Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.370
Filtrar
1.
Physiol Plant ; 176(3): e14375, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38837224

RESUMEN

MicroRNA(miRNA) is a class of non-coding small RNA that plays an important role in plant growth, development, and response to environmental stresses. Unlike most miRNAs, which usually target homologous genes across a variety of species, miR827 targets different types of genes in different species. Research on miR827 mainly focuses on its role in regulating phosphate (Pi) homeostasis of plants, however, little is known about its function in plant response to virus infection. In the present study, miR827 was significantly upregulated in the recovery tissue of virus-infected Nicotiana tabacum. Overexpression of miR827 could improve plants resistance to the infection of chilli veinal mottle virus (ChiVMV) in Nicotiana benthamiana, whereas interference of miR827 increased the susceptibility of the virus-infected plants. Further experiments indicated that the antiviral defence regulated by miR827 was associated with the reactive oxygen species and salicylic acid signalling pathways. Then, fructose-1,6-bisphosphatase (FBPase) was identified to be a target of miR827, and virus infection could affect the expression of FBPase. Finally, transient expression of FBPase increased the susceptibility to ChiVMV-GFP infection in N. benthamiana. By contrast, silencing of FBPase increased plant resistance. Taken together, our results demonstrate that miR827 plays a positive role in tobacco response to virus infection, thus providing new insights into understanding the role of miR827 in plant-virus interaction.


Asunto(s)
Resistencia a la Enfermedad , Regulación de la Expresión Génica de las Plantas , MicroARNs , Nicotiana , Enfermedades de las Plantas , Nicotiana/virología , Nicotiana/genética , MicroARNs/genética , MicroARNs/metabolismo , Enfermedades de las Plantas/virología , Enfermedades de las Plantas/genética , Enfermedades de las Plantas/inmunología , Resistencia a la Enfermedad/genética , Fructosa-Bifosfatasa/genética , Fructosa-Bifosfatasa/metabolismo , Ácido Salicílico/metabolismo , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Tobamovirus/fisiología , Tobamovirus/genética , Plantas Modificadas Genéticamente
2.
Cell Death Dis ; 15(6): 392, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38834617

RESUMEN

Keratinocyte proliferation and differentiation in epidermis are well-controlled and essential for reacting to stimuli such as ultraviolet light. Imbalance between proliferation and differentiation is a characteristic feature of major human skin diseases such as psoriasis and squamous cell carcinoma. However, the effect of keratinocyte metabolism on proliferation and differentiation remains largely elusive. We show here that the gluconeogenic enzyme fructose-1,6-bisphosphatase 1 (FBP1) promotes differentiation while inhibits proliferation of keratinocyte and suppresses psoriasis development. FBP1 is identified among the most upregulated genes induced by UVB using transcriptome sequencing and is elevated especially in upper epidermis. Fbp1 heterozygous mice exhibit aberrant epidermis phenotypes with local hyperplasia and dedifferentiation. Loss of FBP1 promotes proliferation and inhibits differentiation of keratinocytes in vitro. Mechanistically, FBP1 loss facilitates glycolysis-mediated acetyl-CoA production, which increases histone H3 acetylation at lysine 9, resulting in enhanced transcription of proliferation genes. We further find that the expression of FBP1 is dramatically reduced in human psoriatic lesions and in skin of mouse imiquimod psoriasis model. Fbp1 deficiency in mice facilitates psoriasis-like skin lesions development through glycolysis and acetyl-CoA production. Collectively, our findings reveal a previously unrecognized role of FBP1 in epidermal homeostasis and provide evidence for FBP1 as a metabolic psoriasis suppressor.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Fructosa-Bifosfatasa , Histonas , Queratinocitos , Psoriasis , Psoriasis/patología , Psoriasis/metabolismo , Psoriasis/genética , Animales , Queratinocitos/metabolismo , Queratinocitos/patología , Humanos , Acetilación , Histonas/metabolismo , Fructosa-Bifosfatasa/metabolismo , Fructosa-Bifosfatasa/genética , Ratones , Glucólisis , Ratones Endogámicos C57BL , Acetilcoenzima A/metabolismo , Modelos Animales de Enfermedad
3.
Sheng Wu Gong Cheng Xue Bao ; 40(5): 1559-1570, 2024 May 25.
Artículo en Chino | MEDLINE | ID: mdl-38783816

RESUMEN

To develop an accurate and efficient protocol for multi-fragment assembly and multi-site mutagenesis, we integrated and optimized the common multi-fragment assembly methods and validated the established method by using fructose-1,6-diphosphatase 1 (FBP1) with 4 mutant sites. The fragments containing mutations were assembled by introducing mutant sites and Bsa I recognition sequences. After digestion/ligation, the ligated fragment was amplified with the primers containing overlap region to the linearized vector. The amplified fragment was ligated to the linearized vector and the ligation product was transformed into Escherichia coli. After screening and sequencing, the recombinant plasmid with 4 mutant sites was obtained. This protocol overcame the major defects of Gibson assembly and Golden Gate assembly, serving as an efficient solution for multi-fragment assembly and multi-site mutagenesis.


Asunto(s)
Escherichia coli , Fructosa-Bifosfatasa , Recombinación Homóloga , Escherichia coli/genética , Escherichia coli/metabolismo , Fructosa-Bifosfatasa/genética , Fructosa-Bifosfatasa/metabolismo , Plásmidos/genética , Vectores Genéticos/metabolismo , ADN/genética , Mutación , Mutagénesis Sitio-Dirigida , Clonación Molecular
5.
J Med Case Rep ; 18(1): 166, 2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38589931

RESUMEN

BACKGROUND: Fructose-1,6-bisphosphatase deficiency is a rare autosomal recessive disorder characterized by impaired gluconeogenesis. Fructose-1,6-bisphosphatase 1 (FBP1) mutations demonstrate ethnic patterns. For instance, Turkish populations commonly harbor exon 2 deletions. We present a case report of whole exon 2 deletion in a Syrian Arabian child as the first recording of this mutation among Arabian ethnicity and the first report of FBP1 gene mutation in Syria. CASE PRESENTATION: We present the case of a 2.5-year-old Syrian Arab child with recurrent hypoglycemic episodes, accompanied by nausea and lethargy. The patient's history, physical examination, and laboratory findings raised suspicion of fructose-1,6-bisphosphatase deficiency. Whole exome sequencing was performed, revealing a homozygous deletion of exon 2 in the FBP1 gene, confirming the diagnosis. CONCLUSION: This case highlights a potential novel mutation in the Arab population; this mutation is well described in the Turkish population, which suggests potential shared mutations due to ancestral relationships between the two ethnicities. Further studies are needed to confirm this finding.


Asunto(s)
Deficiencia de Fructosa-1,6-Difosfatasa , Preescolar , Humanos , Documentación , Etnicidad , Fructosa , Deficiencia de Fructosa-1,6-Difosfatasa/complicaciones , Deficiencia de Fructosa-1,6-Difosfatasa/diagnóstico , Deficiencia de Fructosa-1,6-Difosfatasa/genética , Fructosa-Bifosfatasa/genética , Homocigoto , Mutación , Eliminación de Secuencia
6.
Funct Plant Biol ; 51: FP24034, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38640358

RESUMEN

Transgenic Arabidopsis thaliana (ecotype Columbia) was successfully transformed with the gene fructose-1,6-bisphosphatase (FBPas e) and named as AtFBPase plants. Transgenic plants exhibited stable transformation, integration and significantly higher expressions for the transformed gene. Morphological evaluation of transgenic plants showed increased plant height (35cm), number of leaves (25), chlorophyll contents (28%), water use efficiency (increased from 1.5 to 2.6µmol CO2 µmol-1 H2 O) and stomatal conductance (20%), which all resulted in an enhanced photosynthetic rate (2.7µmolm-2 s-1 ) compared to wild type plants. This study suggests the vital role of FBPase gene in the modification of regulatory pathways to enhance the photosynthetic rate, which can also be utilised for economic crops in future.


Asunto(s)
Arabidopsis , Arabidopsis/genética , Fructosa-Bifosfatasa/genética , Fructosa-Bifosfatasa/metabolismo , Fructosa/metabolismo , Fotosíntesis/genética , Clorofila/genética , Clorofila/metabolismo , Plantas Modificadas Genéticamente/genética , Plantas Modificadas Genéticamente/metabolismo
7.
Plant Cell Physiol ; 65(5): 737-747, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38305687

RESUMEN

Various chloroplast proteins are activated/deactivated during the light/dark cycle via the redox regulation system. Although the photosynthetic electron transport chain provides reducing power to redox-sensitive proteins via the ferredoxin (Fd)/thioredoxin (Trx) pathway for their enzymatic activity control, how the redox states of individual proteins are linked to electron transport efficiency remains uncharacterized. Here we addressed this subject with a focus on the photosynthetic induction phase. We used Arabidopsis plants, in which the amount of Fd-Trx reductase (FTR), a core component in the Fd/Trx pathway, was genetically altered. Several chloroplast proteins showed different redox shift responses toward low- and high-light treatments. The light-dependent reduction of Calvin-Benson cycle enzymes fructose 1,6-bisphosphatase (FBPase) and sedoheptulose 1,7-bisphosphatase (SBPase) was partially impaired in the FTR-knockdown ftrb mutant. Simultaneous analyses of chlorophyll fluorescence and P700 absorbance change indicated that the induction of the electron transport reactions was delayed in the ftrb mutant. FTR overexpression also mildly affected the reduction patterns of FBPase and SBPase under high-light conditions, which were accompanied by the modification of electron transport properties. Accordingly, the redox states of FBPase and SBPase were linearly correlated with electron transport rates. In contrast, ATP synthase was highly reduced even when electron transport reactions were not fully induced. Furthermore, the redox response of proton gradient regulation 5-like photosynthetic phenotype1 (PGRL1; a protein involved in cyclic electron transport) did not correlate with electron transport rates. Our results provide insights into the working dynamics of the redox regulation system and their differential associations with photosynthetic electron transport efficiency.


Asunto(s)
Proteínas de Arabidopsis , Arabidopsis , Oxidación-Reducción , Fotosíntesis , Transporte de Electrón , Arabidopsis/metabolismo , Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Fructosa-Bifosfatasa/metabolismo , Fructosa-Bifosfatasa/genética , Luz , Cloroplastos/metabolismo , Clorofila/metabolismo , Proteínas de Cloroplastos/metabolismo , Proteínas de Cloroplastos/genética , Oxidorreductasas/metabolismo , Oxidorreductasas/genética , Proteínas Hierro-Azufre , Monoéster Fosfórico Hidrolasas
8.
PLoS One ; 19(1): e0294191, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38252660

RESUMEN

Meiotic recombination is a pivotal process that ensures faithful chromosome segregation and contributes to the generation of genetic diversity in offspring, which is initiated by the formation of double-strand breaks (DSBs). The distribution of meiotic DSBs is not uniform and is clustered at hotspots, which can be affected by environmental conditions. Here, we show that non-coding RNA (ncRNA) transcription creates meiotic DSBs through local chromatin remodeling in the fission yeast fbp1 gene. The fbp1 gene is activated upon glucose starvation stress, in which a cascade of ncRNA-transcription in the fbp1 upstream region converts the chromatin configuration into an open structure, leading to the subsequent binding of transcription factors. We examined the distribution of meiotic DSBs around the fbp1 upstream region in the presence and absence of glucose and observed several new DSBs after chromatin conversion under glucose starvation conditions. Moreover, these DSBs disappeared when cis-elements required for ncRNA transcription were mutated. These results indicate that ncRNA transcription creates meiotic DSBs in response to stress conditions in the fbp1 upstream region. This study addressed part of a long-standing unresolved mechanism underlying meiotic recombination plasticity in response to environmental fluctuation.


Asunto(s)
ARN Largo no Codificante , Schizosaccharomyces , Inanición , Humanos , Schizosaccharomyces/genética , ADN , Cromatina , Fructosa-Bifosfatasa/genética , Glucosa , Roturas del ADN
9.
Mol Biol Rep ; 51(1): 78, 2024 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-38183507

RESUMEN

BACKGROUND: Aberrant DNA methylation has been implicated in the development of gastric cancer (GC). In our previous study, we demonstrated that fructose-1,6-bisphosphatase-2 (FBP2), an enzyme that suppresses cell glycolysis and growth, is downregulated in GC due to promoter methylation. However, the precise mechanism underlying this process remains unknown. Thus, this study aimed to elucidate the mechanisms involved in FBP2 promoter hypermethylation. METHODS AND RESULTS: The methylation levels in GC and normal adjacent tissues were quantified using methylation-specific polymerase chain reaction. FBP2 promoter was frequently hypermethylated in primary GC tissues compared to adjacent normal tissues. To explore the functional consequences of this hypermethylation, we employed small interfering RNA-mediated knockdown of DNA methyltransferase 3a (DNMT3a) in GC cells. FBP2 expression increased following DNMT3a knockdown, suggesting that reduced methylation of the FBP2 promoter contributed to this upregulation. To further investigate this interaction, chromatin immunoprecipitation assays were conducted. The results confirmed an interaction between DNMT3a and the FBP2 promoter region, providing evidence that DNMT3a-mediated hypermethylation of the FBP2 promoter promotes GC progression. CONCLUSIONS: This study provides evidence that DNMT3a is involved in the hypermethylation of the FBP2 promoter and regulation of GC cell metabolism. Hypermethylation of the FBP2 promoter may be a promising prognostic biomarker in GC.


Asunto(s)
Metilación de ADN , Neoplasias Gástricas , Humanos , Carcinogénesis , Metilación de ADN/genética , ADN Metiltransferasa 3A , Metilasas de Modificación del ADN , Fructosa , Fructosa-Bifosfatasa/genética , Regiones Promotoras Genéticas/genética , Neoplasias Gástricas/genética
10.
Mol Genet Genomic Med ; 12(1): e2339, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38111981

RESUMEN

BACKGROUND: Fructose-1,6-bisphosphatase (FBPase) deficiency, caused by an FBP1 mutation, is an autosomal recessively inherited metabolic disorder characterized by impaired gluconeogenesis. Due to the rarity of FBPase deficiency, the mechanism by which the mutations cause enzyme activity loss still remains unclear. METHODS: We report a pediatric patient with typical FBPase deficiency who presented with hypoglycemia, hyperlactatemia, metabolic acidosis, and hyperuricemia. Whole-exome sequencing was used to search for pathogenic genes, Sanger sequencing was used for verification, and molecular dynamic simulation was used to evaluate how the novel mutation affects FBPase activity and structural stability. RESULTS: Direct and allele-specific sequence analysis of the FBP1 gene (NM_000507) revealed that the proband had a compound heterozygote for the c. 490 (exon 4) G>A (p. G164S) and c. 861 (exon 7) C>A (p. Y287X, 52), which he inherited from his carrier parents. His father and mother had heterozygous G164S and Y287X mutations, respectively, without any symptoms of hypoglycemia. CONCLUSION: Our results broaden the known mutational spectrum and possible clinical phenotype of FBP1.


Asunto(s)
Acidosis Láctica , Deficiencia de Fructosa-1,6-Difosfatasa , Hipoglucemia , Masculino , Humanos , Niño , Acidosis Láctica/genética , Deficiencia de Fructosa-1,6-Difosfatasa/diagnóstico , Deficiencia de Fructosa-1,6-Difosfatasa/genética , Fructosa-Bifosfatasa/genética , Fructosa-Bifosfatasa/metabolismo , Hipoglucemia/genética , Mutación
11.
Arthritis Res Ther ; 25(1): 235, 2023 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-38049890

RESUMEN

PURPOSE: To identify the role of gluconeogenesis in chondrocytes in osteoarthritis (OA). MATERIALS AND METHODS: Cartilage samples were collected from OA patients and C57 mice and were stained with Safranin O-Fast Green to determine the severity of OA. Periodic acid Schiff staining was used to characterize the contents of polysaccharides and SA-ßGal staining was used to characterize the aging of chondrocytes. Immunohistochemistry and western blotting were used to detect fructose-bisphosphatase1 (FBP1), SOX9, MMP13, P21, and P16 in cartilage or chondrocyte. The mRNA levels of fbp1, mmp13, sox9, colX, and acan were analyzed by qPCR to evaluate the role of FBP1 in chondrocytes. RESULTS: The level of polysaccharides in cartilage was reduced in OA and the expression of FBP1 was also reduced. We treated the chondrocytes with IL-1ß to cause OA in vitro, and then made chondrocytes overexpress FBP1 with plasma. It shows that FBP1 alleviated the degeneration and senescence of chondrocytes in vitro and that it also showed the same effects in vivo experiments. To further understand the mechanism of FBP1, we screened the downstream protein of FBP1 and found that CRB3 was significantly downregulated. And we confirmed that CRB3 suppressed the degeneration and delayed senescence of chondrocytes. CONCLUSIONS: FBP1 promoted the polysaccharide synthesis in cartilage and alleviated the degeneration of cartilage by regulating CRB3, so FBP1 is a potential target in treating OA.


Asunto(s)
Cartílago Articular , Fructosa-Bifosfatasa , Glicoproteínas de Membrana , Osteoartritis , Animales , Humanos , Ratones , Cartílago Articular/metabolismo , Condrocitos/metabolismo , Interleucina-1beta/metabolismo , Metaloproteinasa 13 de la Matriz/metabolismo , Osteoartritis/metabolismo , Polisacáridos/metabolismo , Fructosa-Bifosfatasa/metabolismo , Glicoproteínas de Membrana/metabolismo
12.
Theranostics ; 13(15): 5290-5304, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37908738

RESUMEN

Background: Chronic inflammation caused by immune cells is the central link between obesity and insulin resistance. Targeting the inflammatory process is a highly promising method for reversing systemic insulin resistance. Methods: Blood samples were prospectively collected from 68 patients with type 2 diabetes. C57BL/6J mice were fed either a high-fat diet (HFD) or normal chow (NC). We performed phenotypical and functional analyses of immune cells using flow cytometry. Vitamin D receptor (VDR) knockout γδ T cells were constructed using Cas9-gRNA targeted approaches to identify 1α,25(OH)2D3/VDR signaling pathway-mediated transcriptional regulation of fructose-1,6-bisphosphatase (FBP1) in γδ T cells. Results: Serum vitamin D deficiency aggravates inflammation in circulating γδ T cells in type 2 diabetes patients. We defined a critical role for 1α,25(OH)2D3 in regulating glycolysis metabolism, protecting against inflammation, and alleviating insulin resistance. Mechanistically, 1α,25(OH)2D3-VDR promoted FBP1 expression to suppress glycolysis in γδ T cells, thereby inhibiting Akt/p38 MAPK phosphorylation and reducing inflammatory cytokine production. Notably, therapeutic administration of 1α,25(OH)2D3 restrained inflammation in γδ T cells and ameliorated systemic insulin resistance in obese mice. Conclusions: Collectively, these findings show that 1α,25(OH)2D3 plays an important role in maintaining γδ T cell homeostasis by orchestrating metabolic programs, and is a highly promising target for preventing obesity, inflammation, and insulin resistance.


Asunto(s)
Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Animales , Humanos , Ratones , Calcitriol , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Fructosa-Bifosfatasa , Inflamación , Ratones Endogámicos C57BL , Obesidad , Linfocitos T
13.
Am J Physiol Cell Physiol ; 325(5): C1354-C1368, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37781737

RESUMEN

Glomerular angiogenesis is a characteristic feature of diabetic nephropathy (DN). Enhanced glycolysis plays a crucial role in angiogenesis. The present study was designed to investigate the role of glycolysis in glomerular endothelial cells (GECs) in a mouse model of DN. Mouse renal cortex and isolated glomerular cells were collected for single-cell and RNA sequencing. Cultured GECs were exposed to high glucose in the presence (proangiogenic) and absence of a vascular sprouting regimen. MicroRNA-590-3p was delivered by lipofectamine in vivo and in vitro. In the present study, a subgroup of GECs with proangiogenic features was identified in diabetic kidneys by using sequencing analyses. In cultured proangiogenic GECs, high glucose increased glycolysis and phosphofructokinase/fructose bisphosphatase 3 (PFKFB3) protein expression, which were inhibited by overexpressing miRNA-590-3p. Mimics of miRNA-590-3p also increased receptor for sphingosine 1-phosphate (S1pR1) expression, an angiogenesis regulator, in proangiogenic GECs challenged with high glucose. Inhibition of PFKFB3 by pharmacological and genetic approaches upregulated S1pR1 protein in vitro. Mimics of miRNA-590-3p significantly reduced migration and angiogenic potential in proangiogenic GECs challenged with high glucose. Ten-week-old type 2 diabetic mice had elevated urinary albumin levels, reduced renal cortex miRNA-590-3p expression, and disarrangement of glomerular endothelial cell fenestration. Overexpressing miRNA-590-3p via perirenal adipose tissue injection restored endothelial cell fenestration and reduced urinary albumin levels in diabetic mice. Therefore, the present study identifies a subgroup of GECs with proangiogenic features in mice with DN. Local administration of miRNA-590-3p mimics reduces glycolytic rate and upregulates S1pR1 protein expression in proangiogenic GECs. The protective effects of miRNA-590-3p provide therapeutic potential in DN treatment.NEW & NOTEWORTHY Proangiogenetic glomerular endothelial cells (GECs) are activated in diabetic nephropathy. High glucose upregulates glycolytic enzyme phosphofructokinase/fructose bisphosphatase 3 (PFKFB3) in proangiogenetic cells. PFKFB3 protects the glomerular filtration barrier by targeting endothelial S1pR1. MiRNA-590-3p restores endothelial cell function and mitigates diabetic nephropathy.


Asunto(s)
Diabetes Mellitus Experimental , Nefropatías Diabéticas , MicroARNs , Ratones , Animales , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/metabolismo , Células Endoteliales/metabolismo , Fructosa-Bifosfatasa/metabolismo , Fructosa-Bifosfatasa/farmacología , Fosfofructoquinasas/metabolismo , Diabetes Mellitus Experimental/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo , Fosfofructoquinasa-1/metabolismo , Glucosa/metabolismo , MicroARNs/metabolismo , Albúminas/metabolismo , Albúminas/farmacología , Glucólisis
14.
Commun Biol ; 6(1): 787, 2023 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-37507476

RESUMEN

Fructose-1,6-bisphosphatase (FBPase) deficiency, caused by an FBP1 mutation, is an autosomal recessive disorder characterized by hypoglycemic lactic acidosis. Due to the rarity of FBPase deficiency, the mechanism by which the mutations cause enzyme activity loss still remains unclear. Here we identify compound heterozygous missense mutations of FBP1, c.491G>A (p.G164D) and c.581T>C (p.F194S), in an adult patient with hypoglycemic lactic acidosis. The G164D and F194S FBP1 mutants exhibit decreased FBP1 protein expression and a loss of FBPase enzyme activity. The biochemical phenotypes of all previously reported FBP1 missense mutations in addition to G164D and F194S are classified into three functional categories. Type 1 mutations are located at pivotal residues in enzyme activity motifs and have no effects on protein expression. Type 2 mutations structurally cluster around the substrate binding pocket and are associated with decreased protein expression due to protein misfolding. Type 3 mutations are likely nonpathogenic. These findings demonstrate a key role of protein misfolding in mediating the pathogenesis of FBPase deficiency, particularly for Type 2 mutations. This study provides important insights that certain patients with Type 2 mutations may respond to chaperone molecules.


Asunto(s)
Acidosis Láctica , Deficiencia de Fructosa-1,6-Difosfatasa , Humanos , Deficiencia de Fructosa-1,6-Difosfatasa/genética , Deficiencia de Fructosa-1,6-Difosfatasa/complicaciones , Fructosa-Bifosfatasa/genética , Fructosa-Bifosfatasa/metabolismo , Fructosa , Acidosis Láctica/complicaciones , Acidosis Láctica/genética , Fenotipo , Genotipo , Hipoglucemiantes
15.
PLoS One ; 18(6): e0274723, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37352301

RESUMEN

Class II Fructose-1,6-bisphosphatases (FBPaseII) (EC: 3.1.3.11) are highly conserved essential enzymes in the gluconeogenic pathway of microorganisms. Previous crystallographic studies of FBPasesII provided insights into various inactivated states of the enzyme in different species. Presented here is the first crystal structure of FBPaseII in an active state, solved for the enzyme from Francisella tularensis (FtFBPaseII), containing native metal cofactor Mn2+ and complexed with catalytic product fructose-6-phosphate (F6P). Another crystal structure of the same enzyme complex is presented in the inactivated state due to the structural changes introduced by crystal packing. Analysis of the interatomic distances among the substrate, product, and divalent metal cations in the catalytic centers of the enzyme led to a revision of the catalytic mechanism suggested previously for class II FBPases. We propose that phosphate-1 is cleaved from the substrate fructose-1,6-bisphosphate (F1,6BP) by T89 in a proximal α-helix backbone (G88-T89-T90-I91-T92-S93-K94) in which the substrate transition state is stabilized by the positive dipole of the 〈-helix backbone. Once cleaved a water molecule found in the active site liberates the inorganic phosphate from T89 completing the catalytic mechanism. Additionally, a crystal structure of Mycobacterium tuberculosis FBPaseII (MtFBPaseII) containing a bound F1,6BP is presented to further support the substrate binding and novel catalytic mechanism suggested for this class of enzymes.


Asunto(s)
Francisella tularensis , Fructosa-Bifosfatasa , Fructosa-Bifosfatasa/metabolismo , Francisella tularensis/metabolismo , Catálisis , Dominio Catalítico , Fructosa/metabolismo , Cristalografía por Rayos X
16.
Arch Biochem Biophys ; 742: 109619, 2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-37142076

RESUMEN

Fructose-1,6-bisphosphatase (FBPase) deficiency is an autosomal recessive disorder characterized by impaired gluconeogenesis caused by mutations in the fructose-1,6-bisphosphatase 1 (FBP1) gene. The molecular mechanisms underlying FBPase deficiency caused by FBP1 mutations require investigation. Herein, we report the case of a Chinese boy with FBPase deficiency who presented with hypoglycemia, ketonuria, metabolic acidosis, and repeated episodes of generalized seizures that progressed to epileptic encephalopathy. Whole-exome sequencing revealed compound heterozygous variants, c.761 A > G (H254R) and c.962C > T (S321F), in FBP1. The variants, especially the novel H254R, reduced protein stability and enzymatic activity in patient-derived leukocytes and transfected HepG2 and U251 cells. Mutant FBP1 undergoes enhanced ubiquitination and proteasomal degradation. NEDD4-2 was identified as an E3 ligase for FBP1 ubiquitination in transfected cells and the liver and brain of Nedd4-2 knockout mice. The H254R mutant FBP1 interacted with NEDD4-2 at significantly higher levels than the wild-type control. Our study identified a novel H254R variant of FBP1 underlying FBPase deficiency and elucidated the molecular mechanism underlying the enhanced NEDD4-2-mediated ubiquitination and proteasomal degradation of mutant FBP1.


Asunto(s)
Deficiencia de Fructosa-1,6-Difosfatasa , Fructosa-Bifosfatasa , Animales , Ratones , Fructosa , Deficiencia de Fructosa-1,6-Difosfatasa/genética , Fructosa-Bifosfatasa/genética , Mutación , Ubiquitinación , Humanos , Masculino , Niño
17.
Cell Metab ; 35(6): 1009-1021.e9, 2023 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-37084733

RESUMEN

Insulin inhibits gluconeogenesis and stimulates glucose conversion to glycogen and lipids. How these activities are coordinated to prevent hypoglycemia and hepatosteatosis is unclear. Fructose-1,6-bisphosphatase (FBP1) is rate controlling for gluconeogenesis. However, inborn human FBP1 deficiency does not cause hypoglycemia unless accompanied by fasting or starvation, which also trigger paradoxical hepatomegaly, hepatosteatosis, and hyperlipidemia. Hepatocyte FBP1-ablated mice exhibit identical fasting-conditional pathologies along with AKT hyperactivation, whose inhibition reversed hepatomegaly, hepatosteatosis, and hyperlipidemia but not hypoglycemia. Surprisingly, fasting-mediated AKT hyperactivation is insulin dependent. Independently of its catalytic activity, FBP1 prevents insulin hyperresponsiveness by forming a stable complex with AKT, PP2A-C, and aldolase B (ALDOB), which specifically accelerates AKT dephosphorylation. Enhanced by fasting and weakened by elevated insulin, FBP1:PP2A-C:ALDOB:AKT complex formation, which is disrupted by human FBP1 deficiency mutations or a C-terminal FBP1 truncation, prevents insulin-triggered liver pathologies and maintains lipid and glucose homeostasis. Conversely, an FBP1-derived complex disrupting peptide reverses diet-induced insulin resistance.


Asunto(s)
Fructosa , Hipoglucemia , Humanos , Ratones , Animales , Fructosa-Bifosfatasa/genética , Proteínas Proto-Oncogénicas c-akt , Insulina , Hepatomegalia/complicaciones , Hipoglucemia/etiología , Glucosa
18.
Endocrinology ; 164(6)2023 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-36964915

RESUMEN

Fructose intolerance in mammals is caused by defects in fructose absorption and metabolism. Fructose-1,6-bisphosphatase 1 (FBP1) is a key enzyme in gluconeogenesis, and its deficiency results in hypoglycemia as well as intolerance to fructose. However, the mechanism about fructose intolerance caused by FBP1 deficiency has not been fully elucidated. Here, we demonstrate that hepatic but not intestinal FBP1 is required for fructose metabolism and tolerance. We generated inducible knockout mouse models specifically lacking FBP1 in adult intestine or liver. Intestine-specific deletion of Fbp1 in adult mice does not compromise fructose tolerance, as evidenced by no significant body weight loss, food intake reduction, or morphological changes of the small intestine during 4 weeks of exposure to a high-fructose diet. By contrast, liver-specific deletion of Fbp1 in adult mice leads to fructose intolerance, as manifested by substantial weight loss, hepatomegaly, and liver injury after exposure to a high-fructose diet. Notably, the fructose metabolite fructose-1-phosphate is accumulated in FBP1-deficient liver after fructose challenge, which indicates a defect of fructolysis, probably due to competitive inhibition by fructose-1,6-bisphosphate and may account for the fructose intolerance. In conclusion, these data have clarified the essential role of hepatic but not intestinal FBP1 in fructose metabolism and tolerance.


Asunto(s)
Intolerancia a la Fructosa , Fructosa , Animales , Ratones , Fructosa-Bifosfatasa/genética , Gluconeogénesis/genética , Intestinos , Hígado , Mamíferos
19.
Cell Res ; 33(3): 245-257, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36646759

RESUMEN

Emerging evidence demonstrates that some metabolic enzymes that phosphorylate soluble metabolites can also phosphorylate a variety of protein substrates as protein kinases to regulate cell cycle, apoptosis and many other fundamental cellular processes. However, whether a metabolic enzyme dephosphorylates protein as a protein phosphatase remains unknown. Here we reveal the gluconeogenic enzyme fructose 1,6-biphosphatase 1 (FBP1) that catalyzes the hydrolysis of fructose 1,6-bisphosphate (F-1,6-BP) to fructose 6-phosphate (F-6-P) as a protein phosphatase by performing a high-throughput screening of metabolic phosphatases with molecular docking followed by molecular dynamics (MD) simulations. Moreover, we identify IκBα as the substrate of FBP1-mediated dephosphorylation by performing phosphoproteomic analysis. Mechanistically, FBP1 directly interacts with and dephosphorylates the serine (S) 32/36 of IκBα upon TNFα stimulation, thereby inhibiting NF-κB activation. MD simulations indicate that the catalytic mechanism of FBP1-mediated IκBα dephosphorylation is similar to F-1,6-BP dephosphorylation, except for higher energetic barriers for IκBα dephosphorylation. Functionally, FBP1-dependent NF-κB inactivation suppresses colorectal tumorigenesis by sensitizing tumor cells to inflammatory stresses and preventing the mobilization of myeloid-derived suppressor cells. Our finding reveals a previously unrecognized role of FBP1 as a protein phosphatase and establishes the critical role of FBP1-mediated IκBα dephosphorylation in colorectal tumorigenesis.


Asunto(s)
Neoplasias Colorrectales , Fructosa-Bifosfatasa , Humanos , Fructosa-Bifosfatasa/análisis , Fructosa-Bifosfatasa/metabolismo , FN-kappa B , Inhibidor NF-kappaB alfa , Simulación del Acoplamiento Molecular , Carcinogénesis , Monoéster Fosfórico Hidrolasas , Transformación Celular Neoplásica , Fructosa
20.
Science ; 379(6628): 185-190, 2023 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-36634192

RESUMEN

Hummingbirds possess distinct metabolic adaptations to fuel their energy-demanding hovering flight, but the underlying genomic changes are largely unknown. Here, we generated a chromosome-level genome assembly of the long-tailed hermit and screened for genes that have been specifically inactivated in the ancestral hummingbird lineage. We discovered that FBP2 (fructose-bisphosphatase 2), which encodes a gluconeogenic muscle enzyme, was lost during a time period when hovering flight evolved. We show that FBP2 knockdown in an avian muscle cell line up-regulates glycolysis and enhances mitochondrial respiration, coincident with an increased mitochondria number. Furthermore, genes involved in mitochondrial respiration and organization have up-regulated expression in hummingbird flight muscle. Together, these results suggest that FBP2 loss was likely a key step in the evolution of metabolic muscle adaptations required for true hovering flight.


Asunto(s)
Adaptación Fisiológica , Aves , Vuelo Animal , Fructosa-Bifosfatasa , Gluconeogénesis , Músculo Esquelético , Animales , Aves/genética , Aves/metabolismo , Metabolismo Energético/genética , Vuelo Animal/fisiología , Gluconeogénesis/genética , Adaptación Fisiológica/genética , Fructosa-Bifosfatasa/genética , Músculo Esquelético/enzimología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA