Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20.882
Filtrar
1.
Curr Biol ; 34(13): R623-R625, 2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-38981426

RESUMEN

The fly Drosophila yakuba has lost an ancestral component of the male courtship song: this is due to ontogenetic death of effector neurons in the ventral nerve cord, a result of the D. yakuba sex-determining gene dsx producing a male isoform, dsxM, with cell-death-promoting activity similar to that of the female isoform, dsxF, in D. melanogaster.


Asunto(s)
Cortejo , Proteínas de Drosophila , Drosophila , Conducta Sexual Animal , Animales , Masculino , Conducta Sexual Animal/fisiología , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Femenino , Drosophila/fisiología , Drosophila/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Neuronas/fisiología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
2.
Methods Mol Biol ; 2836: 3-17, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38995532

RESUMEN

Proteogenomics has revealed the translation of unannotated open reading frames (ORFs) present in mRNAs and in noncoding RNAs (ncRNAs). OpenProt annotates all ORFs with a minimum of 30 codons in the transcriptome of several species and displays many functional features associated with the corresponding proteins. Two types of proteins are annotated: reference or canonical proteins which are proteins already annotated in UniProt, RefSeq, or Ensembl and noncanonical proteins. Noncanonical proteins form two groups: predicted novel isoforms that display a significant level of homology with a reference protein and alternative proteins that are new proteins with no significant homology to known proteins. This chapter describes how to check whether a gene and/or transcript contains multiple open reading frames and how to use OpenProt databases for the detection of alternative proteins and novel isoforms by mass spectrometry-based proteomics.


Asunto(s)
Espectrometría de Masas , Sistemas de Lectura Abierta , Proteoma , Espectrometría de Masas/métodos , Proteómica/métodos , Bases de Datos de Proteínas , Humanos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Anotación de Secuencia Molecular , Proteogenómica/métodos
3.
Int J Mol Sci ; 25(13)2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-39000396

RESUMEN

Latrophilins (LPHNs), a group of the G-protein-coupled receptor to which a spider venom latrotoxin (LTX) is known to bind, remain largely uncharacterized in neoplastic diseases. In the present study, we aimed to determine the role of LPHNs in the progression of prostate cancer. We assessed the actions of LPHNs, including LPHN1, LPHN2, and LPHN3, in human prostate cancer lines via their ligand (e.g., α-LTX, FLRT3) treatment or shRNA infection, as well as in surgical specimens. In androgen receptor (AR)-positive LNCaP/C4-2/22Rv1 cells, dihydrotestosterone considerably increased the expression levels of LPHNs, while chromatin immunoprecipitation assay revealed the binding of endogenous ARs, including AR-V7, to the promoter region of each LPHN. Treatment with α-LTX or FLRT3 resulted in induction in the cell viability and migration of both AR-positive and AR-negative lines. α-LTX and FLRT3 also enhanced the expression of Bcl-2 and phosphorylated forms of JAK2 and STAT3. Meanwhile, the knockdown of each LPHN showed opposite effects on all of those mediated by ligand treatment. Immunohistochemistry in radical prostatectomy specimens further showed the significantly elevated expression of each LPHN in prostate cancer, compared with adjacent normal-appearing prostate, which was associated with a significantly higher risk of postoperative biochemical recurrence in both univariate and multivariable settings. These findings indicate that LPHNs function as downstream effectors of ARs and promote the growth of androgen-sensitive, castration-resistant, or even AR-negative prostate cancer.


Asunto(s)
Progresión de la Enfermedad , Neoplasias de la Próstata , Receptores Androgénicos , Masculino , Humanos , Receptores Androgénicos/metabolismo , Receptores Androgénicos/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Línea Celular Tumoral , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/genética , Movimiento Celular/genética , Movimiento Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Janus Quinasa 2/metabolismo , Janus Quinasa 2/genética , Receptores de Péptidos/metabolismo , Receptores de Péptidos/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/genética , Transducción de Señal , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Empalme Alternativo
4.
Mol Neurodegener ; 19(1): 54, 2024 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-39026372

RESUMEN

Abnormal accumulation of misfolded and hyperphosphorylated tau protein in brain is the defining feature of several neurodegenerative diseases called tauopathies, including Alzheimer's disease (AD). In AD, this pathological change is reflected by highly specific cerebrospinal fluid (CSF) tau biomarkers, including both phosphorylated and non-phosphorylated variants. Interestingly, despite tau pathology being at the core of all tauopathies, CSF tau biomarkers remain unchanged in certain tauopathies, e.g., progressive supranuclear palsy (PSP), Pick's disease (PiD), and corticobasal neurodegeneration (CBD). To better understand commonalities and differences between tauopathies, we report a multiplex assay combining immunoprecipitation and high-resolution mass spectrometry capable of detecting and quantifying peptides from different tau protein isoforms as well as non-phosphorylated and phosphorylated peptides, including those carrying multiple phosphorylations. We investigated the tau proteoforms in soluble and insoluble fractions of brain tissue from subjects with autopsy-confirmed tauopathies, including sporadic AD (n = 10), PSP (n = 11), PiD (n = 10), and CBD (n = 10), and controls (n = 10). Our results demonstrate that non-phosphorylated tau profiles differ across tauopathies, generally showing high abundance of microtubule-binding region (MTBR)-containing peptides in insoluble protein fractions compared with controls; the AD group showed 12-72 times higher levels of MTBR-containing aggregates. Quantification of tau isoforms showed the 3R being more abundant in PiD and the 4R isoform being more abundant in CBD and PSP in the insoluble fraction. Twenty-three different phosphorylated peptides were quantified. Most phosphorylated peptides were measurable in all investigated tauopathies. All phosphorylated peptides were significantly increased in AD insoluble fraction. However, doubly and triply phosphorylated peptides were significantly increased in AD even in the soluble fraction. Results were replicated using a validation cohort comprising AD (n = 10), CBD (n = 10), and controls (n = 10). Our study demonstrates that abnormal levels of phosphorylation and aggregation do indeed occur in non-AD tauopathies, however, both appear pronouncedly increased in AD, becoming a distinctive characteristic of AD pathology.


Asunto(s)
Encéfalo , Tauopatías , Proteínas tau , Humanos , Proteínas tau/metabolismo , Tauopatías/metabolismo , Anciano , Encéfalo/metabolismo , Encéfalo/patología , Masculino , Femenino , Persona de Mediana Edad , Fosforilación , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Anciano de 80 o más Años , Parálisis Supranuclear Progresiva/metabolismo , Isoformas de Proteínas/metabolismo
5.
Molecules ; 29(13)2024 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-38998944

RESUMEN

Actin, which plays a crucial role in cellular structure and function, interacts with various binding proteins, notably myosin. In mammals, actin is composed of six isoforms that exhibit high levels of sequence conservation and structural similarity overall. As a result, the selection of actin isoforms was considered unimportant in structural studies of their binding with myosin. However, recent high-resolution structural research discovered subtle structural differences in the N-terminus of actin isoforms, suggesting the possibility that each actin isoform may engage in specific interactions with myosin isoforms. In this study, we aimed to explore this possibility, particularly by understanding the influence of different actin isoforms on the interaction with myosin 7A. First, we compared the reported actomyosin structures utilizing the same type of actin isoforms as the high-resolution filamentous skeletal α-actin (3.5 Å) structure elucidated using cryo-EM. Through this comparison, we confirmed that the diversity of myosin isoforms leads to differences in interaction with the actin N-terminus, and that loop 2 of the myosin actin-binding sites directly interacts with the actin N-terminus. Subsequently, with the aid of multiple sequence alignment, we observed significant variations in the length of loop 2 across different myosin isoforms. We predicted that these length differences in loop 2 would likely result in structural variations that would affect the interaction with the actin N-terminus. For myosin 7A, loop 2 was found to be very short, and protein complex predictions using skeletal α-actin confirmed an interaction between loop 2 and the actin N-terminus. The prediction indicated that the positively charged residues present in loop 2 electrostatically interact with the acidic patch residues D24 and D25 of actin subdomain 1, whereas interaction with the actin N-terminus beyond this was not observed. Additionally, analyses of the actomyosin-7A prediction models generated using various actin isoforms consistently yielded the same results regardless of the type of actin isoform employed. The results of this study suggest that the subtle structural differences in the N-terminus of actin isoforms are unlikely to influence the binding structure with short loop 2 myosin 7A. Our findings are expected to provide a deeper understanding for future high-resolution structural binding studies of actin and myosin.


Asunto(s)
Actinas , Miosinas , Unión Proteica , Isoformas de Proteínas , Actinas/química , Actinas/metabolismo , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Miosinas/química , Miosinas/metabolismo , Sitios de Unión , Animales , Modelos Moleculares , Secuencia de Aminoácidos , Microscopía por Crioelectrón , Humanos
6.
Int J Mol Sci ; 25(13)2024 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-38999987

RESUMEN

The actin cytoskeleton is one of the most important players in cell motility, adhesion, division, and functioning. The regulation of specific microfilament formation largely determines cellular functions. The main actin-binding protein in animal cells is tropomyosin (Tpm). The unique structural and functional diversity of microfilaments is achieved through the diversity of Tpm isoforms. In our work, we studied the properties of the cytoplasmic isoforms Tpm1.8 and Tpm1.9. The results showed that these isoforms are highly thermostable and differ in the stability of their central and C-terminal fragments. The properties of these isoforms were largely determined by the 6th exons. Thus, the strength of the end-to-end interactions, as well as the affinity of the Tpm molecule for F-actin, differed between the Tpm1.8 and Tpm1.9 isoforms. They were determined by whether an alternative internal exon, 6a or 6b, was included in the Tpm isoform structure. The strong interactions of the Tpm1.8 and Tpm1.9 isoforms with F-actin led to the formation of rigid actin filaments, the stiffness of which was measured using an optical trap. It is quite possible that the structural and functional features of the Tpm isoforms largely determine the appearance of these isoforms in the rigid actin structures of the cell cortex.


Asunto(s)
Citoesqueleto de Actina , Actinas , Isoformas de Proteínas , Tropomiosina , Tropomiosina/metabolismo , Tropomiosina/química , Tropomiosina/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Citoesqueleto de Actina/metabolismo , Animales , Actinas/metabolismo , Actinas/química , Citoplasma/metabolismo , Humanos , Exones , Unión Proteica , Estabilidad Proteica
7.
Biochem Biophys Res Commun ; 727: 150321, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-38954982

RESUMEN

Vascular endothelial growth factor (VEGF) is a pleiotropic growth factor that binds a broad spectrum of cell types and regulates diverse cellular processes, including angiogenesis, growth and survival. However, it is technically difficult to quantify VEGF-cell binding activity because of reversible nature of ligand-receptor interactions. Here we used T7 bacteriophage display to quantify and compare binding activity of three human VEGF-A (hVEGF) isoforms, including hVEGF111, 165 and 206. All three isoforms bound equally well to immobilized aflibercept, a decoy VEGF receptor. hVEGF111-Phage exhibited minimal binding to immobilized heparan sulfate, whereas hVEGF206-Phage and hVEGF165-Phage had the highest and intermediate binding to heparan, respectively. In vitro studies revealed that all three isoforms bound to human umbilical vein endothelial cells (HUVECs), HEK293 epithelial and SK-N-AS neuronal cells. hVEGF111-Phage has the lowest binding activity, while hVEGF206-Phage has the highest binding. hVEGF206-Phage was the most sensitive to detect VEGF-cell binding, albeit with the highest background binding to SK-N-AS cells. These results suggest that hVEGF206-Phage is the best-suited isoform to quantify VEGF-cell binding even though VEGF165 is the most biologically active. Furthermore, this study demonstrates the utility of T7 phage display as a platform for rapid and convenient ligand-cell binding quantification with pros and cons discussed.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana , Unión Proteica , Factor A de Crecimiento Endotelial Vascular , Humanos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células HEK293 , Isoformas de Proteínas/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Bacteriófago T7/metabolismo , Bacteriófago T7/genética , Técnicas de Visualización de Superficie Celular/métodos , Heparitina Sulfato/metabolismo , Proteínas Recombinantes de Fusión
8.
Dev Comp Immunol ; 159: 105225, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38992732

RESUMEN

Members of the myxovirus resistance (Mx) protein family play an essential role in antiviral immunity. They are Dynamin-like GTPases, induced by interferons. In the current study, we have characterized two predicted MX genes (MX1 and MX2) from lumpfish (Cyclopterus lumpus L.), having 12 and 13 exons, respectively. Mx2 has two isoforms (Mx2-X1 and Mx2-X2) which differ in exon 1. The lumpfish Mx proteins contain an N-terminal Dynamin-like GTPase domain, the middle domain (MD) and GTPase effector domain (GED) characteristic for Mx proteins. Phylogenetic analyses grouped all the lumpfish Mx sequences in group 1, and synteny analyses showed that both genes were localized at chromosome 5 in proximity to the genes Tohc7, Atxn7 and Psmd6. In vitro stimulation experiment showed that both MX1 and MX2-X2 were highly upregulated upon exposure to poly(I:C), but not bacteria, 24 h post exposure, indicating their role in antiviral immunity.


Asunto(s)
Proteínas de Peces , Proteínas de Resistencia a Mixovirus , Filogenia , Proteínas de Resistencia a Mixovirus/genética , Proteínas de Resistencia a Mixovirus/metabolismo , Animales , Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Poli I-C/inmunología , Inmunidad Innata/genética , Perciformes/inmunología , Perciformes/genética , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/virología , Peces/inmunología , Peces/genética , Sintenía , Familia de Multigenes , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
9.
Proc Natl Acad Sci U S A ; 121(29): e2319829121, 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-38976736

RESUMEN

In the developing human brain, only 53 stochastically expressed clustered protocadherin (cPcdh) isoforms enable neurites from individual neurons to recognize and self-avoid while simultaneously maintaining contact with neurites from other neurons. Cell assays have demonstrated that self-recognition occurs only when all cPcdh isoforms perfectly match across the cell boundary, with a single mismatch in the cPcdh expression profile interfering with recognition. It remains unclear, however, how a single mismatched isoform between neighboring cells is sufficient to block erroneous recognitions. Using systematic cell aggregation experiments, we show that abolishing cPcdh interactions on the same membrane (cis) results in a complete loss of specific combinatorial binding between cells (trans). Our computer simulations demonstrate that the organization of cPcdh in linear array oligomers, composed of cis and trans interactions, enhances self-recognition by increasing the concentration and stability of cPcdh trans complexes between the homotypic membranes. Importantly, we show that the presence of mismatched isoforms between cells drastically diminishes the concentration and stability of the trans complexes. Overall, we provide an explanation for the role of the cPcdh assembly arrangements in neuronal self/non-self-discrimination underlying neuronal self-avoidance.


Asunto(s)
Cadherinas , Neuronas , Isoformas de Proteínas , Humanos , Neuronas/metabolismo , Cadherinas/metabolismo , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/genética , Comunicación Celular , Simulación por Computador , Neuritas/metabolismo , Membrana Celular/metabolismo
10.
Cell Mol Life Sci ; 81(1): 301, 2024 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-39003683

RESUMEN

Voltage-gated K+ (KV) channels govern K+ ion flux across cell membranes in response to changes in membrane potential. They are formed by the assembly of four subunits, typically from the same family. Electrically silent KV channels (KVS), however, are unable to conduct currents on their own. It has been assumed that these KVS must obligatorily assemble with subunits from the KV2 family into heterotetrameric channels, thereby giving rise to currents distinct from those of homomeric KV2 channels. Herein, we show that KVS subunits indeed also modulate the activity, biophysical properties and surface expression of recombinant KV7 isoforms in a subunit-specific manner. Employing co-immunoprecipitation, and proximity labelling, we unveil the spatial coexistence of KVS and KV7 within a single protein complex. Electrophysiological experiments further indicate functional interaction and probably heterotetramer formation. Finally, single-cell transcriptomic analyses identify native cell types in which this KVS and KV7 interaction may occur. Our findings demonstrate that KV cross-family interaction is much more versatile than previously thought-possibly serving nature to shape potassium conductance to the needs of individual cell types.


Asunto(s)
Subunidades de Proteína , Humanos , Animales , Subunidades de Proteína/metabolismo , Células HEK293 , Potenciales de la Membrana , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/genética , Canales de Potasio con Entrada de Voltaje/metabolismo , Canales de Potasio con Entrada de Voltaje/genética , Canal de Potasio KCNQ1/metabolismo , Canal de Potasio KCNQ1/genética
11.
Sci Rep ; 14(1): 13246, 2024 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-38853173

RESUMEN

Although alternative splicing (AS) is a major mechanism that adds diversity to gene expression patterns, its precise role in generating variability in ribosomal proteins, known as ribosomal heterogeneity, remains unclear. The ribosomal protein S24 (RPS24) gene, encoding a ribosomal component, undergoes AS; however, in-depth studies have been challenging because of three microexons between exons 4 and 6. We conducted a detailed analysis of RPS24 AS isoforms using a direct approach to investigate the splicing junctions related to these microexons, focusing on four AS isoforms. Each of these isoforms showed tissue specificity and relative differences in expression among cancer types. Significant differences in the proportions of these RPS24 AS isoforms between cancerous and normal tissues across diverse cancer types were also observed. Our study highlighted a significant correlation between the expression levels of a specific RPS24 AS isoform and the epithelial-mesenchymal transition process in lung and breast cancers. Our research contributes to a better understanding of the intricate regulatory mechanisms governing AS of ribosomal protein genes and highlights the biological implications of RPS24 AS isoforms in tissue development and tumorigenesis.


Asunto(s)
Empalme Alternativo , Biomarcadores de Tumor , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Proteínas Ribosómicas , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo , Transición Epitelial-Mesenquimal/genética , Humanos , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Neoplasias/genética , Neoplasias/patología , Progresión de la Enfermedad , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Femenino , Línea Celular Tumoral , Exones/genética
12.
Int J Mol Sci ; 25(11)2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38892298

RESUMEN

Periostin, a multifunctional 90 kDa protein, plays a pivotal role in the pathogenesis of fibrosis across various tissues, including skeletal muscle. It operates within the transforming growth factor beta 1 (Tgf-ß1) signalling pathway and is upregulated in fibrotic tissue. Alternative splicing of Periostin's C-terminal region leads to six protein-coding isoforms. This study aimed to elucidate the contribution of the isoforms containing the amino acids encoded by exon 17 (e17+ Periostin) to skeletal muscle fibrosis and investigate the therapeutic potential of manipulating exon 17 splicing. We identified distinct structural differences between e17+ Periostin isoforms, affecting their interaction with key fibrotic proteins, including Tgf-ß1 and integrin alpha V. In vitro mouse fibroblast experimentation confirmed the TGF-ß1-induced upregulation of e17+ Periostin mRNA, mitigated by an antisense approach that induces the skipping of exon 17 of the Postn gene. Subsequent in vivo studies in the D2.mdx mouse model of Duchenne muscular dystrophy (DMD) demonstrated that our antisense treatment effectively reduced e17+ Periostin mRNA expression, which coincided with reduced full-length Periostin protein expression and collagen accumulation. The grip strength of the treated mice was rescued to the wild-type level. These results suggest a pivotal role of e17+ Periostin isoforms in the fibrotic pathology of skeletal muscle and highlight the potential of targeted exon skipping strategies as a promising therapeutic approach for mitigating fibrosis-associated complications.


Asunto(s)
Empalme Alternativo , Moléculas de Adhesión Celular , Exones , Fibrosis , Ratones Endogámicos mdx , Oligonucleótidos Antisentido , Animales , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Ratones , Oligonucleótidos Antisentido/farmacología , Oligonucleótidos Antisentido/genética , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/genética , Fibroblastos/metabolismo , Modelos Animales de Enfermedad , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Masculino
13.
BMB Rep ; 57(6): 263-272, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38835114

RESUMEN

Amyloid-ß (Aß) is one of the amyloidogenic intrinsically disordered proteins (IDPs) that self-assemble to protein aggregates, incurring cell malfunction and cytotoxicity. While Aß has been known to regulate multiple physiological functions, such as enhancing synaptic functions, aiding in the recovery of the blood-brain barrier/brain injury, and exhibiting tumor suppression/antimicrobial activities, the hydrophobicity of the primary structure promotes pathological aggregations that are closely associated with the onset of Alzheimer's disease (AD). Aß proteins consist of multiple isoforms with 37-43 amino acid residues that are produced by the cleavage of amyloid-ß precursor protein (APP). The hydrolytic products of APP are secreted to the extracellular regions of neuronal cells. Aß 1-42 (Aß42) and Aß 1-40 (Aß40) are dominant isoforms whose significance in AD pathogenesis has been highlighted in numerous studies to understand the molecular mechanism and develop AD diagnosis and therapeutic strategies. In this review, we focus on the differences between Aß42 and Aß40 in the molecular mechanism of amyloid aggregations mediated by the two additional residues (Ile41 and Ala42) of Aß42. The current comprehension of Aß42 and Aß40 in AD progression is outlined, together with the structural features of Aß42/Aß40 amyloid fibrils, and the aggregation mechanisms of Aß42/Aß40. Furthermore, the impact of the heterogeneous distribution of Aß isoforms during amyloid aggregations is discussed in the system mimicking the coexistence of Aß42 and Aß40 in human cerebrospinal fluid (CSF) and plasma. [BMB Reports 2024; 57(6): 263-272].


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Isoformas de Proteínas , Animales , Humanos , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/química , Agregado de Proteínas/fisiología , Agregación Patológica de Proteínas/metabolismo , Isoformas de Proteínas/metabolismo
14.
Int J Mol Sci ; 25(11)2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38891862

RESUMEN

RNA processing is a highly conserved mechanism that serves as a pivotal regulator of gene expression. Alternative processing generates transcripts that can still be translated but lead to potentially nonfunctional proteins. A plethora of respiratory viruses, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), strategically manipulate the host's RNA processing machinery to circumvent antiviral responses. We integrated publicly available omics datasets to systematically analyze isoform-level expression and delineate the nascent peptide landscape of SARS-CoV-2-infected human cells. Our findings explore a suggested but uncharacterized mechanism, whereby SARS-CoV-2 infection induces the predominant expression of unproductive splicing isoforms in key IFN signaling, interferon-stimulated (ISGs), class I MHC, and splicing machinery genes, including IRF7, HLA-B, and HNRNPH1. In stark contrast, cytokine and chemokine genes, such as IL6 and TNF, predominantly express productive (protein-coding) splicing isoforms in response to SARS-CoV-2 infection. We postulate that SARS-CoV-2 employs an unreported tactic of exploiting the host splicing machinery to bolster viral replication and subvert the immune response by selectively upregulating unproductive splicing isoforms from antigen presentation and antiviral response genes. Our study sheds new light on the molecular interplay between SARS-CoV-2 and the host immune system, offering a foundation for the development of novel therapeutic strategies to combat COVID-19.


Asunto(s)
Empalme Alternativo , COVID-19 , Interferones , Isoformas de Proteínas , SARS-CoV-2 , Humanos , SARS-CoV-2/genética , COVID-19/virología , COVID-19/genética , COVID-19/inmunología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Interferones/metabolismo , Interferones/genética , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/metabolismo
15.
Cells ; 13(12)2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38920651

RESUMEN

Bone formation is a complex process regulated by a variety of pathways that are not yet fully understood. One of the proteins involved in multiple osteogenic pathways is TID (DNAJA3). The aim of this work was to study the association of TID with osteogenesis. Therefore, the expression profiles of the TID splice variants (TID-L, TID-I) and their protein products were analyzed during the proliferation and differentiation of bone marrow mesenchymal stromal cells (B-MSCs) into osteoblasts. As the reference, the hFOB1.19 cell line was used. The phenotype of B-MSCs was confirmed by the presence of CD73, CD90, and CD105 surface antigens on ~97% of cells. The osteoblast phenotype was confirmed by increased alkaline phosphatase activity, calcium deposition, and expression of ALPL and SPP1. The effect of silencing the TID gene on the expression of ALPL and SPP1 was also investigated. The TID proteins and the expression of TID splice variants were detected. After differentiation, the expression of TID-L and TID-I increased 5-fold and 3.7-fold, respectively, while their silencing resulted in increased expression of SPP1. Three days after transfection, the expression of SPP1 increased 7.6-fold and 5.6-fold in B-MSCs and differentiating cells, respectively. Our preliminary study demonstrated that the expression of TID-L and TID-I changes under differentiation of B-MSCs into osteoblasts and may influence the expression of SPP1. However, for better understanding the functional association of these results with the relevant osteogenic pathways, further studies are needed.


Asunto(s)
Diferenciación Celular , Células Madre Mesenquimatosas , Osteoblastos , Osteogénesis , Humanos , Osteoblastos/metabolismo , Osteoblastos/citología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Diferenciación Celular/genética , Osteogénesis/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/genética , Fosfatasa Alcalina/metabolismo , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/citología , Proliferación Celular
16.
Sci Rep ; 14(1): 14715, 2024 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-38926482

RESUMEN

Opioids are the gold standard for the treatment of chronic pain but are limited by adverse side effects. In our earlier work, we showed that Heat shock protein 90 (Hsp90) has a crucial role in regulating opioid signaling in spinal cord; Hsp90 inhibition in spinal cord enhances opioid anti-nociception. Building on these findings, we injected the non-selective Hsp90 inhibitor KU-32 by the intrathecal route into male and female CD-1 mice, showing that morphine anti-nociceptive potency was boosted by 1.9-3.5-fold in acute and chronic pain models. At the same time, tolerance was reduced from 21-fold to 2.9 fold and established tolerance was rescued, while the potency of constipation and reward was unchanged. These results demonstrate that spinal Hsp90 inhibition can improve the therapeutic index of morphine. However, we also found that systemic non-selective Hsp90 inhibition blocked opioid pain relief. To avoid this effect, we used selective small molecule inhibitors and CRISPR gene editing to identify 3 Hsp90 isoforms active in spinal cord (Hsp90α, Hsp90ß, and Grp94) while only Hsp90α was active in brain. We thus hypothesized that a systemically delivered selective inhibitor to Hsp90ß or Grp94 could selectively inhibit spinal cord Hsp90 activity, resulting in enhanced opioid therapy. We tested this hypothesis using intravenous delivery of KUNB106 (Hsp90ß) and KUNG65 (Grp94), showing that both drugs enhanced morphine anti-nociceptive potency while rescuing tolerance. Together, these results suggest that selective inhibition of spinal cord Hsp90 isoforms is a novel, translationally feasible strategy to improve the therapeutic index of opioids.


Asunto(s)
Analgésicos Opioides , Proteínas HSP90 de Choque Térmico , Morfina , Médula Espinal , Animales , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP90 de Choque Térmico/metabolismo , Médula Espinal/metabolismo , Médula Espinal/efectos de los fármacos , Ratones , Analgésicos Opioides/farmacología , Masculino , Femenino , Morfina/farmacología , Isoformas de Proteínas/metabolismo , Tolerancia a Medicamentos , Dolor Crónico/tratamiento farmacológico , Dolor Crónico/metabolismo , Modelos Animales de Enfermedad , Inyecciones Espinales
17.
Genes (Basel) ; 15(6)2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38927715

RESUMEN

Peroxisome proliferator-activated receptor γ (PPARG) has various splicing variants and plays essential roles in the regulation of adipocyte differentiation and lipogenesis. However, little is known about the expression pattern and effect of the PPARG on milk fat synthesis in the buffalo mammary gland. In this study, we found that only PPARG-X17 and PPARG-X21 of the splicing variant were expressed in the buffalo mammary gland. Amino acid sequence characterization showed that the proteins encoded by PPARG-X17 and PPARG-X21 are endonuclear non-secreted hydrophilic proteins. Protein domain prediction found that only the PPARG-X21-encoded protein had PPAR ligand-binding domains (NR_LBD_PPAR), which may lead to functional differences between the two splices. RNA interference (RNAi) and the overexpression of PPARG-X17 and PPARG-X21 in buffalo mammary epithelial cells (BMECs) were performed. Results showed that the expression of fatty acid synthesis-related genes (ACACA, CD36, ACSL1, GPAT, AGPAT6, DGAT1) was significantly modified (p < 0.05) by the RNAi and overexpression of PPARG-X17 and PPARG-X21. All kinds of FAs detected in this study were significantly decreased (p < 0.05) after RNAi of PPARG-X17 or PPARG-X21. Overexpression of PPARG-X17 or PPARG-X21 significantly decreased (p < 0.05) the SFA content, while significantly increased (p < 0.05) the UFA, especially the MUFA in the BMECs. In conclusion, there are two PPARG splicing variants expressed in the BMECs that can regulate FA synthesis by altering the expression of diverse fatty acid synthesis-related genes. This study revealed the expression characteristics and functions of the PPARG gene in buffalo mammary glands and provided a reference for further understanding of fat synthesis in buffalo milk.


Asunto(s)
Búfalos , Glándulas Mamarias Animales , PPAR gamma , Animales , Búfalos/genética , Búfalos/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , Glándulas Mamarias Animales/metabolismo , Femenino , Células Epiteliales/metabolismo , Empalme Alternativo , Ácidos Grasos/metabolismo , Ácidos Grasos/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Leche/metabolismo
18.
Front Endocrinol (Lausanne) ; 15: 1395750, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38859907

RESUMEN

Background: The beneficial effect of thermogenic adipocytes in maintaining body weight and protecting against metabolic disorders has raised interest in understanding the regulatory mechanisms defining white and beige adipocyte identity. Although alternative splicing has been shown to propagate adipose browning signals in mice, this has yet to be thoroughly investigated in human adipocytes. Methods: We performed parallel white and beige adipogenic differentiation using primary adipose stem cells from 6 unrelated healthy subjects and assessed differential gene and isoform expression in mature adipocytes by RNA sequencing. Results: We find 777 exon junctions with robust differential usage between white and beige adipocytes in all 6 subjects, mapping to 562 genes. Importantly, only 10% of these differentially spliced genes are also differentially expressed, indicating that alternative splicing constitutes an additional layer of gene expression regulation during beige adipocyte differentiation. Functional classification of alternative isoforms points to a gain of function for key thermogenic transcription factors such as PPARG and CITED1, and enzymes such as PEMT, or LPIN1. We find that a large majority of the splice variants arise from differential TSS usage, with beige-specific TSSs being enriched for PPARγ and MED1 binding compared to white-specific TSSs. Finally, we validate beige specific isoform expression at the protein level for two thermogenic regulators, PPARγ and PEMT. Discussion: These results suggest that differential isoform expression through alternative TSS usage is an important regulatory mechanism for human adipocyte thermogenic specification.


Asunto(s)
Adipocitos Beige , Empalme Alternativo , Isoformas de Proteínas , Termogénesis , Humanos , Adipocitos Beige/metabolismo , Termogénesis/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Diferenciación Celular , Adipogénesis/genética , Masculino , Femenino , Adulto , Células Cultivadas , Regulación de la Expresión Génica , PPAR gamma/genética , PPAR gamma/metabolismo
19.
Cell Death Dis ; 15(6): 412, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38866752

RESUMEN

Full-length p53 (p53α) plays a pivotal role in maintaining genomic integrity and preventing tumor development. Over the years, p53 was found to exist in various isoforms, which are generated through alternative splicing, alternative initiation of translation, and internal ribosome entry site. p53 isoforms, either C-terminally altered or N-terminally truncated, exhibit distinct biological roles compared to p53α, and have significant implications for tumor development and therapy resistance. Due to a lack of part and/or complete C- or N-terminal domains, ectopic expression of some p53 isoforms failed to induce expression of canonical transcriptional targets of p53α like CDKN1A or MDM2, even though they may bind their promoters. Yet, p53 isoforms like Δ40p53α still activate subsets of targets including MDM2 and BAX. Furthermore, certain p53 isoforms transactivate even novel targets compared to p53α. More recently, non-canonical functions of p53α in DNA repair and of different isoforms in DNA replication unrelated to transcriptional activities were discovered, amplifying the potential of p53 as a master regulator of physiological and tumor suppressor functions in human cells. Both regarding canonical and non-canonical functions, alternative p53 isoforms frequently exert dominant negative effects on p53α and its partners, which is modified by the relative isoform levels. Underlying mechanisms include hetero-oligomerization, changes in subcellular localization, and aggregation. These processes ultimately influence the net activities of p53α and give rise to diverse cellular outcomes. Biological roles of p53 isoforms have implications for tumor development and cancer therapy resistance. Dysregulated expression of isoforms has been observed in various cancer types and is associated with different clinical outcomes. In conclusion, p53 isoforms have expanded our understanding of the complex regulatory network involving p53 in tumors. Unraveling the mechanisms underlying the biological roles of p53 isoforms provides new avenues for studies aiming at a better understanding of tumor development and developing therapeutic interventions to overcome resistance.


Asunto(s)
Neoplasias , Isoformas de Proteínas , Proteína p53 Supresora de Tumor , Humanos , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/genética , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Animales , Resistencia a Antineoplásicos/genética , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/genética
20.
J Neurosci ; 44(28)2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38858079

RESUMEN

Tau pathologies are detected in the brains of some of the most common neurodegenerative diseases including Alzheimer's disease (AD), Lewy body dementia (LBD), chronic traumatic encephalopathy (CTE), and frontotemporal dementia (FTD). Tau proteins are expressed in six isoforms with either three or four microtubule-binding repeats (3R tau or 4R tau) due to alternative RNA splicing. AD, LBD, and CTE brains contain pathological deposits of both 3R and 4R tau. FTD patients can exhibit either 4R tau pathologies in most cases or 3R tau pathologies less commonly in Pick's disease, which is a subfamily of FTD. Here, we report the isoform-specific roles of tau in FTD. The P301L mutation, linked to familial 4R tau FTD, induces mislocalization of 4R tau to dendritic spines in primary hippocampal cultures that were prepared from neonatal rat pups of both sexes. Contrastingly, the G272V mutation, linked to familial Pick's disease, induces phosphorylation-dependent mislocalization of 3R tau but not 4R tau proteins to dendritic spines. The overexpression of G272V 3R tau but not 4R tau proteins leads to the reduction of dendritic spine density and suppression of mEPSCs in 5-week-old primary rat hippocampal cultures. The decrease in mEPSC amplitude caused by G272V 3R tau is dynamin-dependent whereas that caused by P301L 4R tau is dynamin-independent, indicating that the two tau isoforms activate different signaling pathways responsible for excitatory synaptic dysfunction. Our 3R and 4R tau studies here will shed new light on diverse mechanisms underlying FTD, AD, LBD, and CTE.


Asunto(s)
Espinas Dendríticas , Demencia Frontotemporal , Mutación , Isoformas de Proteínas , Proteínas tau , Proteínas tau/metabolismo , Proteínas tau/genética , Animales , Demencia Frontotemporal/genética , Demencia Frontotemporal/metabolismo , Demencia Frontotemporal/patología , Espinas Dendríticas/metabolismo , Espinas Dendríticas/patología , Ratas , Masculino , Humanos , Femenino , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Sinapsis/metabolismo , Sinapsis/patología , Ratas Sprague-Dawley , Hipocampo/metabolismo , Hipocampo/patología , Células Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...