Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
J Biol Chem ; 300(8): 107541, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38992438

RESUMEN

The amyloid precursor protein (APP) is a key protein in Alzheimer's disease synthesized in the endoplasmic reticulum (ER) and translocated to the plasma membrane where it undergoes proteolytic cleavages by several proteases. Conversely, to other known proteases, we previously elucidated rhomboid protease RHBDL4 as a novel APP processing enzyme where several cleavages likely occur already in the ER. Interestingly, the pattern of RHBDL4-derived large APP C-terminal fragments resembles those generated by the η-secretase or MT5-MMP, which was described to generate so-called Aη fragments. The similarity in large APP C-terminal fragments between both proteases raised the question of whether RHBDL4 may contribute to η-secretase activity and Aη-like fragments. Here, we identified two cleavage sites of RHBDL4 in APP by mass spectrometry, which, intriguingly, lie in close proximity to the MT5-MMP cleavage sites. Indeed, we observed that RHBDL4 generates Aη-like fragments in vitro without contributions of α-, ß-, or γ-secretases. Such Aη-like fragments are likely generated in the ER since RHBDL4-derived APP-C-terminal fragments do not reach the cell surface. Inherited, familial APP mutations appear to not affect this processing pathway. In RHBDL4 knockout mice, we observed increased cerebral full-length APP in comparison to wild type (WT) in support of RHBDL4 being a physiologically relevant protease for APP. Furthermore, we found secreted Aη fragments in dissociated mixed cortical cultures from WT mice, however significantly fewer Aη fragments in RHBDL4 knockout cultures. Our data underscores that RHBDL4 contributes to the η-secretease-like processing of APP and that RHBDL4 is a physiologically relevant protease for APP.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide , Precursor de Proteína beta-Amiloide , Precursor de Proteína beta-Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Secretasas de la Proteína Precursora del Amiloide/genética , Ratones , Humanos , Proteolisis , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Células HEK293 , Ratones Noqueados , Retículo Endoplásmico/metabolismo
2.
bioRxiv ; 2024 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-38464180

RESUMEN

Characteristic cerebral pathological changes of Alzheimer's disease (AD) such as glucose hypometabolism or the accumulation of cleavage products of the amyloid precursor protein (APP), known as Aß peptides, lead to sustained endoplasmic reticulum (ER) stress and neurodegeneration. To preserve ER homeostasis, cells activate their unfolded protein response (UPR). The rhomboid-like-protease 4 (RHBDL4) is an enzyme that participates in the UPR by targeting proteins for proteasomal degradation. We demonstrated previously that RHBLD4 cleaves APP in HEK293T cells, leading to decreased total APP and Aß. More recently, we showed that RHBDL4 processes APP in mouse primary mixed cortical cultures as well. Here, we aim to examine the physiological relevance of RHBDL4 in the brain. We first found that brain samples from AD patients and an AD mouse model (APPtg) showed increased RHBDL4 mRNA and protein expression. To determine the effects of RHBDL4's absence on APP physiology in vivo, we crossed APPtg mice to a RHBDL4 knockout (R4-/-) model. RHBDL4 deficiency in APPtg mice led to increased total cerebral APP and amyloidogenic processing when compared to APPtg controls. Contrary to expectations, as assessed by cognitive tests, RHBDL4 absence rescued cognition in 5-month-old female APPtg mice. Informed by unbiased RNAseq data, we demonstrated in vitro and in vivo that RHBDL4 absence leads to greater levels of active ß-catenin due to decreased proteasomal clearance. Decreased ß-catenin activity is known to underlie cognitive defects in APPtg mice and AD. Our work suggests that RHBDL4's increased expression in AD, in addition to regulating APP levels, leads to aberrant degradation of ß-catenin, contributing to cognitive impairment.

3.
Biochem Cell Biol ; 97(3): 265-269, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30102867

RESUMEN

Proteases carry out a wide variety of physiological functions. This review presents a brief history of protease research, starting with the original discovery of pepsin in 1836. Following the path of time, we revisit how proteases were originally classified based on their catalytic mechanism and how chemical and crystallographic studies unravelled the mechanism of serine proteases. Ongoing research on proteases addresses their biological roles, small molecule inhibitors for therapeutic uses, and protein engineering to modify their activities. The discovery of intramembrane proteases is more recent, beginning with the discovery of site-2 protease in 1997. Since then, different mechanistic classes of intramembrane proteases have been characterized, and many of these act in regulated intramembrane proteolysis in signaling pathways. Furthermore, the rhomboid intramembrane proteases were discovered by genetic and biochemical experiments in Drosophila and then in human cells. Research on the intramembrane proteases is expanding, as their biological importance is recognized.


Asunto(s)
Membrana Celular/metabolismo , Péptido Hidrolasas/metabolismo , Proteolisis , Humanos
4.
Biol Chem ; 399(12): 1399-1408, 2018 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-30171808

RESUMEN

Since the first genetic description of a rhomboid in Drosophila melanogaster, tremendous efforts have been geared towards elucidating the proteolytic mechanism of this particular class of intramembrane proteases. In particular, mammalian rhomboid proteases sparked our interest and we aimed to investigate the human homologue RHBDL4. In light of our recent finding of the amyloid precursor protein (APP) family as efficient substrates of RHBDL4, we were enticed to further study the specific proteolytic mechanism of this enzyme by comparing cleavage patterns of wild type APP and APP TMS chimeras. Here, we demonstrate that the introduction of positively charged amino acid residues in the TMS redirects the RHBDL4-mediated cleavage of APP from its ectodomain closer towards the TMS, possibly inducing an ER-associated degradation (ERAD) of the substrate. In addition, we concluded that the cytoplasmic tail and proposed palmitoylation sites in the ectodomain of APP are not essential for the RHBDL4-mediated APP processing. In summary, our previously identified APP ectodomain cleavages by RHBDL4 are a subsidiary mechanism to the proposed RHBDL4-mediated ERAD of substrates likely through a single cleavage near or within the TMS.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Proteínas de la Membrana/metabolismo , Procesamiento Proteico-Postraduccional , Precursor de Proteína beta-Amiloide/antagonistas & inhibidores , Células Cultivadas , Relación Dosis-Respuesta a Droga , Degradación Asociada con el Retículo Endoplásmico/efectos de los fármacos , Células HEK293 , Humanos , Leupeptinas/farmacología , Proteínas de la Membrana/antagonistas & inhibidores , Inhibidores de Proteasas/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Relación Estructura-Actividad
5.
J Biol Chem ; 293(40): 15556-15568, 2018 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-30143535

RESUMEN

In the last decade, intramembrane proteases have gained increasing attention because of their many links to various diseases. Nevertheless, our understanding as to how they function or how they are regulated is still limited, especially when it comes to human homologues. In this regard, here we sought to unravel mechanisms of regulation of the protease rhomboid-like protein-4 (RHBDL4), one of five active human serine intramembrane proteases. In view of our recent finding that human RHBDL4 efficiently cleaves the amyloid precursor protein (APP), a key protein in the pathology of Alzheimer's disease, we used established reagents to modulate the cellular cholesterol content and analyzed the effects of this modulation on RHBDL4-mediated processing of endogenous APP. We discovered that lowering membrane cholesterol levels increased the levels of RHBDL4-specific endogenous APP fragments, whereas high cholesterol levels had the opposite effect. Direct binding of cholesterol to APP did not mediate these modulating effects of cholesterol. Instead, using homology modeling, we identified two potential cholesterol-binding motifs in the transmembrane helices 3 and 6 of RHBDL4. Substitution of the essential tyrosine residues of the potential cholesterol-binding motifs to alanine increased the levels of endogenous APP C-terminal fragments, reflecting enhanced RHBDL4 activity. In summary, we provide evidence that the activity of RHBDL4 is regulated by cholesterol likely through a direct binding of cholesterol to the enzyme.


Asunto(s)
Precursor de Proteína beta-Amiloide/genética , Membrana Celular/efectos de los fármacos , Colesterol/farmacología , Proteínas de la Membrana/genética , Secuencia de Aminoácidos , Precursor de Proteína beta-Amiloide/metabolismo , Anticolesterolemiantes/farmacología , Sitios de Unión , Membrana Celular/química , Membrana Celular/metabolismo , Colesterol/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Humanos , Lipoproteínas LDL/farmacología , Proteínas de la Membrana/metabolismo , Metaloproteasas/genética , Metaloproteasas/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Modelos Moleculares , Proproteína Convertasas/genética , Proproteína Convertasas/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Dominios y Motivos de Interacción de Proteínas , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Transducción de Señal , Simvastatina/farmacología , Proteínas de Unión a los Elementos Reguladores de Esteroles/genética , Proteínas de Unión a los Elementos Reguladores de Esteroles/metabolismo
6.
J Membr Biol ; 251(3): 369-378, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29260282

RESUMEN

Proteases, sharp yet unforgivable tools of every cell, require tight regulation to ensure specific non-aberrant cleavages. The relatively recent discovered class of intramembrane proteases has gained increasing interest due to their involvement in important signaling pathways linking them to diseases including Alzheimer's disease and cancer. Despite tremendous efforts, their regulatory mechanisms have only started to unravel. There is evidence that the membrane composition itself can regulate intramembrane protease activity and specificity. In this review, we highlight the work on γ-secretase and rhomboid proteases and summarize several studies as to how different lipids impact on enzymatic activity.


Asunto(s)
Membrana Celular/metabolismo , Lípidos de la Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Endopeptidasas/metabolismo , Proteínas de la Membrana/genética , Unión Proteica , Proteolisis , Especificidad por Sustrato
7.
J Biol Chem ; 291(42): 21903-21912, 2016 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-27563067

RESUMEN

The amyloid precursor protein (APP) is an ubiquitously expressed cell surface protein and a key molecule in the etiology of Alzheimer disease. Amyloidogenic processing of APP through secretases leads to the generation of toxic amyloid ß (Aß) peptides, which are regarded as the molecular cause of the disease. We report here an alternative processing pathway of APP through the mammalian intramembrane rhomboid protease RHBDL4. RHBDL4 efficiently cleaves APP inside the cell, thus bypassing APP from amyloidogenic processing, leading to reduced Aß levels. RHBDL4 cleaves APP multiple times in the ectodomain, resulting in several N- and C-terminal fragments that are not further degraded by classical APP secretases. Knockdown of endogenous RHBDL4 results in decreased levels of C-terminal fragments derived from endogenous APP. Similarly, we found the APP family members APLP1 and APLP2 to be substrates of RHBDL4. We conclude that RHBDL4-mediated APP processing provides insight into APP and rhomboid physiology and qualifies for further investigations to elaborate its impact on Alzheimer disease pathology.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Proteínas de la Membrana/metabolismo , Procesamiento Proteico-Postraduccional , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Línea Celular , Humanos , Proteínas de la Membrana/genética , Dominios Proteicos
8.
J Biol Chem ; 287(47): 39492-504, 2012 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-22961979

RESUMEN

Bone (or body) morphogenetic proteins (BMPs) belong to the TGFß superfamily and are crucial for embryonic patterning and organogenesis as well as for adult tissue homeostasis and repair. Activation of BMP receptors by their ligands leads to induction of several signaling cascades. Using fluorescence recovery after photobleaching, FRET, and single particle tracking microscopy, we demonstrate that BMP receptor type I and II (BMPRI and BMPRII) have distinct lateral mobility properties within the plasma membrane, which is mandatory for their involvement in different signaling pathways. Before ligand binding, BMPRI and a subpopulation of BMPRII exhibit confined motion, reflecting preassembled heteromeric receptor complexes. A second free diffusing BMPRII population only becomes restricted after ligand addition. This paper visualizes time-resolved BMP receptor complex formation and demonstrates that the lateral mobility of BMPRI has a major impact in stabilizing heteromeric BMPRI-BMPRII receptor complexes to differentially stimulate SMAD versus non-SMAD signaling.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Membrana Celular/metabolismo , Complejos Multiproteicos/metabolismo , Transducción de Señal/fisiología , Proteínas Smad/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Membrana Celular/genética , Células HEK293 , Humanos , Complejos Multiproteicos/genética , Transporte de Proteínas/fisiología , Proteínas Smad/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA