cells and HEK293 cells. Concomitant knockdown of RacGAP1 expression reverted APPL2 deficiency-induced defective GSIS, F-actin remodeling, and Rac1 activation in INS-1E cells. Our data indicate that APPL2 interacts with RacGAP1 and suppresses its negative action on Rac1 activity and F-actin depolymerization thereby enhancing GSIS in pancreatic ß-cells.


Subject(s)
Actins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/pharmacology , Glucose/metabolism , Insulin Secretion/drug effects , Insulin-Secreting Cells/metabolism , Actin Cytoskeleton/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Diabetes Mellitus, Type 2/metabolism , Female , GTPase-Activating Proteins/metabolism , Gene Knockdown Techniques , Glucose Intolerance , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , Neuropeptides/genetics , Neuropeptides/metabolism , SNARE Proteins/metabolism , Transcriptome , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
6.
Neurosci Bull ; 36(9): 997-1008, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32468397

ABSTRACT

Adult olfactory neurogenesis plays critical roles in maintaining olfactory functions. Newly-generated neurons in the subventricular zone migrate to the olfactory bulb (OB) and determine olfactory discrimination, but the mechanisms underlying the regulation of olfactory neurogenesis remain unclear. Our previous study indicated the potential of APPL2 (adaptor protein, phosphotyrosine interacting with PH domain and leucine zipper 2) as a modulating factor for neurogenesis in the adult olfactory system. In the present study, we report how APPL2 affects neurogenesis in the OB and thereby mediates olfactory discrimination by using both in vitro neural stem cells (NSCs) and an in vivo animal model-APPL2 transgenic (Tg) mice. In the in vitro study, we found that APPL2 overexpression resulted in NSCs switching from neuronal differentiation to gliogenesis while APPL2 knockdown promoted neurogenesis. In the in vivo study, APPL2 Tg mice had a higher population of glial cells and dampened neuronal production in the olfactory system, including the corpus callosum, OB, and rostral migratory stream. Adult APPL2 Tg mice displayed impaired performance in olfactory discrimination tests compared with wild-type mice. Furthermore, we found that an interaction of APPL2 with Notch1 contributed to the roles of APPL2 in modulating the neurogenic lineage-switching and olfactory behaviors. In conclusion, APPL2 controls olfactory discrimination by switching the fate choice of NSCs via interaction with Notch1 signaling.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Neural Stem Cells/cytology , Olfactory Bulb/cytology , Receptor, Notch1/physiology , Smell , Animals , Cell Differentiation , Lateral Ventricles , Mice , Mice, Transgenic , Neurogenesis , Primary Cell Culture , Rats , Rats, Sprague-Dawley
7.
Neuroscience Bulletin ; (6): 997-1008, 2020.
Article in English | WPRIM (Western Pacific) | ID: wpr-826757

ABSTRACT

Adult olfactory neurogenesis plays critical roles in maintaining olfactory functions. Newly-generated neurons in the subventricular zone migrate to the olfactory bulb (OB) and determine olfactory discrimination, but the mechanisms underlying the regulation of olfactory neurogenesis remain unclear. Our previous study indicated the potential of APPL2 (adaptor protein, phosphotyrosine interacting with PH domain and leucine zipper 2) as a modulating factor for neurogenesis in the adult olfactory system. In the present study, we report how APPL2 affects neurogenesis in the OB and thereby mediates olfactory discrimination by using both in vitro neural stem cells (NSCs) and an in vivo animal model-APPL2 transgenic (Tg) mice. In the in vitro study, we found that APPL2 overexpression resulted in NSCs switching from neuronal differentiation to gliogenesis while APPL2 knockdown promoted neurogenesis. In the in vivo study, APPL2 Tg mice had a higher population of glial cells and dampened neuronal production in the olfactory system, including the corpus callosum, OB, and rostral migratory stream. Adult APPL2 Tg mice displayed impaired performance in olfactory discrimination tests compared with wild-type mice. Furthermore, we found that an interaction of APPL2 with Notch1 contributed to the roles of APPL2 in modulating the neurogenic lineage-switching and olfactory behaviors. In conclusion, APPL2 controls olfactory discrimination by switching the fate choice of NSCs via interaction with Notch1 signaling.

8.
Neuroscience Bulletin ; (6): 997-1008, 2020.
Article in English | WPRIM (Western Pacific) | ID: wpr-828345

ABSTRACT

Adult olfactory neurogenesis plays critical roles in maintaining olfactory functions. Newly-generated neurons in the subventricular zone migrate to the olfactory bulb (OB) and determine olfactory discrimination, but the mechanisms underlying the regulation of olfactory neurogenesis remain unclear. Our previous study indicated the potential of APPL2 (adaptor protein, phosphotyrosine interacting with PH domain and leucine zipper 2) as a modulating factor for neurogenesis in the adult olfactory system. In the present study, we report how APPL2 affects neurogenesis in the OB and thereby mediates olfactory discrimination by using both in vitro neural stem cells (NSCs) and an in vivo animal model-APPL2 transgenic (Tg) mice. In the in vitro study, we found that APPL2 overexpression resulted in NSCs switching from neuronal differentiation to gliogenesis while APPL2 knockdown promoted neurogenesis. In the in vivo study, APPL2 Tg mice had a higher population of glial cells and dampened neuronal production in the olfactory system, including the corpus callosum, OB, and rostral migratory stream. Adult APPL2 Tg mice displayed impaired performance in olfactory discrimination tests compared with wild-type mice. Furthermore, we found that an interaction of APPL2 with Notch1 contributed to the roles of APPL2 in modulating the neurogenic lineage-switching and olfactory behaviors. In conclusion, APPL2 controls olfactory discrimination by switching the fate choice of NSCs via interaction with Notch1 signaling.

9.
Mol Biol Rep ; 45(6): 1889-1895, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30218350

ABSTRACT

Breast cancer is the most common cancer type and the second cause of cancer death in women. Different mechanisms are contributed to the initiation and progression of the breast cancer. OCC-1 and APPL2 neighboring genes located in 12q.23.3 human chromosome region are related to colorectal cancer. Here, we intended to investigate OCC-1 newly reported transcript variants and APPL2 gene expression alteration in breast cancer specimens and investigate OCC-1 variants overexpression effect on APPL2 and on cell cycle status. Rt-qPCR analysis indicated that the expression level of OCC-1A/B and OCC-1D (not OCC-1C) transcript variants has been increased while, APPL2 gene expression level has been decreased in breast cancer specimen, compared to their normal pairs. Therefore, a negative correlation of expression is evident between APPL2 and OCC-1 genes in breast cancer specimen. Unlike OCC-1A/B which encodes a small protein, OCC-1D noncoding RNA overexpression lead to APPL2 downregulation in MCF7 cells. Consistently, OCC-1D overexpression resulted in increased sub-G1 cell population in MCF7 cells, detected by flow cytometry. Altogether, these results suggest that OCC1-D variant have an inhibitory effect on APPL2 expression and may regulate the cell cycle status.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Breast Neoplasms/genetics , Neoplasm Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle/genetics , Cell Line, Tumor , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Neoplasm Proteins/metabolism , Transcriptional Activation
10.
Life Sci ; 210: 209-213, 2018 Oct 01.
Article in English | MEDLINE | ID: mdl-30189216

ABSTRACT

AIMS: The aim of this study was to evaluate the effects of aging on intracellular adiponectin signaling and the possible therapeutic effect of physical exercise. MAIN METHODS: Fischer 344 rats were distributed in the following groups: Young (3 months old); Sedentary Old (Old, 27 months old); and Old Exercised (Old-Exe, 27 months old), which were subjected to a short-term exercise training protocol. KEY FINDINGS: The results showed that the old rats presented glucose intolerance without increased adiposity. However, short-term exercise training reversed this disorder, which was associated with a decrease in the pleckstrin homology domain, phosphotyrosine-binding domain, and leucine zipper motif (APPL) isoform 2 (APPL2) content. The APPL isoform 1 (APPL1) and TRB3 (Tribbles homolog 3) contents were not altered. Akt phosphorylation was only increased in the old exercised rats. There was a reduction in the content of adiponectin receptor 1 in the old rats. SIGNIFICANCE: The short-term exercise training protocol was able to decrease APPL2 content in the skeletal muscle, which was accompanied by an improvement in the glucose tolerance of the old Fischer 344 rats. These findings provide new evidence supporting the role of physical exercise as a non-pharmacological therapeutic intervention to attenuate age-related deficits.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Aging , Glucose Intolerance/therapy , Muscle, Skeletal/metabolism , Nerve Tissue Proteins/metabolism , Physical Conditioning, Animal , Animals , Glucose Intolerance/physiopathology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Inbred F344
11.
Mol Neurobiol ; 55(12): 9334-9348, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29675572

ABSTRACT

Olfactory dysfunction is often accompanied with anxiety- and depressive-like behaviors in depressive patients. Impaired neurogenesis in hippocampus and subventricular zone (SVZ)-olfactory bulb (OB) contribute to anxiety- and depressive-like behaviors and olfactory dysfunctions. However, the underlying mechanisms of olfactory dysfunction remain unclear. Our previous study indicates that adaptor protein, phosphotyrosine interacting with PH domain and leucine zipper 2 (APPL2), could affect the activity and sensitivity of glucocorticoid receptor (GR) and mediate impaired hippocampal neurogenesis, which contribute the development of depression. In the present study, we further identified the roles of APPL2 in olfactory functions. APPL2 Tg mice displayed higher GR activity and less capacity of neurogenesis at olfactory system with less olfactory sensitivity than WT mice, indicating that APPL2 could be a potential therapeutic target for depression and olfactory deficits. We then studied the effects of baicalin, a medicinal herbal compound, on modulating APPL2/GR signaling pathway for promoting neurogenesis and antidepressant as well as improving olfactory functions. Baicalin treatment inhibited APPL2/GR signaling pathway and improved neurogenesis at SVZ, OB, and hippocampus in APPL2 Tg mice and chronic corticosterone-induced depression mouse model. Behavioral tests revealed that baicalin attenuated depressive- and anxiety-like behaviors and improve olfactory functions in the chronic depression mouse model and APPL2 Tg mice. Taken together, APPL2 could be a novel therapeutic target for improving depressant-related olfactory dysfunctions and baicalin could inhibit APPL2-mediated GR hyperactivity and promote adult neurogenesis, subsequently releasing depressive and anxiety symptoms and improving olfactory functions for antidepressant therapy.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Depression/drug therapy , Emotions , Flavonoids/therapeutic use , Neurogenesis , Olfactory Bulb/physiopathology , Receptors, Glucocorticoid/metabolism , Signal Transduction , Animals , Anxiety/drug therapy , Anxiety/pathology , Anxiety/physiopathology , Behavior, Animal , Brain/drug effects , Brain/metabolism , Brain/pathology , Cell Line, Tumor , Corticosterone/adverse effects , Depression/chemically induced , Depression/physiopathology , Disease Models, Animal , Flavonoids/pharmacology , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Neurogenesis/drug effects , Olfactory Bulb/drug effects , Olfactory Bulb/pathology , Signal Transduction/drug effects
12.
Mol Neurobiol ; 55(7): 5537-5547, 2018 Jul.
Article in English | MEDLINE | ID: mdl-28965332

ABSTRACT

Adaptor proteins containing the pleckstrin homology domain, phosphotyrosine binding domain, and leucine zipper motif (APPLs) are multifunctional adaptor proteins involved in regulating many biological activities and processes. The newly identified metabolic factor APPL2 showed the potentials to modulate cell growth, but whether APPL2 could affect adult neurogenesis and animal mood behaviors remains unknown. In the present study, APPL2 transgenic (Tg) mice and wild-type littermates were used for testing our hypothesis that APPL2 could affect glucocorticoid receptor (GR) signaling and modulate hippocampal neurogenesis, which contributes to depressive and anxiety behaviors. Compared with WT littermates, APPL2 Tg mice had enhanced GR phosphorylation under basic condition but had no different plasma corticosterone (CORT) level and GR phosphorylation under stress stimulation. APPL2 Tg mice had decreased hippocampal neurogenesis that was reversed by GR antagonist RU486. APPL2 Tg mice also showed the impaired hippocampal neurogenesis and presented the depressive and anxiety behaviors. In conclusion, APPL2 could be an important regulator for adult neurogenesis. APPL2 overexpression could blunt the activation of glucocorticoid receptor when undergoing environmental stress. Our study suggests that APPL2 might be a new therapeutic target for mental disorders.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Aging/metabolism , Antidepressive Agents/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Neurogenesis , Receptors, Glucocorticoid/metabolism , Animals , Anxiety/complications , Anxiety/metabolism , Behavior, Animal , Cell Differentiation , Cell Lineage , Cell Proliferation , Depression/complications , Depression/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Neurons/metabolism , Phosphorylation , Retinal Ganglion Cells/metabolism , Signal Transduction , Stress, Physiological
13.
Adv Exp Med Biol ; 960: 415-441, 2017.
Article in English | MEDLINE | ID: mdl-28585210

ABSTRACT

The decrease in adiponectin levels are negatively correlated with chronic subclinical inflammation markers in obesity. The hypertrophic adipocytes cause obesity-linked insulin resistance and metabolic syndrome. Furthermore, macrophage polarization is a key determinant regulating adiponectin receptor (AdipoR1/R2) expression and differential adiponectin-mediated macrophage inflammatory responses in obese individuals. In addition to decrease in adiponectin concentrations, the decline in AdipoR1/R2 mRNA expression leads to a decrement in adiponectin binding to cell membrane, and this turns into attenuation in the adiponectin effects. Within the receptor complex, adaptor protein-containing pleckstrin homology domain, phosphotyrosine-binding domain, and leucine zipper motif 1 (APPL1) is the intracellular binding partner of AdipoR1 and AdipoR2. The expression levels of APPL1 or APPL2 lead to an altered adiponectin activity. Despite normal or high adiponectin levels, an impaired post receptor signaling due to APPL1/APPL2 may alter adiponectin efficiency and activity. However, APPL2 blocks adiponectin signaling through AdipoR1 and AdipoR2 by competitive inhibition of APPL1. APPL1 is also an important mediator of adiponectin dependent insulin sensitization. In this context, adiponectin resistance is associated with insulin resistance and is thought to be partly due to the down-regulation of the AdipoRs in high-fat diet fed subjects. Actually, adiponectin resistance occurs very rapidly after saturated fatty acid feeding, this metabolic disturbance is not due to a decrease in AdipoR1 protein content. Intra-abdominal adipose tissue-AdipoR2 expression is reduced in obesity, whereas AdipoR1 expression is not changed. Adiponectin resistance together with insulin resistance forms a vicious cycle. The elevated adiponectin levels with adiponectin resistance is a compensatory response in the condition of an unusual discordance between insulin resistance and adiponectin unresponsiveness.Additionally, different mechanisms are involved in vascular adiponectin resistance at different stages of obesity. Nevertheless, diet-induced hyperlipidemia is the leading cause of vascular adiponectin resistance. Leptin/adiponectin imbalance may also be an important marker of the elevated risk of developing abdominal obesity-associated cardiovascular diseases.


Subject(s)
Adiponectin/metabolism , Obesity/metabolism , Obesity/pathology , Animals , Humans , Inflammation/metabolism , Inflammation/pathology , Insulin Resistance/physiology , Metabolic Syndrome/metabolism , Metabolic Syndrome/pathology , Receptors, Adiponectin/metabolism
14.
Cell Signal ; 32: 76-84, 2017 04.
Article in English | MEDLINE | ID: mdl-28108259

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.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Receptors, Adiponectin/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adiponectin/metabolism , Animals , Endosomes/metabolism , Humans , Insulin/metabolism , Protein Binding
15.
RNA ; 23(1): 70-85, 2017 01.
Article in English | MEDLINE | ID: mdl-27986894

ABSTRACT

The Wnt signaling pathway is hyperactivated in most colorectal cancers (CRC). Finding new regulators of this pathway represents the potential for cancer diagnosis or treatment. OCC-1 was initially reported as an up-regulated gene in colon carcinoma, without knowing its mechanism of action. Here, two novel transcript variants and an exonic microRNA that originated from the OCC-1 gene are reported, showing positive effects on Wnt activity. Up-regulation of the known OCC-1 variant (assigned as OCC-1A/B) was limited to CRC, and its overexpression increased survival of CRC-originated SW480 cells (Wnt+), while resulting in apoptosis of Wnt-suppressed SW480 cells or HeLa cells (Wnt-) detected by PI staining. Immunocytochemistry showed that the OCC-1A/B-encoded peptide was localized to the nucleus, where its overexpression resulted in Wnt signaling up-regulation, detected by TOP/FOPflash assay. The noncoding portion of the OCC-1A/B transcript had a suppressive effect on Wnt activity and had a negative correlation with APPL2 neighboring gene expression. Unlike OCC-1A/B, the novel OCC-1C splice variant had no expression alteration in CRC, and it seemed to encode a smaller peptide with cytoplasmic localization. A 60-nucleotide (nt) fragment containing an AUG start codon is spliced out to produce an OCC-1D noncoding RNA variant. The 60-nt RNA was validated as the precursor of a novel microRNA, which we named miR-ex1 Both OCC-1D and miR-ex1 were coordinately up-regulated in CRC. MiR-ex1 functional analysis revealed that it is targeting the APC2 tumor suppressor gene and is an activator of the Wnt signaling pathway. Overall, the OCC-1 gene is now introduced as a novel Wnt signaling regulator and as a potential therapeutic target.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Alternative Splicing , Colorectal Neoplasms/genetics , MicroRNAs/genetics , Neoplasm Proteins/genetics , Proteins/genetics , Apoptosis , Cell Line, Tumor , Cell Survival , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Organ Specificity , Up-Regulation , Wnt Signaling Pathway
16.
J Mol Cell Biol ; 8(2): 101-9, 2016 04.
Article in English | MEDLINE | ID: mdl-26993044

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.


Subject(s)
Adiponectin/metabolism , Insulin/metabolism , Organ Specificity , Animals , Humans , Models, Biological , Receptors, Adiponectin/metabolism , Signal Transduction
17.
Cell Biosci ; 4(1): 60, 2014.
Article in English | MEDLINE | ID: mdl-25328665

ABSTRACT

BACKGROUND: The adapter proteins Appl1 (adaptor protein containing pleckstrin homology domain, phosphotyrosine domain, and leucine zipper motif 1) and Appl2 are highly homologous and involved in several signaling pathways. While previous studies have shown that Appl1 plays a pivotal role in adiponectin signaling and insulin secretion, the physiological functions of Appl2 are largely unknown. RESULTS: In the present study, the role of Appl2 in sepsis shock was investigated by using Appl2 knockout (KO) mice. When challenged with lipopolysaccharides (LPS), Appl2 KO mice exhibited more severe symptoms of endotoxin shock, accompanied by increased production of proinflammatory cytokines. In comparison with the wild-type control, deletion of Appl2 led to higher levels of TNF-α and IL-1ß in primary macrophages. In addition, phosphorylation of Akt and its downstream effector NF-κB was significantly enhanced. By co-immunoprecipitation, we found that Appl2 and Appl1 interacted with each other and formed a complex with PI3K regulatory subunit p85α, which is an upstream regulator of Akt. Consistent with these results, deletion of Appl1 in macrophages exhibited characteristics of reduced Akt activation and decreased the production of TNFα and IL-1ß when challenged by LPS. CONCLUSIONS: Results of the present study demonstrated that Appl2 is a critical negative regulator of innate immune response via inhibition of PI3K/Akt/NF-κB signaling pathway by forming a complex with Appl1 and PI3K.

SELECTION OF CITATIONS
SEARCH DETAIL
...