Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.900
Filtrar
2.
J Cancer Res Clin Oncol ; 148(2): 283-291, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35037101

RESUMEN

BACKGROUND: Leucine-rich alpha-2-glycoprotein-1 (LRG1) is widely involved in proliferation, migration, and invasion of various tumor cells. Recent studies have evaluated the potential of LRG1 as both an early tumor and a prognostic biomarker. METHOD: The relevant literature from PubMed is reviewed in this article. RESULTS: It has been found that LRG1 mainly acts on the regulatory mechanisms of angiogenesis, epithelial-mesenchymal transition (EMT), and apoptosis by transforming growth factor (TGF-ß) signaling pathway as well as affecting the occurrence and development of the tumors. Moreover, with advancement of research, LRG1 regulation pathways which are independent of TGF-ß signaling pathway have been gradually revealed in different tumor cells; There are several studies on the biological effects of LRG1 as an inflammatory factor, vascular growth regulator, cell adhesion, and a cell viability influencing factor. In addition, various tumor suppression methods which are based on regulation of LRG1 levels have also shown high potential clinical value. CONCLUSIONS: LRG1 are critical for the processes of tumorigenesis, development, and metastasis in various tumors. The present study reviewed the latest research on the achievements of LRG1 in tumor genesis and development. Further, this study also discussed the related molecular mechanisms of various biological functions of LRG1.


Asunto(s)
Movimiento Celular/genética , Proliferación Celular/genética , Glicoproteínas/fisiología , Invasividad Neoplásica/genética , Animales , Apoptosis/genética , Supervivencia Celular/genética , Transición Epitelial-Mesenquimal/genética , Humanos , Neoplasias/genética , Neoplasias/patología , Neovascularización Patológica/genética , Neovascularización Patológica/patología
3.
Front Endocrinol (Lausanne) ; 12: 731966, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34671318

RESUMEN

The glycoprotein hormones (GPH) are heterodimers composed of a common α subunit and a specific ß subunit. They act by activating specific leucine-rich repeat G protein-coupled receptors. However, individual subunits have been shown to elicit responses in cells devoid of the receptor for the dimeric hormones. The α subunit is involved in prolactin production from different tissues. The human chorionic gonadotropin ß subunit (ßhCG) plays determinant roles in placentation and in cancer development and metastasis. A truncated form of the thyrotropin (TSH) ß subunit is also reported to have biological effects. The GPH α- and ß subunits are derived from precursor genes (gpa and gpb, respectively), which are expressed in most invertebrate species and are still represented in vertebrates as GPH subunit paralogs (gpa2 and gpb5, respectively). No specific receptor has been found for the vertebrate GPA2 and GPB5 even if their heterodimeric form is able to activate the TSH receptor in mammals. Interestingly, GPA and GPB are phylogenetically and structurally related to cysteine-knot growth factors (CKGF) and particularly to a group of antagonists that act independently on any receptor. This review article summarizes the observed actions of individual GPH subunits and presents the current hypotheses of how these actions might be induced. New approaches are also proposed in light of the evolutionary relatedness with antagonists of the CKGF family of proteins.


Asunto(s)
Glicoproteínas/fisiología , Hormonas Peptídicas/fisiología , Secuencia de Aminoácidos , Animales , Hormonas Glicoproteicas de Subunidad alfa/fisiología , Glicoproteínas/química , Humanos , Subunidades de Proteína/fisiología , Receptores Acoplados a Proteínas G/fisiología
4.
Genome Biol ; 22(1): 297, 2021 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-34686207

RESUMEN

BACKGROUND: Rhinoviruses (RVs) cause more than half of common colds and, in some cases, more severe diseases. Functional genomics analyses of RVs using siRNA or genome-wide CRISPR screen uncovered a limited set of host factors, few of which have proven clinical relevance. RESULTS: Herein, we systematically compare genome-wide CRISPR screen and surface protein-focused CRISPR screen, referred to as surfaceome CRISPR screen, for their efficiencies in identifying RV host factors. We find that surfaceome screen outperforms the genome-wide screen in the success rate of hit identification. Importantly, using the surfaceome screen, we identify olfactomedin-like 3 (OLFML3) as a novel host factor of RV serotypes A and B, including a clinical isolate. We find that OLFML3 is a RV-inducible suppressor of the innate immune response and that OLFML3 antagonizes type I interferon (IFN) signaling in a SOCS3-dependent manner. CONCLUSION: Our study suggests that RV-induced OLFML3 expression is an important mechanism for RV to hijack the immune system and underscores surfaceome CRISPR screen in identifying viral host factors.


Asunto(s)
Sistemas CRISPR-Cas , Glicoproteínas/metabolismo , Interferón Tipo I/antagonistas & inhibidores , Rhinovirus/fisiología , Genoma Humano , Glicoproteínas/fisiología , Células HeLa , Humanos , Inmunidad Innata , Transducción de Señal , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , Proteínas de Unión al GTP rab5/fisiología
5.
Mol Vis ; 27: 480-493, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34497454

RESUMEN

Glaucoma a debilitating disease, is globally the second most common kind of permanent blindness. Primary open-angle glaucoma (POAG) is its most prevalent form and is often linked with alterations in the myocilin gene (MYOC). MYOC encodes the myocilin protein, which is expressed throughout the body, but primarily in trabecular meshwork (TM) tissue in the eyes. TM is principally involved in regulating intraocular pressure (IOP), and elevated IOP is the main risk factor associated with glaucoma. The myocilin protein's function remains unknown; however, mutations compromise its folding and processing inside TM cells, contributing to the glaucoma phenotype. While glaucoma is a complex disease with various molecules and factors as contributing causes, the role played by myocilin has been the most widely studied. The current review describes the present understanding of myocilin and its association with glaucoma and aims to shift the focus toward developing targeted therapies for treating glaucoma patients with variations in MYOC.


Asunto(s)
Proteínas del Citoesqueleto/fisiología , Proteínas del Ojo/fisiología , Glaucoma de Ángulo Abierto/fisiopatología , Glicoproteínas/fisiología , Humanos , Presión Intraocular/fisiología , Terapia Molecular Dirigida , Malla Trabecular/metabolismo
6.
Int J Mol Sci ; 22(14)2021 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-34299131

RESUMEN

The use of immune checkpoint inhibitors has revolutionized the treatment of melanoma patients, leading to remarkable improvements in the cure. However, to ensure a safe and effective treatment, there is the need to develop markers to identify the patients that would most likely respond to the therapies. The microenvironment is gaining attention in this context, since it can regulate both the immunotherapy efficacyand angiogenesis, which is known to be affected by treatment. Here, we investigated the putative role of the ECM molecule EMILIN-2, a tumor suppressive and pro-angiogenic molecule. We verified that the EMILIN2 expression is variable among melanoma patients and is associated with the response to PD-L1 inhibitors. Consistently, in preclinical settings,the absence of EMILIN-2 is associated with higher PD-L1 expression and increased immunotherapy efficacy. We verified that EMILIN-2 modulates PD-L1 expression in melanoma cells through indirect immune-dependent mechanisms. Notably, upon PD-L1 blockage, Emilin2-/- mice displayed improved intra-tumoral vessel normalization and decreased tumor hypoxia. Finally, we provide evidence indicating that the inclusion of EMILIN2 in a number of gene expression signatures improves their predictive potential, a further indication that the analysis of this molecule may be key for the development of new markers to predict immunotherapy efficacy.


Asunto(s)
Antígeno B7-H1/antagonistas & inhibidores , Glicoproteínas/fisiología , Inhibidores de Puntos de Control Inmunológico/farmacología , Melanoma Experimental/tratamiento farmacológico , Neovascularización Patológica/prevención & control , Microambiente Tumoral/inmunología , Animales , Antígeno B7-H1/inmunología , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
7.
Med Sci Monit ; 27: e931238, 2021 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-34210950

RESUMEN

BACKGROUND Oral squamous cell carcinoma (OSCC) is a common tumor of the head and neck. Its treatment usually requires multiple modalities. Currently, there are no molecular biomarkers to guide these treatment strategies. Studies have shown that microfibril-associated protein 4 (MFAP4) is potentially useful for non-invasive assessment of various diseases; however, its biological function in tumors is still unknown. In this study, we propose that MFAP4 is a new prognostic target for OSCC. MATERIAL AND METHODS First, we collected OSCC data (GSE25099 and GSE30784 datasets) from the Gene Expression Omnibus (GEO) database and compared the differential expression of MFAP4 gene between the patients (tumor) and normal (control) groups. The comparison was done with University of California Santa Cruz Xena (https://xenabrowser.net/Datapages/), and we calculated the difference in MFAP4 gene expression between normal and tumor tissues in a pan-cancer analysis. Then, we compared the 2 groups with high and low expression of MFAP4 gene in terms of tumor mutation burden (TMB), miRNA regulation, and immune cell infiltration. RESULTS We found that the expression of MFAP4 gene was significantly decreased in tumors. Our research also showed that high expression of MFAP4 was related to better prognosis of patients and may be related to tumor gene mutation, miRNA regulation, and infiltration of different immune cells. CONCLUSIONS Our work provides evidence that expression of MFAP4 can be used as a prognostic biomarker for risk stratification of OSCC patients and elaborates on its relation with the regulation of TMB, miRNAs, and immune cell infiltration.


Asunto(s)
Proteínas Portadoras/fisiología , Proteínas de la Matriz Extracelular/fisiología , Glicoproteínas/fisiología , Neoplasias de la Boca/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Biomarcadores de Tumor/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos
8.
Adv Sci (Weinh) ; 8(16): e2100841, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34184839

RESUMEN

Dry eye disease (DED) has high personal and societal costs, but its pathology remains elusive due to intertwined biophysical and biochemical processes at the ocular surface. Specifically, mucin deficiency is reported in a subset of DED patients, but its effects on ocular interfacial properties remain unclear. Herein a novel in vitro mucin-deficient mimetic ocular surface (Mu-DeMOS) with a controllable amount of membrane-tethered mucin molecules is developed to represent the diseased ocular surfaces. Contact angle goniometry on mimetic ocular surfaces reveals that high surface roughness, but not the presence of hydrophilic mucin molecules, delivers constant hydration over native ocular surface epithelia. Live-cell rheometry confirms that the presence of mucin-like glycoproteins on ocular epithelial cells reduces shear adhesive strength at cellular interfaces. Together, optimal surface roughness and surface chemistry facilitate sustainable lubrication for healthy ocular surfaces, while an imbalance between them contributes to lubrication-related dysfunction at diseased ocular epithelial surfaces. Furthermore, the restoration of low adhesive strength at Mu-DeMOS interfaces through a mucin-like glycoprotein, recombinant human lubricin, suggests that increased frictional damage at mucin-deficient cellular surfaces may be reversible. More broadly, these results demonstrate that Mu-DeMOS is a promising platform for drug screening assays and fundamental studies on ocular physiology.


Asunto(s)
Síndromes de Ojo Seco/fisiopatología , Células Epiteliales/fisiología , Glicoproteínas/fisiología , Mucinas/fisiología , Fenómenos Fisiológicos Oculares , Animales , Células Cultivadas , Humanos
9.
Hum Cell ; 34(5): 1517-1531, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34115333

RESUMEN

The dysregulated long non-coding RNA A1BG antisense RNA 1 (A1BG-AS1) has been implicated in the oncogenicity of hepatocellular carcinoma. Using reverse transcription quantitative polymerase chain reaction in this study, we detected A1BG-AS1 expression in breast cancer and elucidated the regulatory functions and exact mechanisms of A1BG-AS1 in breast cancer cells. The regulatory functions of A1BG-AS1 were examined in vitro using the Cell Counting Kit-8 assay, flow cytometric, and Transwell migration and invasion assays and in vivo through tumor xenograft experiments. In addition, we performed bioinformatics analysis, luciferase reporter assay, RNA immunoprecipitation, and rescue experiments to verify the interaction among A1BG-AS1, microRNA-485-5p (miR-485-5p), and flotillin-1 (FLOT1) in breast cancer. We found A1BG-AS1 to be highly expressed in breast cancer tissues and cell lines. In terms of function, depleted A1BG-AS1 markedly suppressed cell proliferation, accelerated cell apoptosis, and hindered cell migration and invasion in breast cancer. Furthermore, A1BG-AS1 interference reduced tumor growth in vivo. Mechanistic investigations confirmed that A1BG-AS1 directly interacted with miR-485-5p as a molecular sponge. We demonstrated that FLOT1 is a direct target of miR-485-5p, which could be positively regulated by A1BG-AS1 by competing for miR-485-5p. Rescue experiments clearly showed that the downregulation of miR-485-5p and upregulation of FLOT1 were capable of reversing the anticancer activities of A1BG-AS1 deficiency in terms of breast cancer cell malignancy. A1BG-AS1 acts as a miR-485-5p sponge and subsequently increases FLOT1 expression in breast cancer cells, ultimately facilitating cancer progression. Hence, the A1BG-AS1/miR-485-5p/FLOT1 pathway might offer a novel therapeutic perspective for breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Carcinogénesis/genética , Regulación Neoplásica de la Expresión Génica/genética , Glicoproteínas/fisiología , Inmunoglobulinas/fisiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , ARN Largo no Codificante/fisiología , Neoplasias de la Mama/terapia , Línea Celular Tumoral , Movimiento Celular/genética , Femenino , Glicoproteínas/genética , Humanos , Inmunoglobulinas/genética , Terapia Molecular Dirigida , Invasividad Neoplásica/genética , ARN Largo no Codificante/genética , Transducción de Señal/genética , Transducción de Señal/fisiología
10.
Sci Rep ; 11(1): 7581, 2021 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-33828185

RESUMEN

The basilar membrane (BM) of the mammalian cochlea constitutes a spiraling acellular ribbon that is intimately attached to the organ of Corti. Its graded stiffness, increasing from apex to the base of the cochlea provides the mechanical basis for sound frequency analysis. Despite its central role in auditory signal transduction, virtually nothing is known about the BM's structural development. Using polarized light microscopy, the present study characterized the architectural transformations of freshly dissected BM at time points during postnatal development and maturation. The results indicate that the BM structural elements increase progressively in size, becoming radially aligned and more tightly packed with maturation and reach the adult structural signature by postnatal day 20 (P20). The findings provide insight into structural details and developmental changes of the mammalian BM, suggesting that BM is a dynamic structure that changes throughout the life of an animal.


Asunto(s)
Membrana Basilar/anatomía & histología , Membrana Basilar/crecimiento & desarrollo , Animales , Membrana Basilar/fisiología , Birrefringencia , Glicoproteínas/deficiencia , Glicoproteínas/genética , Glicoproteínas/fisiología , Audición/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Sonido
11.
Am J Nephrol ; 52(3): 228-238, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33823527

RESUMEN

INTRODUCTION: Increasing evidence has demonstrated that loss of peritubular capillaries plays a critical role in renal interstitial fibrosis. Leucine-rich α2-glycoprotein-1 (LRG1) has been observed promoting angiogenesis in the ocular disease mouse model and myocardial infarction model. We aimed to explore the role of LRG1 in renal interstitial fibrosis. METHODS: We analyzed the expression of LRG1 in the plasma and kidney of CKD patients by ELISA and immunohistochemistry. Relationships between the expression of LRG1 in plasma and kidney and renal fibrosis and inflammation were analyzed. Tube formation assay was used to detect the angiogenesis in the human umbilical vein endothelial cell lines (HUVECs). And real-time PCR was used to detect the mRNA expression of LRG1, inflammatory factors, renal tubular injury indicators, pro-fibrotic cytokines, and CD31. We examined the effects of genetic ablation of LRG1 on renal fibrosis induced by unilateral ureteral obstruction (UUO) mice model at day 7. RESULTS: We demonstrated that the expression of LRG1 in renal tissues and plasma samples was upregulated in CKD patients. And the expression of LRG1 was elevated in human renal tubular epithelial cell line (HK-2) cells in response to the stimulation of TNF-α in vitro, and in kidney after UUO in vivo. The deficiency of the LRG1 gene aggravated renal fibrosis, inflammatory cells infiltration, and capillary rarefaction after UUO. In vitro, LRG1 promoted the tube formation of HUVEC cells. LRG1 inhibits fibronectin secretion induced by TGF-ß1 in HK-2 and overexpression of LRG1 in HK-2 cells decreased fibronectin secretion. CONCLUSION: LRG1 may prevent renal fibrosis by inhibiting the secretion of inflammatory and pro-fibrotic cytokines and promoting angiogenesis.


Asunto(s)
Citocinas/fisiología , Glicoproteínas/fisiología , Enfermedades Renales/etiología , Riñón/patología , Rarefacción Microvascular/etiología , Adulto , Animales , Femenino , Fibrosis/etiología , Humanos , Masculino , Ratones , Persona de Mediana Edad
12.
Mol Immunol ; 130: 159-163, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33384157

RESUMEN

The CD1 antigen presenting system is evolutionary conserved and found in mammals, birds and reptiles. Humans express five isoforms, of which CD1a, CD1b and CD1c represent the group 1 CD1-molecules. They are recognized by T cells that express diverse αß-T cell receptors. Investigation of the role of group 1 CD1 function has been hampered by the fact that CD1a, CD1b and CD1c are not expressed by mice. However, other animals, such as guinea pigs or cattle, serve as alternative models and have established basic aspects of CD1-dependent, antimicrobial immune functions. Group 1 CD1 transgenic mouse models became available about ten years ago. In a series of seminal studies these mouse models coined the mechanistical understanding of the role of the corresponding CD1 restricted T cell responses. This review gives a short overview of available animal studies and the lessons that have been and still can be learned.


Asunto(s)
Presentación de Antígeno , Antígenos CD1/fisiología , Modelos Animales , Animales , Presentación de Antígeno/genética , Presentación de Antígeno/inmunología , Bovinos , Glicoproteínas/fisiología , Cobayas , Humanos , Ratones , Linfocitos T/inmunología , Linfocitos T/metabolismo
13.
Front Immunol ; 12: 784473, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34975875

RESUMEN

Lectin-glycan interactions, in particular those mediated by the galectin family, regulate many processes required for a successful pregnancy. Over the past decades, increasing evidence gathered from in vitro and in vivo experiments indicate that members of the galectin family specifically bind to both intracellular and membrane bound carbohydrate ligands regulating angiogenesis, immune-cell adaptations required to tolerate the fetal semi-allograft and mammalian embryogenesis. Therefore, galectins play important roles in fetal development and placentation contributing to maternal and fetal health. This review discusses the expression and role of galectins during the course of pregnancy, with an emphasis on maternal immune adaptions and galectin-glycan interactions uncovered in the recent years. In addition, we summarize the galectin fingerprints associated with pathological gestation with particular focus on preeclampsia.


Asunto(s)
Adaptación Fisiológica , Desarrollo Fetal/fisiología , Galectinas/fisiología , Placentación/fisiología , Femenino , Galectinas/química , Glicoproteínas/fisiología , Humanos , Embarazo
14.
Med Mycol ; 59(5): 441-452, 2021 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-32766889

RESUMEN

The genus Scedosporium is composed of clinically relevant fungal species, such as Scedosporium aurantiacum, Scedosporium apiospermum, and Scedosporium boydii. Surface molecules have been described that play crucial roles in fungi-macrophage interaction, and many of them are pathogen-associated molecular patterns (PAMPs). The present study aims to characterize peptidoglycans obtained from Scedosporium aurantiacum and Scedosporium minutisporum, a clinical and an environmental isolate, respectively, and compare their roles in pathogen-host interaction. Both molecules were characterized as peptidorhamnomannans (PRMs), similar to what has been already described for other Scedosporium species. Rabbit immune sera obtained by injecting whole cells from each species recognized both fungal cells and purified PRMs, suggesting that a cross-reaction occur between both fungi. Immunofluorescent microscopy revealed that PRMs are exposed on fungal surface. Prior incubation of purified molecules with immune sera before adding to cells led to loss of fluorescent, indicating that PRM is a major molecule recognized by immune sera. Fungi-macrophage interaction revealed that S. aurantiacum is able to survive more inside phagocytic cells than S. minutisporum, and PRM from both fungi plays a role in phagocytosis when the purified molecule is pre-incubated with macrophage. In addition, PRM induce nitric oxide release by macrophages. Our data indicate that PRM is an important PAMP exposed on fungal surface with the potential of immune modulation.


In this work, peptidorhamnomannans from Scedosporium aurantiacum and Scedosporium minutisporum have been characterized. These molecules play important roles in phagocytosis and oxidative burst in peritoneal macrophages and are recognized by immune sera.


Asunto(s)
Glicoproteínas/química , Glicoproteínas/fisiología , Macrófagos/metabolismo , Macrófagos/microbiología , Scedosporium/metabolismo , Animales , Anticuerpos Antifúngicos/química , Anticuerpos Antifúngicos/inmunología , Femenino , Interacciones Microbiota-Huesped , Humanos , Infecciones Fúngicas Invasoras/inmunología , Infecciones Fúngicas Invasoras/microbiología , Ratones , Ratones Endogámicos BALB C , Óxido Nítrico/metabolismo , Moléculas de Patrón Molecular Asociado a Patógenos/metabolismo , Fagocitosis , Conejos
15.
J Biol Chem ; 296: 100070, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33187980

RESUMEN

Lipids in complex, protein-enriched films at air/liquid interfaces reduce surface tension. In the absence of this benefit, the light refracting and immunoprotective tear film on eyes would collapse. Premature collapse, coupled with chronic inflammation compromising visual acuity, is a hallmark of dry eye disease affecting 7 to 10% of individuals worldwide. Although collapse seems independent of mutation (unlike newborn lung alveoli), selective proteome and possible lipidome changes have been noted. These include elevated tissue transglutaminase and consequent inactivation through C-terminal cross-linking of the tear mitogen lacritin, leading to significant loss of lacritin monomer. Lacritin monomer restores homeostasis via autophagy and mitochondrial fusion and promotes basal tearing. Here, we discover that lacritin monomer C-terminal processing, inclusive of cysteine, serine, and metalloproteinase activity, generates cationic amphipathic α-helical proteoforms. Such proteoforms (using synthetic peptide surrogates) act like alveolar surfactant proteins to rapidly bind and stabilize the tear lipid layer. Immunodepletion of C- but not N-terminal proteoforms nor intact lacritin, from normal human tears promotes loss of stability akin to human dry eye tears. Stability of these and dry eye tears is rescuable with C- but not N-terminal proteoforms. Repeated topical application in rabbits reveals a proteoform turnover time of 7 to 33 h with gradual loss from human tear lipid that retains bioactivity without further processing. Thus, the processed C-terminus of lacritin that is deficient or absent in dry eye tears appears to play a key role in preventing tear film collapse and as a natural slow release mechanism that restores epithelial homeostasis.


Asunto(s)
Síndromes de Ojo Seco/fisiopatología , Proteínas del Ojo/metabolismo , Glicoproteínas/fisiología , Isoformas de Proteínas/fisiología , Lágrimas/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Glándulas Tarsales/fisiología , Conejos
16.
Hum Mol Genet ; 29(21): 3554-3565, 2021 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-33219378

RESUMEN

The glycogenin knockout mouse is a model of Glycogen Storage Disease type XV. These animals show high perinatal mortality (90%) due to respiratory failure. The lungs of glycogenin-deficient embryos and P0 mice have a lower glycogen content than that of wild-type counterparts. Embryonic lungs were found to have decreased levels of mature surfactant proteins SP-B and SP-C, together with incomplete processing of precursors. Furthermore, non-surviving pups showed collapsed sacculi, which may be linked to a significantly reduced amount of surfactant proteins. A similar pattern was observed in glycogen synthase1-deficient mice, which are devoid of glycogen in the lungs and are also affected by high perinatal mortality due to atelectasis. These results indicate that glycogen availability is a key factor for the burst of surfactant production required to ensure correct lung expansion at the establishment of air breathing. Our findings confirm that glycogen deficiency in lungs can cause respiratory distress syndrome and suggest that mutations in glycogenin and glycogen synthase 1 genes may underlie cases of idiopathic neonatal death.


Asunto(s)
Glucosiltransferasas/fisiología , Glucógeno Sintasa/fisiología , Glicoproteínas/fisiología , Surfactantes Pulmonares/metabolismo , Síndrome de Dificultad Respiratoria/patología , Animales , Animales Recién Nacidos , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Síndrome de Dificultad Respiratoria/etiología , Síndrome de Dificultad Respiratoria/metabolismo
17.
STAR Protoc ; 1(3): 100133, 2020 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-33377027

RESUMEN

HIV-1 envelope glycoproteins (Envs) bind to CD4 receptor and CCR5/CXCR4 coreceptor and mediate viral entry (Feng et al., 1996; Herschhorn et al., 2016, 2017; Kwong et al., 1998). HIV-1 Envs are the sole target of neutralizing antibodies and a main focus of vaccine development (Flemming et al., 2018). Here, we provide a step-by-step protocol to measure Env sensitivity to ligands, cold, and small molecules, as well as to study viral infectivity and to dissect parameters affecting HIV-1 Env function. For complete details on the use and execution of this protocol, please refer to Harris et al. (2020).


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Productos del Gen env del Virus de la Inmunodeficiencia Humana/efectos de los fármacos , Productos del Gen env del Virus de la Inmunodeficiencia Humana/aislamiento & purificación , Anticuerpos Neutralizantes/inmunología , Antígenos CD4/metabolismo , Genes env/genética , Glicoproteínas/efectos de los fármacos , Glicoproteínas/aislamiento & purificación , Glicoproteínas/fisiología , Anticuerpos Anti-VIH/inmunología , VIH-1/genética , VIH-1/metabolismo , Humanos , Ligandos , Receptores CCR5/metabolismo , Internalización del Virus , Productos del Gen env del Virus de la Inmunodeficiencia Humana/metabolismo
18.
Exp Hematol ; 91: 32-38.e6, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33075436

RESUMEN

The generation of a functional erythrocyte from a committed progenitor requires significant changes in gene expression during hemoglobin accumulation, rapid cell division, and nuclear condensation. Congenital dyserythropoietic anemia type I (CDA-I) is an autosomal recessive disease that presents with erythroid hyperplasia in the bone marrow. Erythroblasts in patients with CDA-I are frequently binucleate and have chromatin bridging and defective chromatin condensation. CDA-1 is most commonly caused by mutations in Codanin-1 (CDAN1). The function of CDAN1 is poorly understood but it is thought to regulate histone incorporation into nascent DNA during cellular replication. The study of CDA-1 has been limited by the lack of in vitro models that recapitulate key features of the disease, and most studies on CDAN1 function have been done in nonerythroid cells. To model CDA-I we generated HUDEP2 mutant lines with deletion or mutation of R1042 of CDAN1, mirroring mutations found in CDA-1 patients. CDAN1 mutant cell lines had decreased viability and increased intercellular bridges and binucleate cells. Further, they had alterations in histone acetylation associated with prematurely elevated erythroid gene expression, including gamma globin. Together, these data imply a specific functional role for CDAN1, specifically R1042 on exon 24, in the regulation of DNA replication and organization during erythroid maturation. Most importantly, generation of models with specific patient mutations, such as R1042, will provide further mechanistic insights into CDA-I pathology.


Asunto(s)
Anemia Diseritropoyética Congénita/genética , Células Eritroides/citología , Eritropoyesis/genética , Glicoproteínas/genética , Proteínas Nucleares/genética , Acetilación , Anemia Diseritropoyética Congénita/sangre , Sistemas CRISPR-Cas , Línea Celular , Núcleo Celular/ultraestructura , Supervivencia Celular , Cromatina/ultraestructura , Células Eritroides/metabolismo , Eritropoyesis/fisiología , Exones/genética , Edición Génica , Glicoproteínas/deficiencia , Glicoproteínas/fisiología , Código de Histonas , Humanos , Proteínas Nucleares/deficiencia , Proteínas Nucleares/fisiología , Fenotipo , Procesamiento Proteico-Postraduccional
19.
FASEB J ; 34(11): 15559-15576, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32997357

RESUMEN

Pericytes promote vessel stability and their dysfunction causes pathologies due to blood vessel leakage. Previously, we reported that Olfactomedin-like 3 (Olfml3) is a matricellular protein with proangiogenic properties. Here, we explored the role of Olfml3 in a knockout mouse model engineered to suppress this protein. The mutant mice exhibited vascular defects in pericyte coverage, suggesting that pericytes influence blood vessel formation in an Olfml3-dependent manner. Olfml3-deficient mice exhibited abnormalities in the vasculature causing partial lethality of embryos and neonates. Reduced pericyte coverage was observed at embryonic day 12.5 and persisted throughout development, resulting in perinatal death of 35% of Olfml3-deficient mice. Cultured Olfml3-deficient pericytes exhibited aberrant motility and altered pericyte association to endothelial cells. Furthermore, the proliferative response of Olfml3-/- pericytes upon PDGF-B stimulation was significantly diminished. Subsequent experiments revealed that intact PDGF-B signaling, mediated via Olfml3 binding, is required for pericyte proliferation and activation of downstream kinase pathways. Our findings suggest a model wherein pericyte recruitment to endothelial cells requires Olfml3 to provide early instructive cue and retain PDGF-B along newly formed vessels to achieve optimal angiogenesis.


Asunto(s)
Movimiento Celular , Proliferación Celular , Glicoproteínas/fisiología , Neovascularización Patológica/patología , Pericitos/patología , Proteínas Proto-Oncogénicas c-sis/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Animales , Desarrollo Embrionario , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Patológica/metabolismo , Pericitos/metabolismo , Embarazo , Transducción de Señal
20.
FASEB J ; 34(11): 14250-14263, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32905637

RESUMEN

Renal fibrosis, which is characterized by excessive extracellular matrix (ECM) accumulation in the renal tubulointerstitium, can lead to chronic kidney disease (CKD). The role of microfiber-associated protein 4 (MFAP4), which is an ECM protein that interacts with elastin and collagen, in renal fibrosis has not been investigated. The aim of this study was to examine the role of MFAP4 in the pathogenesis of renal fibrosis and the underlying mechanism using in vivo and in vitro models. The MFAP4-/- mice were subjected to unilateral ureteral obstruction (UUO) to elucidate the role of MFAP4 in renal fibrosis in vivo. Compared to the wild-type mice, the MFAP4-/- mice exhibited decreased protein expression of p-p65 and p-IKBα and ECM deposition after UUO. The MFAP4-/- mice exhibited attenuated nuclear translocation of p65 (the hub subunit of nuclear factor (NF)-κB signaling pathway), suppressed activation of transforming growth factor (TGF)-ß/Smad pathways, and downregulated expression of fibronectin, collagen I, and plasminogen activator inhibitor-1. The knockdown of MFAP4 mitigated the TGF-ß-induced upregulated expression of fibronectin, collagen I, and plasminogen activator inhibitor-1 in the human proximal tubular epithelial cells (HK-2). Compared to the HK-2 cells transfected with sh-MFAP4, the HK-2 cells co-transfected with sh-MFAP4 and Ad-MFAP4 exhibited severe inflammatory response and increased fibrosis-related proteins expression. Mechanistically, the knockdown of MFAP4 inhibited the activation of NF-κB and TGF-ß/Smad signaling pathways and downregulated the expression of fibrosis-related proteins. The findings of this study indicate that MFAP4 is involved in UUO-induced renal fibrosis through regulation of NF-κB and TGF-ß/Smad pathways.


Asunto(s)
Proteínas Portadoras/fisiología , Proteínas de la Matriz Extracelular/fisiología , Fibrosis/prevención & control , Glicoproteínas/fisiología , Enfermedades Renales/prevención & control , FN-kappa B/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Obstrucción Ureteral/complicaciones , Animales , Modelos Animales de Enfermedad , Fibronectinas/genética , Fibronectinas/metabolismo , Fibrosis/etiología , Fibrosis/metabolismo , Fibrosis/patología , Enfermedades Renales/etiología , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/genética , FN-kappa B/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...