Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters








Publication year range
1.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(7): 1355-1360, 2024 Jul 20.
Article in Chinese | MEDLINE | ID: mdl-39051081

ABSTRACT

OBJECTIVE: To explore the effects of deletion of protein 4.1R on hepatocyte proliferation, apoptosis, and glycolysis and the molecular mechanisms. METHODS: A 4.1R-/- HL-7702 cell line was constructed using CRISPR/Cas9 technique, and with 4.1R+/+HL-7702 cells as the control, its proliferative capacity and cell apoptosis were assessed using CCK-8 assay, EdU-488 staining, flow cytometry and Annexin V-FITC/PI staining at 24, 48, 72 h of cell culture. The changes in glucose uptake, lactate secretion, ATP production and pH value of the culture supernatant of 4.1R-/- HL-7702 cells were determined. The mRNA expressions of the key regulatory enzymes HK2, PFKL, PKM2 and LDHA in glycolysis were detected with qRT-PCR, and the protein expressions of AMPK, p-AMPK, Raptor and p-Raptor were determined using Western blotting. RESULTS: Western blotting and sequencing analysis both confirmed the successful construction of 4.1R-/- HL-7702 cell line. Compared with the wild-type cells, 4.1R-/- HL-7702 cells exhibited a lowered proliferative activity with increased cell apoptosis. The deletion of protein 4.1R also resulted in significantly decreased glucose uptake, lactate secretion and ATP production of the cells and increased pH value of the cell culture supernatant. qRT-PCR showed significantly decreased mRNA expressions of the key regulatory enzymes in glycolysis in 4.1R-/- HL-7702 cells. Compared with those in HL-7702 cells, the expression levels of AMPK and Raptor proteins were decreased while the expression levels of p-AMPK and p-Raptor proteins increased significantly in 4.1R-/- HL-7702 cells. CONCLUSION: Deletion of protein 4.1R in HL-7702 cells results in reduced proliferative capacity, increased apoptosis and suppression of glycolysis, and this regulatory mechanism is closely related with the activation of the downstream AMPK-mTORC1 signaling pathway.


Subject(s)
Apoptosis , Cell Proliferation , Glycolysis , Hepatocytes , Humans , Hepatocytes/metabolism , Hepatocytes/cytology , Cell Line , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/genetics , CRISPR-Cas Systems , Glucose/metabolism , Signal Transduction
2.
Biomolecules ; 14(2)2024 Feb 11.
Article in English | MEDLINE | ID: mdl-38397451

ABSTRACT

The protein 4.1R is an essential component of the erythrocyte membrane skeleton, serving as a key structural element and contributing to the regulation of the membrane's physical properties, including mechanical stability and deformability, through its interaction with spectrin-actin. Recent research has uncovered additional roles of 4.1R beyond its function as a linker between the plasma membrane and the membrane skeleton. It has been found to play a crucial role in various biological processes, such as cell fate determination, cell cycle regulation, cell proliferation, and cell motility. Additionally, 4.1R has been implicated in cancer, with numerous studies demonstrating its potential as a diagnostic and prognostic biomarker for tumors. In this review, we provide an updated overview of the gene and protein structure of 4.1R, as well as its cellular functions in both physiological and pathological contexts.


Subject(s)
Cytoskeletal Proteins , Membrane Proteins , Membrane Proteins/metabolism , Cytoskeletal Proteins/metabolism , Spectrin/chemistry , Spectrin/genetics , Spectrin/metabolism , Actins/metabolism , Erythrocyte Membrane/metabolism
3.
Int Immunopharmacol ; 128: 111546, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38237224

ABSTRACT

Acute liver injury (ALI) is a common clinical disease caused by sepsis, metabolic syndrome, hepatitis virus. Macrophage plays an important role in the development of ALI, which is characterized by polarization and inflammatory regulation. The polarization process of macrophages is related to membrane binding proteins and adaptors. Protein 4.1R acts as an adaptor, linking membrane proteins to the cytoskeleton, and is involved in cell activation and cytokine secretion. However, whether protein 4.1R is involved in regulating macrophage polarization and inflammation-induced liver injury remains unknown. In this study, protein 4.1R is identified with the special effect on macrophage M1 polarization. And it is further demonstrated that protein 4.1R deficiency significantly enhance glycolytic metabolism. Mechanistically, the regulation of protein 4.1R on pyruvate kinase M2 (PKM2) plays a key role in glycolysis metabolism. In addition, we found that protein 4.1R directly interacts with toll-like receptor 4 (TLR4), inhibits the activation of the AKT/HIF-1α signaling pathway. In conclusion, protein 4.1R targets HIF-1α mediated glycolysis regulates M1 macrophage polarization, indicating that protein 4.1R is a candidate for regulating macrophage mediated inflammatory response. In conclusion, we have revealed a novel function of protein 4.1R in macrophage polarization and ALI, providing important insights into the metabolic reprogramming, which is important for ALI therapy. We have revealed a novel function of protein 4.1R in macrophage polarization and ALI, providing important insights into the metabolic reprogramming, which is important for ALI therapy.


Subject(s)
Chemical and Drug Induced Liver Injury, Chronic , Sepsis , Mice , Animals , Chemical and Drug Induced Liver Injury, Chronic/metabolism , Lipopolysaccharides/pharmacology , Macrophages , Glycolysis , Sepsis/metabolism
4.
J Biol Chem ; 299(2): 102877, 2023 02.
Article in English | MEDLINE | ID: mdl-36621628

ABSTRACT

The red blood cells (RBCs) of vertebrates have evolved into two basic shapes, with nucleated nonmammalian RBCs having a biconvex ellipsoidal shape and anuclear mammalian RBCs having a biconcave disk shape. In contrast, camelid RBCs are flat ellipsoids with reduced membrane deformability, suggesting altered membrane skeletal organization. However, the mechanisms responsible for their elliptocytic shape and reduced deformability have not been determined. We here showed that in alpaca RBCs, protein 4.1R, a major component of the membrane skeleton, contains an alternatively spliced exon 14-derived cassette (e14) not observed in the highly conserved 80 kDa 4.1R of other highly deformable biconcave mammalian RBCs. The inclusion of this exon, along with the preceding unordered proline- and glutamic acid-rich peptide (PE), results in a larger and unique 90 kDa camelid 4.1R. Human 4.1R containing e14 and PE, but not PE alone, showed markedly increased ability to form a spectrin-actin-4.1R ternary complex in viscosity assays. A similar facilitated ternary complex was formed by human 4.1R possessing a duplication of the spectrin-actin-binding domain, one of the mutations known to cause human hereditary elliptocytosis. The e14- and PE-containing mutant also exhibited an increased binding affinity to ß-spectrin compared with WT 4.1R. Taken together, these findings indicate that 4.1R protein with the e14 cassette results in the formation and maintenance of a hyperstable membrane skeleton, resulting in rigid red ellipsoidal cells in camelid species, and suggest that membrane structure is evolutionarily regulated by alternative splicing of exons in the 4.1R gene.


Subject(s)
Alternative Splicing , Camelids, New World , Cell Shape , Cytoskeletal Proteins , Erythrocytes , Animals , Humans , Actins/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Erythrocytes/cytology , Erythrocytes/metabolism , Membranes/metabolism , Peptides/metabolism , Protein Binding , Spectrin/genetics , Spectrin/metabolism , Cell Shape/genetics
5.
Exp Cell Res ; 409(1): 112896, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34717920

ABSTRACT

M2 macrophages are crucial components of the tumour microenvironment and have been shown to be closely related to tumour progression. Co-culture with 4.1R-/- M2 macrophages enhances the malignancy of colon cancer (CC), but the mechanism remains unclear. Here, we report that protein 4.1R knockout reduced the phagocytosis of M2 macrophages (M-CSF/IL-4-treated bone marrow cells) and promoted MC38 colon cancer cell proliferation, migration, invasion, tumour formation and epithelial-mesenchymal transition (EMT), which are regulated by M2 macrophages. Further mechanistic dissection revealed that the 4.1R knockout upregulated vascular endothelial growth factor A (VEGFA) secreted by M2 macrophages and promoted colon cancer progression by activating the PI3K/AKT signalling pathway. In summary, our present study identified that 4.1R downregulates VEGFA secretion in M2 macrophages and delays the malignant potential of colon cancer by inhibiting the PI3K/AKT signalling pathway.


Subject(s)
Colonic Neoplasms/genetics , Down-Regulation/genetics , Macrophages/physiology , Microfilament Proteins/genetics , Vascular Endothelial Growth Factor A/genetics , Animals , Cell Line , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Colonic Neoplasms/pathology , Epithelial-Mesenchymal Transition/genetics , Female , Macrophage Activation , Macrophage Colony-Stimulating Factor/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Signal Transduction/genetics , Tumor Microenvironment/genetics
6.
Virus Res ; 306: 198593, 2021 12.
Article in English | MEDLINE | ID: mdl-34637814

ABSTRACT

Zika virus (ZIKV) is a typical mosquito-borne flavivirus known to cause severe fetal microcephaly and adult Guillain-Barré syndrome. Currently, there are no specific drugs or licensed vaccines available for ZIKV infection, and further research is required to identify host cell proteins involved in the virus's life cycle. Viruses are known to use host cell membrane skeletal proteins, such as actin and spectrin, to complete cell entry, transportation, and release. Here, based on immunoprecipitation, the Axl and ZIKV envelope (E) protein were shown to interact with the cell membrane skeleton protein 4.1R. Furthermore, deletion of 4.1R significantly reduced virus titer and viral protein synthesis. Our study showed that 4.1R is an important host cell protein during ZIKV infection and may be involved in the process of viral entry into host cells.


Subject(s)
Zika Virus Infection , Zika Virus , Animals , Cell Membrane/metabolism , Cytoskeletal Proteins/metabolism , Membrane Proteins/metabolism , Virus Internalization , Virus Replication , Zika Virus/metabolism
7.
J Biol Chem ; 297(3): 101051, 2021 09.
Article in English | MEDLINE | ID: mdl-34364872

ABSTRACT

The asymmetric cell division of stem or progenitor cells generates daughter cells with distinct fates that balance proliferation and differentiation. Asymmetric segregation of Notch signaling regulatory protein Numb plays a crucial role in cell diversification. However, the molecular mechanism remains unclear. Here, we examined the unequal distribution of Numb in the daughter cells of murine erythroleukemia cells (MELCs) that undergo DMSO-induced erythroid differentiation. In contrast to the cytoplasmic localization of Numb during uninduced cell division, Numb is concentrated at the cell boundary in interphase, near the one-spindle pole in metaphase, and is unequally distributed to one daughter cell in anaphase in induced cells. The inheritance of Numb guides this daughter cell toward erythroid differentiation while the other cell remains a progenitor cell. Mitotic spindle orientation, critical for distribution of cell fate determinants, requires complex communication between the spindle microtubules and the cell cortex mediated by the NuMA-LGN-dynein/dynactin complex. Depletion of each individual member of the complex randomizes the position of Numb relative to the mitotic spindle. Gene replacement confirms that multifunctional erythrocyte protein 4.1R (4.1R) functions as a member of the NuMA-LGN-dynein/dynactin complex and is necessary for regulating spindle orientation, in which interaction between 4.1R and NuMA plays an important role. These results suggest that mispositioning of Numb is the result of spindle misorientation. Finally, disruption of the 4.1R-NuMA-LGN complex increases Notch signaling and decreases the erythroblast population. Together, our results identify a critical role for 4.1R in regulating the asymmetric segregation of Numb to mediate erythropoiesis.


Subject(s)
Asymmetric Cell Division , Erythroid Cells/cytology , Erythroid Cells/metabolism , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation , Cell Line, Tumor , Dynactin Complex/genetics , Dynactin Complex/metabolism , Dyneins/genetics , Dyneins/metabolism , Membrane Proteins/genetics , Mice , Microfilament Proteins/genetics , Mitosis , Nerve Tissue Proteins/genetics , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Spindle Apparatus/genetics , Spindle Apparatus/metabolism
8.
Acta Physiol (Oxf) ; 232(3): e13647, 2021 07.
Article in English | MEDLINE | ID: mdl-33729672

ABSTRACT

AIMS: Total haemoglobin mass (tot-Hb) increases during high-altitude acclimatization. Normalization of tot-Hb upon descent is thought to occur via neocytolysis, the selective destruction of newly formed erythrocytes. Because convincing experimental proof of neocytolysis is lacking, we performed a prospective study on erythrocyte survival after a stay at the Jungfraujoch Research Station (JFJRS; 3450 m). METHODS: Newly formed erythrocytes of 12 male subjects (mean age 23.3 years) were age cohort labelled in normoxia (110 m) and during a 19-day high-altitude sojourn by ingestion of 13 C2- and 15 N-labelled glycine respectively. Elimination dynamics for erythrocytes produced in normoxia and at high altitude were measured by isotope ratio mass spectrometry of haem, by determining tot-Hb, reticulocyte counts, erythrocyte membrane protein 4.1a/4.1b ratio and by mathematical modelling. RESULTS: Tot-Hb increased by 4.7% ± 2.7% at high altitude and returned to pre-altitude values within 11 days after descent. Elimination of 13 C- (normoxia) and 15 N- (high altitude) labelled erythrocytes was not different. Erythropoietin levels and counts of CD71-positive reticulocytes decreased rapidly after descent. The band 4.1a/4.1b ratio decreased at altitude and remained low for 3-4 days after descent and normalized slowly. There was no indication of haemolysis. CONCLUSION: We confirm a rapid normalization of tot-Hb upon descent. Based on the lack of accelerated removal of age cohorts of erythrocytes labelled at high altitude, on patterns of changes in reticulocyte counts and of the band 4.1a/4.1b ratio and on modelling, this decrease did not occur via neocytolysis, but by a reduced rate of erythropoiesis along with normal clearance of senescent erythrocytes.


Subject(s)
Altitude , Erythropoietin , Adult , Erythrocytes , Humans , Male , Prospective Studies , Reticulocytes , Young Adult
9.
Exp Cell Res ; 399(2): 112465, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33385415

ABSTRACT

Melanoma is the most aggressive malignant tumor of skin cancer as it can grow rapidly and metastasize. Photodynamic therapy (PDT) is a promising cancer ablation method for skin tumors, although it lacks efficiency owing to factors such as tumor characteristics, delivery of photosensitizers, immune response in vivo etc. Extensive investigation of molecules that can potentially modulate treatment efficacy is required. Protein 4.1R is a cytoskeletal protein molecule. Previous studies have shown that protein 4.1R knockdown reduces PDT sensitivity in mouse embryonic fibroblast cells. However, the functional role of protein 4.1R in melanoma is unclear. In this study, we aimed to elucidate the effect of protein 4.1R on PDT for melanoma in mice and the mechanism of anti-tumor immunity. Our results indicated that CRISPR/Cas9-mediated protein 4.1R knockout promotes the proliferation, migration, and invasion of B16 cells. We further investigated the potential mechanism of protein 4.1R on tumor cell PDT sensitivity. Our results showed that protein 4.1R knockout reduced the expression of membrane transporters γ-aminobutyric acid transporter (GAT)-1 and (GAT)-2 in B16 cells, which affected 5-ALA transmembrane transport and reduced the efficiency of PDT on B16 cells. Protein 4.1R knockout downregulated the anti-tumor immune response triggered by PDT in vivo. In conclusion, our data suggest that protein 4.1R is an important regulator in PDT for tumors and may promote the progress and efficacy of melanoma treatment.


Subject(s)
Cytoskeletal Proteins/physiology , Levulinic Acids/metabolism , Melanoma, Experimental/drug therapy , Membrane Proteins/physiology , Skin Neoplasms/drug therapy , Animals , Biological Transport/drug effects , Biological Transport/genetics , Cell Line, Tumor , Cytoskeletal Proteins/genetics , Gene Knockout Techniques , HEK293 Cells , Humans , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Photochemotherapy/methods , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Aminolevulinic Acid
10.
Exp Cell Res ; 398(2): 112403, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33271128

ABSTRACT

The proliferation of mast cells (MCs) plays a crucial role in either physiological or pathological progression of human physical. C-Kit-mediated signaling pathway has been confirmed to play a key role in MCs proliferation, and the regulatory mechanisms of C-Kit-mediated MCs proliferation need to be further explored. Our previous study found that protein 4.1R could negatively regulate T cell receptor (TCR) mediated signal pathways in CD4+ T cells. Little is known about the function of 4.1R in C-Kit-mediated proliferation of MCs. In this study, P815-4.1R-/- cells were constructed by using CRISPR/Cas9 technique. Lack of 4.1R significantly enhanced P815 cells proliferation by accelerating the progression of cell cycle. 4.1R could also significantly alleviate the clinical symptoms of systemic mastocytosis (SM) and improve the overall survival of SM mice. Further study showed that 4.1R could interact directly with C-Kit to inhibit the activation of C-Kit-mediated Ras-Raf-MAPKs and PI3K-AKT signal pathways. Taken together, our findings demonstrate that protein 4.1R, a novel negative regulator, negatively regulates MCs proliferation by inhibiting C-Kit-mediated signal transduction, which maybe provide a potential target to the prevention and treatment of abnormal MCs proliferation-related diseases.


Subject(s)
Cytoskeletal Proteins/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Signal Transduction , Animals , Cell Proliferation , Cells, Cultured , Humans , Mice, Inbred DBA
11.
Biochem Biophys Res Commun ; 534: 172-178, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33298314

ABSTRACT

Cell adhesion molecule 1 (CADM1), which mediates intercellular adhesion between epithelial cells, is shown to be highly expressed in small-cell lung cancer (SCLC) and to enhance tumorigenicity of SCLC cells in nude mice. Here, we investigated the molecular mechanism underlying the oncogenic role of CADM1 in SCLC. CADM1 promoted colony formation of SCLC cells in soft agar. Analysis of deletion and point mutants of the conserved protein-binding motifs in CADM1 revealed that the 4.1 protein-binding motif in the cytoplasmic domain is responsible for the promotion of colony formation. Among the actin-binding 4.1 proteins, 4.1R was the only protein whose localization to the plasma membrane is dependent on CADM1 expression in SCLC cells. Knockdown of 4.1R suppressed the colony formation enhanced by CADM1, suggesting that 4.1R is required for the oncogenic role of CADM1 in SCLC. In primary SCLC, CADM1 expression was correlated with membranous localization of 4.1R, as was observed in a SCLC cell line. Moreover, membranous co-localization of CADM1 and 4.1R was associated with more advanced tumor stage. These results suggest that the formation of CADM1-4.1R complex would promote malignant features of SCLC.


Subject(s)
Cell Adhesion Molecule-1/metabolism , Cytoskeletal Proteins/metabolism , Lung Neoplasms/metabolism , Membrane Proteins/metabolism , Small Cell Lung Carcinoma/metabolism , Animals , Cell Adhesion Molecule-1/genetics , Cell Line, Tumor , Cell Membrane/metabolism , Female , Lung Neoplasms/pathology , Mice, Inbred BALB C , Mice, Nude , Mutation , Small Cell Lung Carcinoma/pathology
12.
Immunology ; 161(4): 314-324, 2020 12.
Article in English | MEDLINE | ID: mdl-32852059

ABSTRACT

During the immune response, B cells can enter the memory pathway, which is characterized by class switch recombination (CSR), or they may undergo plasma cell differentiation (PCD) to secrete immunoglobulin. Both of these processes occur in activated B cells, which are reported to relate to membrane-association proteins and adaptors. Protein 4.1R acts as an adaptor, linking membrane proteins to the cytoskeleton, and is involved in many cell events such as cell activation and differentiation, and cytokine secretion. However, the effect of 4.1R on regulating B-cell fate is unclear. Here, we show an important association between B-cell fate and 4.1R. In vitro, primary B cells were stimulated with lipopolysaccharide combined with interleukin-4; results showed that 4.1R-deficient (4.1R-/- ) cells compared with wild-type (4.1R+/+ ) B cells augmented expression of activation-induced cytidine deaminase and germline, resulting in increased IgG1+ B cells, whereas the secretion of IgG1 and IgM was reduced, and CD138+ B cells were also decreased. Throughout the process, 4.1R regulated canonical nuclear factor (NF-κB) rather than non-canonical NF-κB to promote the expression of CSR complex components, leading to up-regulation of B-cell CSR. In contrast, 4.1R-deficient B cells showed reduced expression of Blimp-1, which caused B cells to down-regulate PCD. Furthermore, over-activation of canonical NF-κB may induce apoptosis signaling to cause PCD apoptosis to reduce PCD number. In summary, our results suggest that 4.1R acts as a B-cell fate regulator by inhibiting the canonical NF-κB signaling pathway.


Subject(s)
B-Lymphocytes/immunology , Cytoskeleton/metabolism , Microfilament Proteins/metabolism , NF-kappa B/metabolism , Animals , Cell Differentiation , Cells, Cultured , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Immunoglobulin Class Switching , Immunoglobulin G/metabolism , Immunologic Memory , Immunomodulation , Interleukin-4/metabolism , Lipopolysaccharides/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/genetics , Signal Transduction
13.
Hematology ; 25(1): 321-326, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32807033

ABSTRACT

Objectives: Hereditary elliptocytosis (HE) is inherited in an autosomal dominant fashion, and the majority of HE-associated defects occur due to qualitative and quantitative defects in the RBC membrane skeleton proteins α-spectrin, ß-spectrin, or protein 4.1R. The complex EPB41 gene encodes a diverse family of protein 4.1R isoforms which are key components of the erythroid membrane skeleton that regulates red cell morphology and mechanical stability. The purpose of this study was to investigate the genome of a Korean patient with HE to discover the causative gene mutation using gene panel sequencing. Methods: An 89-year-old female presented to the Emergency Department and was diagnosed with pancreatitis and gallstones. A peripheral blood smear revealed that approximately 60% of the RBCs were abnormally shaped and appeared oval or elongated, from slightly egg-shaped to rod or pencil forms (elliptocytes). Targeted gene panel sequencing consisting of 33 genes related to inherited RBC disorders and Sanger sequencing were performed. Results: A heterozygous c.2112G > A of the EPB41 gene leading to premature termination codon (NM_001166005.1:c.2112G > A, p.Trp704*) was identified. This variant, which had not been previously reported to be related to HE, was confirmed by Sanger sequencing. Thus, the patient's diagnosis of HE-1 was genetically confirmed. Conclusion: The present study confirmed a novel mutation of the EPB41 gene that plays an important role in expanding the mutational distribution in HE-1. It could also be helpful for understanding the correlation between the genotype and phenotype in HE.


Subject(s)
Cytoskeletal Proteins/genetics , Elliptocytosis, Hereditary/genetics , Membrane Proteins/genetics , Point Mutation , Aged, 80 and over , Elliptocytosis, Hereditary/complications , Elliptocytosis, Hereditary/pathology , Erythrocytes, Abnormal/metabolism , Erythrocytes, Abnormal/pathology , Female , Humans , Republic of Korea
14.
J Biol Chem ; 295(1): 191-211, 2020 01 03.
Article in English | MEDLINE | ID: mdl-31776189

ABSTRACT

Epithelial adherens junctions (AJs) and tight junctions (TJs) undergo disassembly and reassembly during morphogenesis and pathological states. The membrane-cytoskeleton interface plays a crucial role in junctional reorganization. Protein 4.1R (4.1R), expressed as a diverse array of spliceoforms, has been implicated in linking the AJ and TJ complex to the cytoskeleton. However, which specific 4.1 isoform(s) participate and the mechanisms involved in junctional stability or remodeling remain unclear. We now describe a role for epithelial-specific isoforms containing exon 17b and excluding exon 16 4.1R (4.1R+17b) in AJs. 4.1R+17b is exclusively co-localized with the AJs. 4.1R+17b binds to the armadillo repeats 1-2 of ß-catenin via its membrane-binding domain. This complex is linked to the actin cytoskeleton via a bispecific interaction with an exon 17b-encoded peptide. Exon 17b peptides also promote fodrin-actin complex formation. Expression of 4.1R+17b forms does not disrupt the junctional cytoskeleton and AJs during the steady-state or calcium-dependent AJ reassembly. Overexpression of 4.1R-17b forms, which displace the endogenous 4.1R+17b forms at the AJs, as well as depletion of the 4.1R+17b forms both decrease junctional actin and attenuate the recruitment of spectrin to the AJs and also reduce E-cadherin during the initial junctional formation of the AJ reassembly process. Expressing 4.1R+17b forms in depleted cells rescues junctional localization of actin, spectrin, and E-cadherin assembly at the AJs. Together, our results identify a critical role for 4.1R+17b forms in AJ assembly and offer additional insights into the spectrin-actin-4.1R-based membrane skeleton as an emerging regulator of epithelial integrity and remodeling.


Subject(s)
Adherens Junctions/metabolism , Cytoskeletal Proteins/metabolism , Membrane Proteins/metabolism , Actins/metabolism , Alternative Splicing , Animals , Binding Sites , Cadherins/metabolism , Calcium/metabolism , Carrier Proteins/metabolism , Cytoskeletal Proteins/genetics , Dogs , Humans , Madin Darby Canine Kidney Cells , Membrane Proteins/genetics , Microfilament Proteins/metabolism , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Spectrin/metabolism , beta Catenin/chemistry , beta Catenin/metabolism
15.
Exp Cell Res ; 384(2): 111648, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31562860

ABSTRACT

The correct functioning of epidermal growth factor receptor (EGFR), a receptor tyrosine kinase, is required for normal skin development and homeostasis. Cellular hyperproliferation induced by dysregulation of EGFR is tightly associated with structural and functional defects of hair follicles, skin lesions, and tumorigenesis. However, a number of questions still remain regarding the mechanism of EGFR activation and signaling. Here, we report that 4.1R, a member of the membrane-cytoskeleton linker FERM family proteins, plays critical roles in EGFR activation and signaling in keratinocytes. We demonstrated that knockout of 4.1R augments the excessive proliferation potential of keratinocytes by immunohistochemical analysis using murine skin samples. 4.1R-/- keratinocytes display enhanced EGFR-mediated Akt/ERK signaling by upregulating EGFR expression and phosphorylation, which can be reversed by either EGFR or MEK phosphorylation inhibitors. Mechanistically, coimmunoprecipitation and immunofluorescent staining results confirmed that 4.1R can impair the activation of EGFR through direct binding to EGFR and reduce the downstream signaling. Taken together, a deficiency of 4.1R would therefore serve to sustain aberrant EGFR-mediated cellular signaling, leading to hyperproliferation. Our findings highlight the role of 4.1R in the regulation of EGFR signaling in keratinocytes and suggest that 4.1R acts as a novel regulator for EGFR activation.


Subject(s)
Cell Proliferation/physiology , ErbB Receptors/metabolism , Keratinocytes/metabolism , MAP Kinase Signaling System/physiology , Microfilament Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Animals , Cell Proliferation/drug effects , Cytoskeletal Proteins/metabolism , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Keratinocytes/drug effects , Keratinocytes/pathology , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation/drug effects , Phosphorylation/physiology , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Skin/drug effects , Skin/metabolism , Up-Regulation/drug effects , Up-Regulation/physiology
16.
Immunology ; 157(4): 312-321, 2019 08.
Article in English | MEDLINE | ID: mdl-31135971

ABSTRACT

Protein 4.1R, an 80 000 MW membrane skeleton protein, is a vital component of the red blood cell membrane cytoskeleton that stabilizes the spectrin-actin network and regulates membrane properties of deformability and mechanical stability. It has been shown that 4.1R is expressed in T cells, including CD8+ T cells, but its role in CD8+ T cells remains unclear. Here, we have explored the role of 4.1R in CD8+ T cells using 4.1R-/- mice. Our results showed that cell activation, proliferation and secretion levels of interleukin-2 and interferon-γ were significantly increased in 4.1R-/- CD8+ T cells. Furthermore, the phosphorylation levels of linker for activation of T cells (LAT) and its downstream signaling molecule extracellular signal-regulated kinase were enhanced in the absence of 4.1R. In vitro co-immunoprecipitation experiments showed a direct interaction between 4.1R and LAT. Moreover, 4.1R-/- CD8+ T cells and mice exhibited an enhanced T-cell-dependent immune response. These data enabled the identification of a negative regulation function for 4.1R in CD8+ T cells by a direct association between 4.1R and LAT, possibly through inhibiting phosphorylation of LAT and then modulating intracellular signal transduction.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , CD8-Positive T-Lymphocytes/immunology , Lymphocyte Activation , Membrane Proteins/immunology , Microfilament Proteins/immunology , Phosphoproteins/immunology , Signal Transduction/immunology , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Line, Tumor , Humans , Membrane Proteins/genetics , Mice , Mice, Knockout , Microfilament Proteins/genetics , Phosphoproteins/genetics , Phosphorylation/genetics , Phosphorylation/immunology , Signal Transduction/genetics
17.
Front Immunol ; 10: 3068, 2019.
Article in English | MEDLINE | ID: mdl-31993060

ABSTRACT

Protein 4.1R, a member of the 4.1 family, functions as a bridge between cytoskeletal and plasma membrane proteins. It is expressed in T cells, where it binds to a linker for activation of T cell (LAT) family member 1 and inhibits its phosphorylation and downstream signaling events after T cell receptor triggering. The role of the 4.1R protein in cell activation through other immunoreceptors is not known. In this study, we used 4.1R-deficient (4.1R-KO) and 4.1R wild-type (WT) mice and explored the role of the 4.1R protein in the high-affinity IgE receptor (FcεRI) signaling in mast cells. We found that bone marrow mast cells (BMMCs) derived from 4.1R-KO mice showed normal growth in vitro and expressed FcεRI and c-KIT at levels comparable to WT cells. However, 4.1R-KO cells exhibited reduced antigen-induced degranulation, calcium response, and secretion of tumor necrosis factor-α. Chemotaxis toward antigen and stem cell factor (SCF) and spreading on fibronectin were also reduced in 4.1R-KO BMMCs, whereas prostaglandin E2-mediated chemotaxis was not affected. Antibody-induced aggregation of tetraspanin CD9 inhibited chemotaxis toward antigen in WT but not 4.1R-KO BMMCs, implying a CD9-4.1R protein cross-talk. Further studies documented that in the absence of 4.1R, antigen-mediated phosphorylation of FcεRI ß and γ subunits was not affected, but phosphorylation of SYK and subsequent signaling events such as phosphorylation of LAT1, phospholipase Cγ1, phosphatases (SHP1 and SHIP), MAP family kinases (p38, ERK, JNK), STAT5, CBL, and mTOR were reduced. Immunoprecipitation studies showed the presence of both LAT1 and LAT2 (LAT, family member 2) in 4.1R immunocomplexes. The positive regulatory role of 4.1R protein in FcεRI-triggered activation was supported by in vivo experiments in which 4.1R-KO mice showed the normal presence of mast cells in the ears and peritoneum, but exhibited impaired passive cutaneous anaphylaxis. The combined data indicate that the 4.1R protein functions as a positive regulator in the early activation events after FcεRI triggering in mast cells.


Subject(s)
Chemotaxis/immunology , Mast Cells/immunology , Microfilament Proteins/immunology , Receptors, IgE/immunology , Animals , Cell Degranulation/immunology , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/metabolism , Passive Cutaneous Anaphylaxis/immunology , Receptors, IgE/metabolism
18.
C R Biol ; 341(4): 219-227, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29705198

ABSTRACT

At the beginning of diplotene, the oocyte of Xenopus laevis is a cell of about 10-20 microns destined to increase 10,000-fold its size when the oocyte becomes filled with yolk platelets and has accumulated a great number of pigment granules in a half of its periphery. Its internal architecture is gradually accomplished during growth because of several factors, especially because of cytoskeletal changes. In the fully-grown oocyte, the cytoskeleton appears to sustain the eccentrically located germinal vesicle through arms radiating from the cortex to the germinal vesicle, a unique organization not to be found in other Amphibians. In this report, we summarized and analysed steps of cytoskeletal proteins and related mRNAs organization and function throughout diplotene stage, highlighting our studies in this animal model. The cytoskeletal proteins appear to exploit their activity with respect to ribosomal 60S subunit maturation and during translation. Most importantly, the polarity of the oocyte is achieved through a sophisticated and highly organized localization of mRNAs and cytoskeletal proteins in one side of the cell. This asymmetry will start the construction of the oocyte polarity that is instrumental for determining the characteristic of this cell, which will become an embryo. Moreover, in the same time membrane composition, conditioned by the underlying cytoskeletal organization, will acquire the prerequisites for sperm binding and fusion.


Subject(s)
Cytoskeleton/physiology , Oocytes/metabolism , Xenopus laevis , Animals , Cytoplasm/physiology , Female , Microtubules/metabolism , Oogenesis/physiology , RNA, Messenger
19.
Mol Cell Biol ; 37(9)2017 05 01.
Article in English | MEDLINE | ID: mdl-28193846

ABSTRACT

Exon 16 of protein 4.1R encodes a spectrin/actin-binding peptide critical for erythrocyte membrane stability. Its expression during erythroid differentiation is regulated by alternative pre-mRNA splicing. A UUUUCCCCCC motif situated between the branch point and the 3' splice site is crucial for inclusion. We show that the UUUU region and the last three C residues in this motif are necessary for the binding of splicing factors TIA1 and Pcbp1 and that these proteins appear to act in a collaborative manner to enhance exon 16 inclusion. This element also activates an internal exon when placed in a corresponding intronic position in a heterologous reporter. The impact of these two factors is further enhanced by high levels of RBM39, whose expression rises during erythroid differentiation as exon 16 inclusion increases. TIA1 and Pcbp1 associate in a complex containing RBM39, which interacts with U2AF65 and SF3b155 and promotes U2 snRNP recruitment to the branch point. Our results provide a mechanism for exon 16 3' splice site activation in which a coordinated effort among TIA1, Pcbp1, and RBM39 stabilizes or increases U2 snRNP recruitment, enhances spliceosome A complex formation, and facilitates exon definition through RBM39-mediated splicing regulation.


Subject(s)
Alternative Splicing/genetics , Cytoskeletal Proteins/genetics , Erythropoiesis/physiology , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Membrane Proteins/genetics , Nuclear Proteins/metabolism , Poly(A)-Binding Proteins/metabolism , RNA-Binding Proteins/metabolism , Animals , Binding Sites/genetics , Cell Line, Tumor , DNA-Binding Proteins , Erythropoiesis/genetics , HEK293 Cells , HeLa Cells , Humans , Mice , Phosphoproteins/metabolism , Protein Binding/genetics , RNA Splicing Factors/metabolism , Ribonucleoproteins, Small Nuclear/metabolism , Spliceosomes/metabolism , Splicing Factor U2AF/metabolism , T-Cell Intracellular Antigen-1
20.
J Biol Chem ; 291(49): 25591-25607, 2016 Dec 02.
Article in English | MEDLINE | ID: mdl-27780863

ABSTRACT

Protein 4.1R (4.1R) isoforms are expressed in both cardiac and skeletal muscle. 4.1R is a component of the contractile apparatus. It is also associated with dystrophin at the sarcolemma in skeletal myofibers. However, the expression and function of 4.1R during myogenesis have not been characterized. We now report that 4.1R expression increases during C2C12 myoblast differentiation into myotubes. Depletion of 4.1R impairs skeletal muscle differentiation and is accompanied by a decrease in the levels of myosin heavy and light chains and caveolin-3. Furthermore, the expression of myogenin at the protein, but not mRNA, level is drastically decreased in 4.1R knockdown myocytes. Similar results were obtained using MyoD-induced differentiation of 4.1R-/- mouse embryonic fibroblast cells. von Hippel-Lindau (VHL) protein is known to destabilize myogenin via the ubiquitin-proteasome pathway. We show that 4.1R associates with VHL and, when overexpressed, reverses myogenin ubiquitination and stability. This suggests that 4.1R may influence myogenesis by preventing VHL-mediated myogenin degradation. Together, our results define a novel biological function for 4.1R in muscle differentiation and provide a molecular mechanism by which 4.1R promotes myogenic differentiation.


Subject(s)
Cell Differentiation/physiology , Cytoskeletal Proteins/metabolism , Membrane Proteins/metabolism , Myoblasts, Skeletal/metabolism , Myogenin/metabolism , Proteolysis , Animals , Cell Line , Cytoskeletal Proteins/genetics , Membrane Proteins/genetics , Mice , Mice, Knockout , MyoD Protein/genetics , MyoD Protein/metabolism , Myogenin/genetics , Protein Stability , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL