Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mol Neurobiol ; 60(5): 2397-2412, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36656458

RESUMEN

Cells perform regular maintenance to avoid the accumulation of misfolded proteins. Prolonged accumulation of these proteotoxic inclusions generates potential risk of ageing-related diseases such as neurodegenerative diseases. Therefore, removal of such abnormal aggregates can ensure the re-establishment of proteostasis. Ubiquitin proteasome system (UPS) actively participates in the selective removal of aberrantly folded clients with the help of complex proteasome machinery. However, specific induction of proteasome functions to remove abnormal proteins remains an open challenge. Here, we show that Itraconazole treatment induces proteasome activities and degrades the accumulation of bonafide-misfolded proteins, including heat-denatured luciferase. Exposure of Itraconazole elevates the degradation of neurodegenerative disease-associated proteins, e.g. expanded polyglutamine, mutant SOD1, and mutant α-synuclein. Our results suggest that Itraconazole treatment prevents the accumulation of neurodegenerative disease-linked misfolded proteins and generates cytoprotection. These findings reveal that Itraconazole removes abnormal proteins through sequential proteasomal activation and represents a potential protective therapeutic role against protein-misfolding neurodegenerative diseases.


Asunto(s)
Enfermedades Neurodegenerativas , Humanos , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Agregado de Proteínas , Itraconazol/farmacología , Itraconazol/uso terapéutico , Citoprotección , Pliegue de Proteína
2.
Cell Physiol Biochem ; 56(5): 530-545, 2022 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-36168821

RESUMEN

BACKGROUND/AIMS: Cells require regular maintenance of proteostasis. Synthesis of new polypeptides and elimination of damaged or old proteins is an uninterrupted mechanism essential for a healthy cellular environment. Impairment in the removal of misfolded proteins can disturb proteostasis; such toxic aggregation of misfolded proteins can act as a primary risk factor for neurodegenerative diseases and imperfect ageing. The critical challenge is to design effective protein quality control (PQC) based molecular tactics that could potentially eliminate aggregation-prone protein load from the cell. Still, targeting specific components of the PQC pathway for the suppression of proteotoxic insults retains several challenges. Earlier, we had observed that LRSAM1 promotes the degradation of aberrant proteins. Here, we examined the effect of resveratrol, a stilbenoid phytoalexin compound, treatment on LRSAM1 E3 ubiquitin ligase, involved in the spongiform neurodegeneration. METHODS: In this study, we reported induction of mRNA and protein levels of LRSAM1 in response to resveratrol treatment via RT-PCR, immunoblotting, and immunofluorescence analysis. The LRSAM1-mediated proteasomal-based clearance of misfolded proteins was also investigated via proteasome activity assays, immunoblotting and immunofluorescence analysis. The increased stability of LRSAM1 by resveratrol was demonstrated by cycloheximide chase analysis. RESULTS: Here, we show that resveratrol treatment induces LRSAM1 E3 ubiquitin ligase expression levels. Further, our findings suggest that overexpression of LRSAM1 significantly elevates proteasome activities and improves the degradation of bona fide heat-denatured luciferase protein. Exposure of resveratrol not only slows down the turnover of LRSAM1 but also effectively degrades abnormal proteinaceous inclusions, which eventually promotes cell viability. CONCLUSION: Our findings suggest that resveratrol facilitates LRSAM1 endogenous establishment, which consequently promotes the proteasome machinery for effective removal of intracellular accumulated misfolded or proteasomal-designated substrates. Altogether, our study proposes a promising molecular approach to specifically trigger PQC signaling for efficacious rejuvenation of defective proteostasis via activation of overburdened proteolytic machinery.


Asunto(s)
Complejo de la Endopetidasa Proteasomal , Ubiquitina-Proteína Ligasas , Cicloheximida , Luciferasas , Péptidos , Complejo de la Endopetidasa Proteasomal/metabolismo , ARN Mensajero , Resveratrol/farmacología , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
3.
Mol Neurobiol ; 58(12): 6593-6609, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34590243

RESUMEN

According to cellular demands, ribosomes synthesize and maintain the desired pool of proteins inside the cell. However, sometimes due to defects in ribosomal machinery and faulty mRNAs, these nascent polypeptides are constantly under threat to become non-functional. In such conditions, cells acquire the help of ribosome-associated quality control mechanisms (RQC) to eliminate such aberrant nascent proteins. The primary regulator of RQC is RING domain containing LISTERIN E3 ubiquitin ligase, which is associated with ribosomes and alleviates non-stop proteins-associated stress in cells. Mouse RING finger protein E3 ubiquitin ligase LISTERIN is crucial for embryonic development, and a loss in its function causes neurodegeneration. LISTERIN is overexpressed in the mouse brain and spinal cord regions, and its perturbed functions generate neurological and motor deficits, but the mechanism of the same is unclear. Overall, LISTERIN is crucial for brain health and brain development. The present article systematically describes the detailed nature, molecular functions, and cellular physiological characterization of LISTERIN E3 ubiquitin ligase. Improve comprehension of LISTERIN's neurological roles may uncover pathways linked with neurodegeneration, which in turn might elucidate a promising novel therapeutic intervention against human neurodegenerative diseases.


Asunto(s)
Encéfalo/metabolismo , Ribosomas/metabolismo , Médula Espinal/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Desarrollo Embrionario/fisiología , Humanos , Neuronas/metabolismo , Pliegue de Proteína , Isoformas de Proteínas/metabolismo , Ubiquitinación
4.
Cell Signal ; 77: 109836, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33207262

RESUMEN

Numerous proteins participate and actively contribute to the various cellular mechanisms, where several of them are crucial for regular metabolism, including survival. Thus, to maintain optimal cellular physiology, cells govern protein quality control functions with the assistance of comprehensive actions of molecular chaperones, the ubiquitin-proteasome system, and autophagy. In the ubiquitin-proteasome pathway, few quality control E3 ubiquitin ligases actively participate against misfolded protein aggregation generated via stress conditions. But how these quality control E3s active expression levels returned to basal levels when cells achieved re-establishment of proteostasis is still poorly understood. Our current study demonstrated that LRSAM1 E3 ubiquitin ligase promotes the proteasomal degradation of quality control E3 ubiquitin ligase E6-AP. We have observed the co-localization and recruitment of LRSAM1 with E6-AP protein and noticed that LRSAM1 induces the endogenous turnover of E6-AP. Partial depletion of LRSAM1 elevates the levels of E6-AP and affects overall cell cycle regulatory proteins (p53 and p27) expression, including the rate of cellular proliferation. The current finding also provides an excellent opportunity to better understand the basis of the E6-AP associated pathomechanism of Angelman Syndrome disorder. Additionally, this study touches upon the novel potential molecular strategy to regulate the levels of one quality control E3 ubiquitin ligase with another E3 ubiquitin ligase and restore proteostasis and provide a possible therapeutic approach against abnormal protein aggregation diseases.


Asunto(s)
Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Células A549 , Animales , Células COS , Proliferación Celular , Chlorocebus aethiops , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Humanos , Agregado de Proteínas , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/genética
5.
Int J Biochem Cell Biol ; 120: 105697, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31982566

RESUMEN

Accumulation of aberrant misfolded proteins is a major hallmark of several neurodegenerative diseases. Intracellular accumulations of such abnormal proteins are selectively cleared by the ubiquitin-proteasome system (UPS). But how the failure of misfolded protein degradation cause proteinopathies is still an unanswered question?. Previous studies have suggested that few selective quality control (QC) E3 ubiquitin ligase from the UPS can selectively target insoluble aggregated proteins for their intracellular degradation. Few reports suggest that lack or aberrant functions of QC E3 ubiquitin ligases can be a possible causative factor of neurodegeneration and aging. Earlier findings indicated that leucine-rich repeat and sterile alpha motif containing-1 (LRSAM1) is associated with Charcot-Marie-Tooth Type 2P (CMT2P) disease in which loss of LRSAM1 function sensitizes peripheral axons for degeneration. Here, our current study for the first time demonstrates that E3 ubiquitin Ligase LRSAM1 is a really interesting new gene (RING) class protein which suppresses the accumulation of misfolded protein aggregates and also alleviates their deleterious cytotoxic effects. We have also observed that LRSAM1 expression is elevated under neurodegenerative stress conditions, and partial depletion of LRSAM1 endogenous levels aggravates mitochondrial abnormalities and severely affects cell survival during proteotoxic insults. Overall, our current finding indicates that LRSAM1 can alleviate cytotoxic insults mediated by a variety of neurodegeneration linked proteotoxic stress events, and most likely LRSAM1 interplay a significant role in between different components of cellular protein quality control mechanism. This study will also allow us to better comprehend the problem of proteinopathies linked with aberrant protein accumulation and open new possibilities to better elucidate the molecular mechanisms involved in the pathologies of neurodegeneration and aging.


Asunto(s)
Enfermedades Neurodegenerativas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Células A549 , Animales , Células COS , Muerte Celular , Chlorocebus aethiops , Humanos , Enfermedades Neurodegenerativas/patología , Proteolisis , Transfección , Ubiquitina/metabolismo
6.
Sci Rep ; 9(1): 16872, 2019 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-31728006

RESUMEN

CAG repeats RNA causes various fatal neurodegenerative diseases exemplified by Huntington's disease (HD) and several spinocerebellar ataxias (SCAs). Although there are differences in the pathogenic mechanisms, these diseases share the common cause, i.e., expansion of CAG repeats. The shared cause of these diseases raises the possibility for the exploiting the common target as a potential therapeutic approach. Oligonucleotide-based therapeutics are designed earlier with the help of the base pairing rule but are not very promiscuous, considering the nonspecific stimulation of the immune system and the poor cellular delivery. Therefore, small molecules-based therapeutics are preferred for targeting the repeats expansion disorders. Here, we have used the chemical similarity search approach to discern the small molecules that selectively target toxic CAG RNA. The lead compounds showed the specificity towards AA mismatch in biophysical studies including CD, ITC, and NMR spectroscopy and thus aided to forestall the polyQ mediated pathogenicity. Furthermore, the lead compounds also explicitly alleviate the polyQ mediated toxicity in HD cell models and patient-derived cells. These findings suggest that the lead compound could act as a chemical probe for AA mismatch containing RNA as well as plays a neuroprotective role in fatal neurodegenerative diseases like HD and SCAs.


Asunto(s)
Fibroblastos/efectos de los fármacos , Flavonoides/farmacología , Fármacos Neuroprotectores/farmacología , Péptidos/antagonistas & inhibidores , ARN/química , Bibliotecas de Moléculas Pequeñas/farmacología , Benzotiazoles/química , Bioensayo , Supervivencia Celular/efectos de los fármacos , Descubrimiento de Drogas , Fibroblastos/metabolismo , Fibroblastos/patología , Flavonoides/química , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Simulación del Acoplamiento Molecular , Fármacos Neuroprotectores/química , Conformación de Ácido Nucleico , Péptidos/química , Péptidos/metabolismo , Cultivo Primario de Células , Agregado de Proteínas/efectos de los fármacos , ARN/genética , ARN/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Expansión de Repetición de Trinucleótido/efectos de los fármacos
7.
Biochimie ; 163: 21-32, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31075282

RESUMEN

Huntington's diseases (HD) is a very devastating disease caused by r(CAG) expansion in HTT gene, encoding the huntingtin protein. r(CAG) expansion causes disease via multiple pathways including, 1) loss of normal protein function like sequestration of RNA binding protein such as Muscleblind-like (MBNL) and nucleolin, 2) Gain of function for mutant proteins and 3) repeat-associated non-ATG (RAN) translation; in which expanded r(CAG) translates into toxic poly glu, poly ser, or poly ala without the use of any canonical start codon. Herein, we have rationally designed and synthesized a unique class of pyridocoumarin derivatives that target the r(CAG)exp involved in HD and spinocerebellar ataxia (SCA) pathogenesis. Notably, compounds 3 and 15 showed higher affinity (nanomolar Kd) and selectivity for diseased r(CAG)exp RNA compared to regular duplex AU-paired RNA. Interestingly, both scaffolds are cell permeable, exhibit low toxicity to healthy fibroblast cells and are also capable of reducing the level of poly Q aggregation in cellular models. Indeed, our current study offers promising facet for selectively targeting repeats containing RNAs that cause severe diseases like HD and SCAs.


Asunto(s)
Cumarinas/química , Enfermedad de Huntington/tratamiento farmacológico , Proteínas Mutantes/genética , ARN Mensajero/química , Ataxias Espinocerebelosas/tratamiento farmacológico , Células Cultivadas , Cumarinas/farmacología , Cumarinas/uso terapéutico , Diseño de Fármacos , Humanos , Proteína Huntingtina , Enfermedad de Huntington/metabolismo , Cinética , Simulación del Acoplamiento Molecular , Conformación de Ácido Nucleico , ARN Mensajero/efectos de los fármacos , Ataxias Espinocerebelosas/metabolismo , Expansión de Repetición de Trinucleótido
8.
J Cell Physiol ; 234(11): 20900-20914, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31004355

RESUMEN

Major neurodegenerative disorders are characterized by the formation of misfolded proteins aggregates inside or outside the neuronal cells. Previous studies suggest that aberrant proteins aggregates play a critical role in protein homeostasis imbalance and failure of protein quality control (PQC) mechanism, leading to disease conditions. However, we still do not understand the precise mechanisms of PQC failure and cellular dysfunctions associated with neurodegenerative diseases caused by the accumulation of protein aggregates. Here, we show that Myricetin, a flavonoid, can eliminate various abnormal proteins from the cellular environment via modulating endogenous levels of Hsp70 chaperone and quality control (QC)-E3 ubiquitin ligase E6-AP. We have observed that Myricetin treatment suppresses the aggregation of different aberrant proteins. Myricetin also enhances the elimination of various toxic neurodegenerative diseases associated proteins from the cells, which could be reversed by the addition of putative proteasome inhibitor (MG132). Remarkably, Myricetin can also stabilize E6-AP and reduce the misfolded proteins inclusions, which further alleviates cytotoxicity. Taken together these findings suggested that new mechanistic and therapeutic insights based on small molecules mediated regulation of disturbed protein quality control mechanism, which may result in the maintenance of the state of proteostasis.


Asunto(s)
Flavonoides/farmacología , Degeneración Nerviosa/metabolismo , Polifenoles/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Agregado de Proteínas , Proteolisis , Regulación hacia Arriba/efectos de los fármacos , Células A549 , Estabilidad de Enzimas/efectos de los fármacos , Humanos , Cuerpos de Inclusión/efectos de los fármacos , Cuerpos de Inclusión/metabolismo , Luciferasas/metabolismo , Modelos Biológicos , Proteínas Mutantes/metabolismo , Degeneración Nerviosa/patología , Péptidos/metabolismo , Agregado de Proteínas/efectos de los fármacos , Pliegue de Proteína/efectos de los fármacos , Proteolisis/efectos de los fármacos , Solubilidad , Estrés Fisiológico/efectos de los fármacos , Superóxido Dismutasa/metabolismo , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/metabolismo , alfa-Sinucleína/metabolismo
9.
Cell Mol Life Sci ; 76(11): 2093-2110, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30826859

RESUMEN

Cellular protein quality control (PQC) plays a significant role in the maintenance of cellular homeostasis. Failure of PQC mechanism may lead to various neurodegenerative diseases due to accumulation of aberrant proteins. To avoid such fatal neuronal conditions PQC employs autophagy and ubiquitin proteasome system (UPS) to degrade misfolded proteins. Few quality control (QC) E3 ubiquitin ligases interplay an important role to specifically recognize misfolded proteins for their intracellular degradation. Leucine-rich repeat and sterile alpha motif-containing 1 (LRSAM1) is a really interesting new gene (RING) class protein that possesses E3 ubiquitin ligase activity with promising applications in PQC. LRSAM1 is also known as RING finger leucine repeat rich (RIFLE) or TSG 101-associated ligase (TAL). LRSAM1 has various cellular functions as it modulates the protein aggregation, endosomal sorting machinery and virus egress from the cells. Thus, this makes LRSAM1 interesting to study not only in protein conformational disorders such as neurodegeneration but also in immunological and other cancerous disorders. Furthermore, LRSAM1 interacts with both cellular protein degradation machineries and hence it can participate in maintenance of overall cellular proteostasis. Still, more research work on the quality control molecular functions of LRSAM1 is needed to comprehend its roles in various protein aggregatory diseases. Earlier findings suggest that in a mouse model of Charcot-Marie-Tooth (CMT) disease, lack of LRSAM1 functions sensitizes peripheral axons to degeneration. It has been observed that in CMT the patients retain dominant and recessive mutations of LRSAM1 gene, which encodes most likely a defective protein. However, still the comprehensive molecular pathomechanism of LRSAM1 in neuronal functions and neurodegenerative diseases is not known. The current article systematically represents the molecular functions, nature and detailed characterization of LRSAM1 E3 ubiquitin ligase. Here, we review emerging molecular mechanisms of LRSAM1 linked with neurobiological functions, with a clear focus on the mechanism of neurodegeneration and also on other diseases. Better understanding of LRSAM1 neurobiological and intracellular functions may contribute to develop promising novel therapeutic approaches, which can also propose new lines of molecular beneficial targets for various neurodegenerative diseases.


Asunto(s)
Proteínas del Tejido Nervioso/genética , Enfermedades Neurodegenerativas/genética , Nervios Periféricos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina-Proteína Ligasas/genética , Animales , Axones/metabolismo , Axones/patología , Regulación de la Expresión Génica , Humanos , Mutación , Proteínas del Tejido Nervioso/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Nervios Periféricos/patología , Agregado de Proteínas , Pliegue de Proteína , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteolisis , Proteostasis/genética , Transducción de Señal , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
10.
Med Res Rev ; 38(6): 1916-1973, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29719055

RESUMEN

Proteins actively participate in a wide range of cellular physiological functions. But aggregation of proteins results in cytotoxicity, and unwanted aggregation of misfolded proteins often causes many diseases. During abnormal protein aggregation events, cells try to cope against such deleterious consequences because of the remarkable functional attempts of two distinct proteolytic mechanisms. These tightly regulative and signaling mechanisms are autophagy pathway and ubiquitin proteasome system. Proteasome complex system holds the elimination capacity of intracellular aberrant protein aggregation. Despite the considerable progress that has been achieved, which elucidates wide function and diverse roles of proteasome system, still several crucial problems remain unanswered. For example, how the complex proteasomes assembly and their interactive pathways determine the precise sense of several proteotoxic insults, which can severely affect the cell survival and homeostasis? The specific degradation of various aberrant proteins that can disturb cellular homeostasis is achieved by proper proteasome functionality, which is yet another unclear and critical challenge. Therefore, a better understanding of the various cellular signaling mechanisms composing the proteasome machinery carries broad therapeutic implications linked with proteopathies. This article signifies the urgent need, which is now crucial for us to improve our understanding of the proteasome architecture, structure, and functions that span multiple level strategies from the molecular level to the cellular level. This systematic in-depth information of proteasome may be helpful in the near future to design a new molecular framework based on intrinsic and extrinsic cellular mechanisms that drive the assembly of proteasome to induce cellular survival against proteostasis imbalance and disease conditions.


Asunto(s)
Enfermedad , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteostasis , Animales , Homeostasis , Humanos , Estrés Fisiológico , Ubiquitina/metabolismo
11.
Mol Neurobiol ; 55(2): 1169-1182, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28102469

RESUMEN

Accumulation of misfolded or aberrant proteins in neuronal cells is linked with neurodegeneration and other pathologies. Which molecular mechanisms fail and cause inappropriate folding of proteins and what is their relationship to cellular toxicity is not well known. How does it happen and what are the probable therapeutic or molecular approaches to counter them are also not clear. Here, we demonstrate that treatment of lanosterol diminishes aberrant proteotoxic aggregation and mitigates their cytotoxicity via induced expression of co-chaperone CHIP and elevated autophagy. The addition of lanosterol not only reduces aggregation of mutant bonafide misfolded proteins but also effectively prevents accumulation of various mutant disease-prone proteotoxic proteins. Finally, we observed that lanosterol mitigates cytotoxicity in cells, mediated by different stress-inducing agents. Taken together, our present results suggest that upregulation of cellular molecular chaperones, primarily using small molecules, can probably offer an efficient therapeutic approach in the future against misfolding of different disease-causing proteins and neurodegenerative disorders. Graphical Abstract ᅟ.


Asunto(s)
Autofagia/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Lanosterol/farmacología , Enfermedades Neurodegenerativas/metabolismo , Agregado de Proteínas/efectos de los fármacos , Pliegue de Proteína/efectos de los fármacos , Células A549 , Animales , Células COS , Chlorocebus aethiops , Humanos , Luciferasas/metabolismo , Chaperonas Moleculares/metabolismo
12.
J Cell Physiol ; 233(2): 1685-1699, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28681929

RESUMEN

Non-steroidal anti-inflammatory drugs (NSAIDs) are a class of drugs that are mainly used to treat pain, inflammation, and fever via cyclooxygenase-2 (COX-2) inhibition. There are abundant findings that uncover the hidden critical chemotherapeutics potential of NSAIDs in cancer treatment. However, still the precise mechanism by which NSAIDs could be used as an effective anti-tumor agent in the prevention of carcinogenesis is not well understood. Here, we show that indomethacin, a well-known NSAID, induces proteasomal dysfunction that results in accumulation of unwanted proteins, mitochondrial abnormalities, and successively stimulate apoptosis in cells. We observed the interaction of indomethacin with proteasome and noticed the massive accumulation of intracellular ubiquitin-positive proteins, which might be due to the suppression of proteasome activities. Furthermore, we also found that exposure of indomethacin causes the accumulation of critical proteasomal substrates that consequently generate severe mitochondrial abnormalities and prompt up key apoptotic events in cells. Our results demonstrate how indomethacin affects normal proteasomal functions and induces mitochondrial apoptosis in cells. These findings also improve our current understanding of how NSAIDs can exhibit crucial anti-proliferative effects in cells. In near future, our findings may suggest a new possible strategy for the development of specific proteasome inhibitors in conjunction with other chemo-preventive anticancer agents.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Indometacina/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Mitocondrias/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Inhibidores de Proteasoma/farmacología , Células A549 , Animales , Antiinflamatorios no Esteroideos/química , Antineoplásicos/química , Células COS , Chlorocebus aethiops , Relación Dosis-Respuesta a Droga , Humanos , Indometacina/química , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/patología , Mitocondrias/metabolismo , Mitocondrias/patología , Simulación del Acoplamiento Molecular , Complejo de la Endopetidasa Proteasomal/química , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/química , Agregado de Proteínas , Unión Proteica , Proteolisis , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad , Factores de Tiempo , Ubiquitinación
13.
Prog Neurobiol ; 159: 1-38, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28870769

RESUMEN

Proteins are ordered useful cellular entities, required for normal health and organism's survival. The proteome is the absolute set of cellular expressed proteins, which regulates a wide range of physiological functions linked with all domains of life. In aging cells or under unfavorable cellular conditions, misfolding of proteins generates common pathological events linked with neurodegenerative diseases and aging. Current advances of proteome studies systematically generates some progress in our knowledge that how misfolding of proteins or their accumulation can contribute to the impairment or depletion of proteome functions. Still, the underlying causes of this unrecoverable loss are not clear that how such unsolved transitions give rise to multifactorial challengeable degenerative pathological conditions in neurodegeneration. In this review, we specifically focus and systematically summarize various molecular mechanisms of proteostasis maintenance, as well as discuss progressing neurobiological strategies, promising natural and pharmacological candidates, which can be useful to counteract the problem of proteopathies. Our article emphasizes an urgent need that now it is important for us to recognize the fundamentals of proteostasis to design a new molecular framework and fruitful strategies to uncover how the proteome defects are associated with aging and neurodegenerative diseases. A enhance understanding of progress link with proteome and neurobiological challenges may provide new basic concepts in the near future, based on pharmacological agents, linked with impaired proteostasis and neurodegenerative diseases.


Asunto(s)
Enfermedades Neurodegenerativas/metabolismo , Proteoma/fisiología , Proteostasis/fisiología , Envejecimiento/genética , Envejecimiento/fisiología , Animales , Humanos , Neoplasias/metabolismo , Enfermedades Neurodegenerativas/genética , Proteoma/genética , Proteostasis/genética
14.
Front Mol Neurosci ; 10: 151, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28579943

RESUMEN

Cells regularly synthesize new proteins to replace old or damaged proteins. Deposition of various aberrant proteins in specific brain regions leads to neurodegeneration and aging. The cellular protein quality control system develop various defense mechanisms against the accumulation of misfolded and aggregated proteins. The mechanisms underlying the selective recognition of specific crucial protein or misfolded proteins are majorly governed by quality control E3 ubiquitin ligases mediated through ubiquitin-proteasome system. Few known E3 ubiquitin ligases have shown prominent neurodevelopmental functions, but their interactions with different developmental proteins play critical roles in neurodevelopmental disorders. Several questions are yet to be understood properly. How E3 ubiquitin ligases determine the specificity and regulate degradation of a particular substrate involved in neuronal proliferation and differentiation is certainly the one, which needs detailed investigations. Another important question is how neurodevelopmental E3 ubiquitin ligases specifically differentiate between their versatile range of substrates and timing of their functional modulations during different phases of development. The premise of this article is to understand how few E3 ubiquitin ligases sense major molecular events, which are crucial for human brain development from its early embryonic stages to throughout adolescence period. A better understanding of these few E3 ubiquitin ligases and their interactions with other potential proteins will provide invaluable insight into disease mechanisms to approach toward therapeutic interventions.

15.
J Cell Biochem ; 118(5): 1014-1027, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27487200

RESUMEN

Diclofenac is the most commonly used phenylacetic acid derivative non-steroidal anti-inflammatory drug (NSAID) that demonstrates significant analgesic, antipyretic, and anti-inflammatory effects. Several epidemiological studies have demonstrated anti-proliferative activity of NSAIDs and examined their apoptotic induction effects in different cancer cell lines. However, the precise molecular mechanisms by which these pharmacological agents induce apoptosis and exert anti-carcinogenic properties are not well known. Here, we have observed that diclofenac treatment induces proteasome malfunction and promotes accumulation of different critical proteasome substrates, including few pro-apoptotic proteins in cells. Exposure of diclofenac consequently elevates aggregation of various ubiquitylated misfolded proteins. Finally, we have shown that diclofenac treatment promotes apoptosis in cells, which could be because of mitochondrial membrane depolarization and cytochrome c release into cytosol. This study suggests possible beneficial insights of NSAIDs-induced apoptosis that may improve our existing knowledge in anti-proliferative interspecific strategies development. J. Cell. Biochem. 118: 1014-1027, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Antineoplásicos/farmacología , Diclofenaco/farmacología , Mitocondrias/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Células A549 , Animales , Apoptosis , Células COS , Supervivencia Celular/efectos de los fármacos , Chlorocebus aethiops , Potencial de la Membrana Mitocondrial/efectos de los fármacos
16.
Front Mol Neurosci ; 9: 93, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27757073

RESUMEN

Cells regularly synthesize new proteins to replace old and abnormal proteins for normal cellular functions. Two significant protein quality control pathways inside the cellular milieu are ubiquitin proteasome system (UPS) and autophagy. Autophagy is known for bulk clearance of cytoplasmic aggregated proteins, whereas the specificity of protein degradation by UPS comes from E3 ubiquitin ligases. Few E3 ubiquitin ligases, like C-terminus of Hsc70-interacting protein (CHIP) not only take part in protein quality control pathways, but also plays a key regulatory role in other cellular processes like signaling, development, DNA damage repair, immunity and aging. CHIP targets misfolded proteins for their degradation through proteasome, as well as autophagy; simultaneously, with the help of chaperones, it also regulates folding attempts for misfolded proteins. The broad range of CHIP substrates and their associations with multiple pathologies make it a key molecule to work upon and focus for future therapeutic interventions. E3 ubiquitin ligase CHIP interacts and degrades many protein inclusions formed in neurodegenerative diseases. The presence of CHIP at various nodes of cellular protein-protein interaction network presents this molecule as a potential candidate for further research. In this review, we have explored a wide range of functionality of CHIP inside cells by a detailed presentation of its co-chaperone, E3 and E4 enzyme like functions, with central focus on its protein quality control roles in neurodegenerative diseases. We have also raised many unexplored but expected fundamental questions regarding CHIP functions, which generate hopes for its future applications in research, as well as drug discovery.

17.
Neurobiol Dis ; 86: 16-28, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26607786

RESUMEN

Proteotoxicity of misfolded, disease-causing proteins is deeply implicated in the pathomechanisms for neurodegenerative diseases including copper-zinc superoxide dismutase (SOD1)-linked amyotrophic lateral sclerosis (ALS). However, the precise cellular quality control (QC) mechanisms against aggregation of misfolded mutant SOD1 proteins remain elusive. Here, we found that the Mahogunin ring finger-1 (MGRN1) E3 ubiquitin ligase, which catalyzes mono-ubiquitination to the substrate, was dysregulated in the cellular and mouse models of ALS and that it preferentially interacted with various mutant forms of SOD1. Intriguingly, the motor neurons of presymptomatic ALS mice have diminished MGRN1 cytoplasmic distribution. MGRN1 was partially recruited to mutant SOD1 inclusions where they were positive for p62 and Lamp2. Moreover, overexpression of MGRN1 reduced mutant SOD1 aggregation and alleviated its proteotoxic effects on cells. Taken together, our findings suggest that MGRN1 contributes to the clearance of toxic mutant SOD1 inclusions likely through autophagic pathway, and, most likely, the sequestration of MGRN1 sensitizes motor neurons to degeneration in the ALS mouse model. Furthermore, the present study identifies the MGRN1-mediated protein QC mechanism as a novel therapeutic target in neurodegenerative diseases.


Asunto(s)
Esclerosis Amiotrófica Lateral/enzimología , Esclerosis Amiotrófica Lateral/patología , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Superóxido Dismutasa/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Células COS , Supervivencia Celular , Chlorocebus aethiops , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Ratones , Ratones Transgénicos , Médula Espinal/metabolismo , Médula Espinal/patología , Superóxido Dismutasa/genética , Superóxido Dismutasa-1
18.
Mol Neurobiol ; 53(7): 4484-96, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26255182

RESUMEN

In healthy cell, inappropriate accumulation of poor or damaged proteins is prevented by cellular quality control system. Autophagy and ubiquitin proteasome system (UPS) provides regular cytoprotection against proteotoxicity induced by abnormal or disruptive proteins. E3 ubiquitin ligases are crucial components in this defense mechanism. Mahogunin Ring Finger-1 (MGRN1), an E3 ubiquitin ligase of the Really Interesting New Gene (RING) finger family, plays a pivotal role in many biological and cellular mechanisms. Previous findings indicate that lack of functions of MGRN1 can cause spongiform neurodegeneration, congenital heart defects, abnormal left-right patterning, and mitochondrial dysfunctions in mice brains. However, the detailed molecular pathomechanism of MGRN1 in cellular functions and diseases is not well known. This article comprehensively represents the molecular nature, characterization, and functions of MGRN1; we also summarize possible beneficiary aspects of this novel E3 ubiquitin ligase. Here, we review recent literature on the role of MGRN1 in the neuro-pathobiological mechanisms, with precise focus on the processes of neurodegeneration, and thereby propose new lines of potential targets for therapeutic intervention.


Asunto(s)
Neurobiología , Ubiquitina-Proteína Ligasas/metabolismo , Secuencia de Aminoácidos , Animales , Humanos , Modelos Biológicos , Enfermedades Neurodegenerativas/enzimología , Enfermedades Neurodegenerativas/patología , Pliegue de Proteína , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética
19.
Mol Neurobiol ; 53(10): 6968-6981, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-26666667

RESUMEN

In routine course of life, nonsteroidal anti-inflammatory drugs (NSAIDs) are widely prescribed antipyretic, analgesic, and anti-inflammatory drugs. It is a well-proposed notion that treatment of NSAIDs may induce anti-proliferative effects in numerous cancer cells. Ibuprofen from isobutylphenylpropanoic acid is NSAID and used to relieve fever, pain, and inflammation. It is also used for juvenile idiopathic arthritis, rheumatoid arthritis, patent ductus arteriosus, and for pericarditis. Despite few emerging studies have expanded the fundamental concept that the treatment of NSAIDs influences apoptosis in cancer cells, however the NSAID-mediated precise mechanisms that determine apoptosis induction without producing adverse consequences in variety of cancer cells are largely unknown. In our present study, we have observed that ibuprofen reduces proteasome activity, enhances the aggregation of ubiquitylated abnormal proteins, and also elevates the accumulation of crucial proteasome substrates. Ibuprofen treatment causes mitochondrial abnormalities and releases cytochrome c into cytosol. Perhaps, the more detailed study is needed in the future to elucidate the molecular mechanisms of NSAIDs that can induce apoptosis without adverse effects and produce effective anti-tumor effects and consequently help in neurodegeneration and ageing.


Asunto(s)
Apoptosis/efectos de los fármacos , Ibuprofeno/farmacología , Mitocondrias/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Células A549 , Animales , Células COS , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Forma del Núcleo Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Chlorocebus aethiops , Cromatina/metabolismo , Citocromos c/metabolismo , Fragmentación del ADN/efectos de los fármacos , Humanos , Cuerpos de Inclusión/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Inhibidor NF-kappaB alfa/metabolismo , Agregado de Proteínas/efectos de los fármacos , Pliegue de Proteína/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Especificidad por Sustrato/efectos de los fármacos , Factores de Tiempo , Proteínas Ubiquitinadas/metabolismo
20.
Ageing Res Rev ; 24(Pt B): 138-59, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26247845

RESUMEN

Efficient and regular performance of Ubiquitin Proteasome System and Autophagy continuously eliminate deleterious accumulation of nonnative protiens. In cellular quality control system, E3 ubiquitin ligases are significant employees for defense mechanism against abnormal toxic proteins. Few findings indicate that lack of functions of E3 ubiquitin ligases can be a causative factor of neurodevelopmental disorders, neurodegeneration, cancer and ageing. However, the detailed molecular pathomechanism implying E3 ubiquitin ligases in cellular functions in multifactorial disease conditions are not well understood. This article systematically represents the unique characteristics, molecular nature, and recent developments in the knowledge of neurobiological functions of few crucial E3 ubiquitin ligases. Here, we review recent literature on the roles of E6-AP, HRD1 and ITCH E3 ubiquitin ligases in the neuro-pathobiological mechanisms, with precise focus on the processes of neurodegeneration, and thereby propose new lines of potential targets for therapeutic interventions.


Asunto(s)
Envejecimiento/fisiología , Degeneración Nerviosa/metabolismo , Enfermedades Neurodegenerativas , Neuroprotección/fisiología , Ubiquitina-Proteína Ligasas , Humanos , Redes y Vías Metabólicas , Modelos Biológicos , Enfermedades Neurodegenerativas/enzimología , Enfermedades Neurodegenerativas/patología , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Ubiquitina-Proteína Ligasas/clasificación , Ubiquitina-Proteína Ligasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...