Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Vet Med Educ ; 48(3): 351-360, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32516075

RESUMO

Working as an equine veterinarian carries a high risk of occupational injury, with the behavior of the horse frequently reported as a cause for these injuries. Risk of injury is one reason cited by undergraduate veterinary students that would prevent them from entering large animal practice, and newly graduated veterinarians have been shown to be at increased risk of sustaining an occupational injury compared with more experienced colleagues. A cohort of pre-final-year undergraduate veterinary students were given a 45-minute lecture on learning theory and its application in equine practice, completing a questionnaire before (pre) and after (immediately [post] and several weeks [delayed post]) to investigate whether receiving a single lecture alters undergraduate veterinary students' perception of dealing with difficult horses in equine practice. The undergraduate veterinary students' attitudes to the behavior scenarios altered from the pre-questionnaire to the post- and delayed post-questionnaires. They were less likely to choose more traditional methods of restraining or controlling the horse (such as a twitch) and more likely to choose an option based on learning theory after the lecture. They also reported that if they had to deal with one of these scenarios in practice following the lecture, they would feel more confident, more likely to succeed in completing the intervention, and less likely to be injured. This study suggests that an educational intervention can help change the attitudes and confidence of undergraduate students when working with difficult horses.


Assuntos
Educação em Veterinária , Animais , Atitude , Currículo , Cavalos , Humanos , Estudantes , Inquéritos e Questionários
2.
J Biol Chem ; 294(18): 7296-7307, 2019 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-30877201

RESUMO

The E3 ubiquitin ligase parkin is a critical regulator of mitophagy and has been identified as a susceptibility gene for type 2 diabetes (T2D). However, its role in metabolically active tissues that precipitate T2D development is unknown. Specifically, pancreatic ß cells and adipocytes both rely heavily on mitochondrial function in the regulation of optimal glycemic control to prevent T2D, but parkin's role in preserving quality control of ß cell or adipocyte mitochondria is unclear. Although parkin has been reported previously to control mitophagy, here we show that, surprisingly, parkin is dispensable for glucose homeostasis in both ß cells and adipocytes during diet-induced insulin resistance in mice. We observed that insulin secretion, ß cell formation, and islet architecture were preserved in parkin-deficient ß cells and islets, suggesting that parkin is not necessary for control of ß cell function and islet compensation for diet-induced obesity. Although transient parkin deficiency mildly impaired mitochondrial turnover in ß cell lines, parkin deletion in primary ß cells yielded no deficits in mitochondrial clearance. In adipocyte-specific deletion models, lipid uptake and ß-oxidation were increased in cultured cells, whereas adipose tissue morphology, glucose homeostasis, and beige-to-white adipocyte transition were unaffected in vivo In key metabolic tissues where mitochondrial dysfunction has been implicated in T2D development, our experiments unexpectedly revealed that parkin is not an essential regulator of glucose tolerance, whole-body energy metabolism, or mitochondrial quality control. These findings highlight that parkin-independent processes maintain ß cell and adipocyte mitochondrial quality control in diet-induced obesity.


Assuntos
Adipócitos/metabolismo , Homeostase , Células Secretoras de Insulina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Adipócitos/citologia , Adipócitos/enzimologia , Adiposidade , Animais , Peso Corporal , Diferenciação Celular , Diabetes Mellitus Tipo 2/metabolismo , Metabolismo Energético , Feminino , Teste de Tolerância a Glucose , Resistência à Insulina , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/enzimologia , Masculino , Camundongos , Mitocôndrias/metabolismo , Oxirredução
3.
Diabetologia ; 57(1): 129-39, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24149836

RESUMO

AIMS/HYPOTHESIS: Lipolytic breakdown of endogenous lipid pools in pancreatic beta cells contributes to glucose-stimulated insulin secretion (GSIS) and is thought to be mediated by acute activation of neutral lipases in the amplification pathway. Recently it has been shown in other cell types that endogenous lipid can be metabolised by autophagy, and this lipophagy is catalysed by lysosomal acid lipase (LAL). This study aimed to elucidate a role for LAL and lipophagy in pancreatic beta cells. METHODS: We employed pharmacological and/or genetic inhibition of autophagy and LAL in MIN6 cells and primary islets. Insulin secretion following inhibition was measured using RIA. Lipid accumulation was assessed by MS and confocal microscopy (to visualise lipid droplets) and autophagic flux was analysed by western blot. RESULTS: Insulin secretion was increased following chronic (≥ 8 h) inhibition of LAL. This was more pronounced with glucose than with non-nutrient stimuli and was accompanied by augmentation of neutral lipid species. Similarly, following inhibition of autophagy in MIN6 cells, the number of lipid droplets was increased and GSIS was potentiated. Inhibition of LAL or autophagy in primary islets also increased insulin secretion. This augmentation of GSIS following LAL or autophagy inhibition was dependent on the acute activation of neutral lipases. CONCLUSIONS/INTERPRETATION: Our data suggest that lysosomal lipid degradation, using LAL and potentially lipophagy, contributes to neutral lipid turnover in beta cells. It also serves as a constitutive negative regulator of GSIS by depletion of substrate for the non-lysosomal neutral lipases that are activated acutely by glucose.


Assuntos
Glucose/farmacologia , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Animais , Linhagem Celular Tumoral , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Camundongos , Microscopia Confocal , Esterol Esterase
4.
Vet Rec ; 194 Suppl 1: 9, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38700153

RESUMO

This year BVA Live will, for the first time, feature farm animal and equine CPD. Among the varied topics on offer, Gemma Pearson will be presenting an introduction to equine behavioural medicine, examining the link between emotional and physical health in horses.


Assuntos
Comportamento Animal , Dor , Animais , Cavalos/psicologia , Dor/veterinária , Dor/psicologia , Medicina Veterinária , Doenças dos Cavalos/psicologia , Humanos , Reino Unido
5.
JCI Insight ; 9(15)2024 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-38935435

RESUMO

ER stress and proinsulin misfolding are heralded as contributing factors to ß cell dysfunction in type 2 diabetes, yet how ER function becomes compromised is not well understood. Recent data identify altered ER redox homeostasis as a critical mechanism that contributes to insulin granule loss in diabetes. Hyperoxidation of the ER delays proinsulin export and limits the proinsulin supply available for insulin granule formation. In this report, we identified glucose metabolism as a critical determinant in the redox homeostasis of the ER. Using multiple ß cell models, we showed that loss of mitochondrial function or inhibition of cellular metabolism elicited ER hyperoxidation and delayed ER proinsulin export. Our data further demonstrated that ß cell ER redox homeostasis was supported by the metabolic supply of reductive redox donors. We showed that limiting NADPH and thioredoxin flux delayed ER proinsulin export, whereas thioredoxin-interacting protein suppression restored ER redox and proinsulin trafficking. Taken together, we propose that ß cell ER redox homeostasis is buffered by cellular redox donor cycles, which are maintained through active glucose metabolism.


Assuntos
Retículo Endoplasmático , Glucose , Células Secretoras de Insulina , Oxirredução , Proinsulina , Tiorredoxinas , Proinsulina/metabolismo , Células Secretoras de Insulina/metabolismo , Animais , Retículo Endoplasmático/metabolismo , Camundongos , Humanos , Glucose/metabolismo , Tiorredoxinas/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Mitocôndrias/metabolismo , Estresse do Retículo Endoplasmático , Homeostase , Insulina/metabolismo , NADP/metabolismo , Transporte Proteico
6.
bioRxiv ; 2024 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-39211191

RESUMO

Pluripotent stem cell (SC)-derived islets offer hope as a renewable source for ß cell replacement for type 1 diabetes (T1D), yet functional and metabolic immaturity may limit their long-term therapeutic potential. Here, we show that limitations in mitochondrial transcriptional programming impede the formation and maturation of SC-derived ß (SC-ß) cells. Utilizing transcriptomic profiling, assessments of chromatin accessibility, mitochondrial phenotyping, and lipidomics analyses, we observed that SC-ß cells exhibit reduced oxidative and mitochondrial fatty acid metabolism compared to primary human islets that are related to limitations in key mitochondrial transcriptional networks. Surprisingly, we found that reductions in glucose- stimulated mitochondrial respiration in SC-islets were not associated with alterations in mitochondrial mass, structure, or genome integrity. In contrast, SC-islets show limited expression of targets of PPARIZ and PPARγ, which regulate mitochondrial programming, yet whose functions in ß cell differentiation are unknown. Importantly, treatment with WY14643, a potent PPARIZ agonist, induced expression of mitochondrial targets, improved insulin secretion, and increased the formation and maturation of SC-ß cells both in vitro and following transplantation. Thus, mitochondrial programming promotes the differentiation and maturation of SC-ß cells and may be a promising target to improve ß cell replacement efforts for T1D.

7.
Nat Microbiol ; 9(4): 949-963, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38528148

RESUMO

A polymorphism causing deficiencies in Toll-interacting protein (TOLLIP), an inhibitory adaptor protein affecting endosomal trafficking, is associated with increased tuberculosis (TB) risk. It is, however, unclear how TOLLIP affects TB pathogenesis. Here we show that TB severity is increased in Tollip-/- mice, characterized by macrophage- and T cell-driven inflammation, foam cell formation and lipid accumulation. Tollip-/- alveolar macrophages (AM) specifically accumulated lipid and underwent necrosis. Transcriptional and protein analyses of Mycobacterium tuberculosis (Mtb)-infected, Tollip-/- AM revealed increased EIF2 signalling and downstream upregulation of the integrated stress response (ISR). These phenotypes were linked, as incubation of the Mtb lipid mycolic acid with Mtb-infected Tollip-/- AM activated the ISR and increased Mtb replication. Correspondingly, the ISR inhibitor, ISRIB, reduced Mtb numbers in AM and improved Mtb control, overcoming the inflammatory phenotype. In conclusion, targeting the ISR offers a promising target for host-directed anti-TB therapy towards improved Mtb control and reduced immunopathology.


Assuntos
Mycobacterium tuberculosis , Tuberculose , Animais , Camundongos , Macrófagos Alveolares/microbiologia , Tuberculose/microbiologia , Mycobacterium tuberculosis/fisiologia , Macrófagos/microbiologia , Lipídeos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo
8.
Animals (Basel) ; 13(19)2023 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-37835743

RESUMO

The 2023 Grand National steeplechase race was delayed when protesters from the animal rights group, 'Animal Rising', gained access to the course just prior to the race. The international media spotlight was focused on what is already a high-profile event and the social licence of both this race and racing in general was scrutinised. Both at the time and for several days afterwards, the general public was exposed to two different narratives from pro- and anti-racing communities. This paper discusses these perspectives and the potential impact on the general public's relationship with racing. Whilst well-meaning and aiming to promote racing, much of the racing industry's commentary inadvertently risked damaging its reputation due to a poor understanding of social licence principles. We explore the reasons for these two groups' alternative perspectives on welfare and suggest considerations for change. Ultimately, if 'the people's race' is to maintain its social licence, the racing community needs to both understand and embrace the concept. Welcoming independent opinions, engaging with different viewpoints, accepting that change is inevitable and, most importantly, being proactive in making changes to prioritise equine welfare will all help racing to move towards greater public acceptance.

9.
Animals (Basel) ; 13(4)2023 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-36830535

RESUMO

Equestrianism is currently facing a range of pressing challenges. These challenges, which are largely based on evolving attitudes to ethics and equine wellbeing, have consequences for the sport's social licence to operate. The factors that may have contributed to the current situation include overarching societal trends, specific aspects of the equestrian sector, and factors rooted in human nature. If equestrianism is to flourish, it is evident that much needs to change, not the least, human behaviour. To this end, using established behaviour change frameworks that have been scientifically validated and are rooted in practice-most notably, Michie et al.'s COM-B model and Behaviour Change Wheel-could be of practical value for developing and implementing equine welfare strategies. This review summarises the theoretical underpinnings of some behaviour change frameworks and provides a practical, step-by-step approach to designing an effective behaviour change intervention. A real-world example is provided through the retrospective analysis of an intervention strategy that aimed to increase the use of learning theory in (educational) veterinary practice. We contend that the incorporation of effective behaviour change interventions into any equine welfare improvement strategy may help to safeguard the future of equestrianism.

10.
Autophagy ; 19(2): 525-543, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-35604110

RESUMO

CLEC16A regulates mitochondrial health through mitophagy and is associated with over 20 human diseases. However, the key structural and functional regions of CLEC16A, and their relevance for human disease, remain unknown. Here, we report that a disease-associated CLEC16A variant lacks a C-terminal intrinsically disordered protein region (IDPR) that is critical for mitochondrial quality control. IDPRs comprise nearly half of the human proteome, yet their mechanistic roles in human disease are poorly understood. Using carbon detect NMR, we find that the CLEC16A C terminus lacks secondary structure, validating the presence of an IDPR. Loss of the CLEC16A C-terminal IDPR in vivo impairs mitophagy, mitochondrial function, and glucose-stimulated insulin secretion, ultimately causing glucose intolerance. Deletion of the CLEC16A C-terminal IDPR increases CLEC16A ubiquitination and degradation, thus impairing assembly of the mitophagy regulatory machinery. Importantly, CLEC16A stability is dependent on proline bias within the C-terminal IDPR, but not amino acid sequence order or charge. Together, we elucidate how an IDPR in CLEC16A regulates mitophagy and implicate pathogenic human gene variants that disrupt IDPRs as novel contributors to diabetes and other CLEC16A-associated diseases.Abbreviations : CAS: carbon-detect amino-acid specific; IDPR: intrinsically disordered protein region; MEFs: mouse embryonic fibroblasts; NMR: nuclear magnetic resonance.


Assuntos
Proteínas Intrinsicamente Desordenadas , Mitofagia , Humanos , Animais , Camundongos , Mitofagia/genética , Proteínas Intrinsicamente Desordenadas/genética , Proteínas Intrinsicamente Desordenadas/química , Proteínas Intrinsicamente Desordenadas/metabolismo , Autofagia , Fibroblastos/metabolismo , Ubiquitinação , Proteínas de Transporte de Monossacarídeos/metabolismo , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo
11.
Nat Commun ; 13(1): 2340, 2022 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-35487893

RESUMO

The dynamin-like GTPases Mitofusin 1 and 2 (Mfn1 and Mfn2) are essential for mitochondrial function, which has been principally attributed to their regulation of fission/fusion dynamics. Here, we report that Mfn1 and 2 are critical for glucose-stimulated insulin secretion (GSIS) primarily through control of mitochondrial DNA (mtDNA) content. Whereas Mfn1 and Mfn2 individually were dispensable for glucose homeostasis, combined Mfn1/2 deletion in ß-cells reduced mtDNA content, impaired mitochondrial morphology and networking, and decreased respiratory function, ultimately resulting in severe glucose intolerance. Importantly, gene dosage studies unexpectedly revealed that Mfn1/2 control of glucose homeostasis was dependent on maintenance of mtDNA content, rather than mitochondrial structure. Mfn1/2 maintain mtDNA content by regulating the expression of the crucial mitochondrial transcription factor Tfam, as Tfam overexpression ameliorated the reduction in mtDNA content and GSIS in Mfn1/2-deficient ß-cells. Thus, the primary physiologic role of Mfn1 and 2 in ß-cells is coupled to the preservation of mtDNA content rather than mitochondrial architecture, and Mfn1 and 2 may be promising targets to overcome mitochondrial dysfunction and restore glucose control in diabetes.


Assuntos
DNA Mitocondrial , Mitocôndrias , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Glucose/metabolismo , Homeostase , Mitocôndrias/metabolismo
12.
Diabetes ; 70(6): 1229-1241, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34016598

RESUMO

Insulin-producing pancreatic ß-cells are central to glucose homeostasis, and their failure is a principal driver of diabetes development. To preserve optimal health ß-cells must withstand both intrinsic and extrinsic stressors, ranging from inflammation to increased peripheral insulin demand, in addition to maintaining insulin biosynthesis and secretory machinery. Autophagy is increasingly being appreciated as a critical ß-cell quality control system vital for glycemic control. Here we focus on the underappreciated, yet crucial, roles for selective and organelle-specific forms of autophagy as mediators of ß-cell health. We examine the unique molecular players underlying each distinct form of autophagy in ß-cells, including selective autophagy of mitochondria, insulin granules, lipid, intracellular amyloid aggregates, endoplasmic reticulum, and peroxisomes. We also describe how defects in selective autophagy pathways contribute to the development of diabetes. As all forms of autophagy are not the same, a refined view of ß-cell selective autophagy may inform new approaches to defend against the various insults leading to ß-cell failure in diabetes.


Assuntos
Autofagia/fisiologia , Células Secretoras de Insulina/fisiologia , Animais , Diabetes Mellitus/etiologia , Diabetes Mellitus/patologia , Diabetes Mellitus/fisiopatologia , Humanos , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Ilhotas Pancreáticas/fisiopatologia , Mitofagia/fisiologia , Agregados Proteicos/fisiologia , Fatores de Transcrição/fisiologia , Ubiquitina-Proteína Ligases/fisiologia
13.
JCI Insight ; 5(24)2020 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-33232298

RESUMO

Inflammatory damage contributes to ß cell failure in type 1 and 2 diabetes (T1D and T2D, respectively). Mitochondria are damaged by inflammatory signaling in ß cells, resulting in impaired bioenergetics and initiation of proapoptotic machinery. Hence, the identification of protective responses to inflammation could lead to new therapeutic targets. Here, we report that mitophagy serves as a protective response to inflammatory stress in both human and rodent ß cells. Utilizing in vivo mitophagy reporters, we observed that diabetogenic proinflammatory cytokines induced mitophagy in response to nitrosative/oxidative mitochondrial damage. Mitophagy-deficient ß cells were sensitized to inflammatory stress, leading to the accumulation of fragmented dysfunctional mitochondria, increased ß cell death, and hyperglycemia. Overexpression of CLEC16A, a T1D gene and mitophagy regulator whose expression in islets is protective against T1D, ameliorated cytokine-induced human ß cell apoptosis. Thus, mitophagy promotes ß cell survival and prevents diabetes by countering inflammatory injury. Targeting this pathway has the potential to prevent ß cell failure in diabetes and may be beneficial in other inflammatory conditions.


Assuntos
Células Secretoras de Insulina/metabolismo , Lectinas Tipo C/metabolismo , Mitofagia/fisiologia , Proteínas de Transporte de Monossacarídeos/metabolismo , Animais , Apoptose , Sobrevivência Celular , Complicações do Diabetes , Diabetes Mellitus/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Inflamação/metabolismo , Células Secretoras de Insulina/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Estresse Oxidativo , Cultura Primária de Células , Substâncias Protetoras/metabolismo , Transdução de Sinais
14.
J Vis Exp ; (147)2019 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-31107439

RESUMO

Mitophagy is an essential mitochondrial quality control pathway, which is crucial for pancreatic islet beta cell bioenergetics to fuel glucose-stimulated insulin release. Assessment of mitophagy is challenging and often requires genetic reporters or multiple complementary techniques not easily utilized in tissue samples, such as primary human pancreatic islets. Here we demonstrate a robust approach to visualize and quantify formation of key endogenous mitophagy complexes in primary human pancreatic islets. Utilizing the sensitive proximity ligation assay technique to detect interaction of the mitophagy regulators NRDP1 and USP8, we are able to specifically quantify formation of essential mitophagy complexes in situ. By coupling this approach to counterstaining for the transcription factor PDX1, we can quantify mitophagy complexes, and the factors that can impair mitophagy, specifically within beta cells. The methodology we describe overcomes the need for large quantities of cellular extracts required for other protein-protein interaction studies, such as immunoprecipitation (IP) or mass spectrometry, and is ideal for precious human islet samples generally not available in sufficient quantities for these approaches. Further, this methodology obviates the need for flow sorting techniques to purify beta cells from a heterogeneous islet population for downstream protein applications. Thus, we describe a valuable protocol for visualization of mitophagy highly compatible for use in heterogeneous and limited cell populations.


Assuntos
Células Secretoras de Insulina/citologia , Mitofagia , Animais , Endopeptidases/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Metabolismo Energético , Regulação da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Humanos , Insulina/metabolismo , Mitocôndrias/metabolismo , Transativadores/metabolismo , Ubiquitina Tiolesterase/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
15.
Autophagy ; 14(7): 1160-1161, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29799764

RESUMO

Mitochondrial autophagy or mitophagy is a key component of mitochondrial quality control, which is necessary to maintain cellular bioenergetics. Pancreatic islet ß-cells, which release insulin in response to circulating blood glucose levels, are particularly susceptible to mitochondrial dysfunction due to their high metabolic activity and energy requirements for insulin processing, maturation, and secretion. Therefore, dysregulated mitophagy has drawn interest in the etiology of ß-cell failure in diabetes. We demonstrate that the pivotal ß-cell mitophagy regulator, CLEC16A, is an E3 ligase that forms a ubiquitin-dependent tripartite complex with RNF41/NRDP1 and USP8. Maintenance of the CLEC16A-RNF41-USP8 mitophagy complex is necessary for maximal cellular respiration and insulin secretion. Further, we observe that diabetogenic metabolic stressors, including elevated glucose and fatty acids, destabilize the CLEC16A-RNF41-USP8 complex and lead to ß-cell apoptosis. Thus, the ß-cell mitophagy pathway requires ubiquitin signals to stabilize the CLEC16A-RNF41-USP8 complex and maintain mitochondrial quality control.


Assuntos
Autofagia , Mitofagia , Secreção de Insulina , Mitocôndrias , Ubiquitina , Ubiquitina-Proteína Ligases
16.
Diabetes ; 67(2): 265-277, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29180353

RESUMO

Mitophagy is a cellular quality-control pathway, which is essential for elimination of unhealthy mitochondria. While mitophagy is critical to pancreatic ß-cell function, the posttranslational signals governing ß-cell mitochondrial turnover are unknown. Here, we report that ubiquitination is essential for the assembly of a mitophagy regulatory complex, comprised of the E3 ligase Nrdp1, the deubiquitinase enzyme USP8, and Clec16a, a mediator of ß-cell mitophagy with unclear function. We discover that the diabetes gene Clec16a encodes an E3 ligase, which promotes nondegradative ubiquitin conjugates to direct its mitophagy effectors and stabilize the Clec16a-Nrdp1-USP8 complex. Inhibition of the Clec16a pathway by the chemotherapeutic lenalidomide, a selective ubiquitin ligase inhibitor associated with new-onset diabetes, impairs ß-cell mitophagy, oxygen consumption, and insulin secretion. Indeed, patients treated with lenalidomide develop compromised ß-cell function. Moreover, the ß-cell Clec16a-Nrdp1-USP8 mitophagy complex is destabilized and dysfunctional after lenalidomide treatment as well as after glucolipotoxic stress. Thus, the Clec16a-Nrdp1-USP8 complex relies on ubiquitin signals to promote mitophagy and maintain mitochondrial quality control necessary for optimal ß-cell function.


Assuntos
Endopeptidases/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Células Secretoras de Insulina/metabolismo , Lectinas Tipo C/metabolismo , Mitofagia , Proteínas de Transporte de Monossacarídeos/metabolismo , Ubiquitina Tiolesterase/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Inibidores da Angiogênese/farmacologia , Animais , Linhagem Celular , Células Cultivadas , Cruzamentos Genéticos , Endopeptidases/química , Endopeptidases/genética , Complexos Endossomais de Distribuição Requeridos para Transporte/antagonistas & inibidores , Complexos Endossomais de Distribuição Requeridos para Transporte/química , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Inibidores Enzimáticos/farmacologia , Glucose/metabolismo , Humanos , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Lectinas Tipo C/antagonistas & inibidores , Lectinas Tipo C/química , Lectinas Tipo C/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mitofagia/efeitos dos fármacos , Proteínas de Transporte de Monossacarídeos/antagonistas & inibidores , Proteínas de Transporte de Monossacarídeos/química , Proteínas de Transporte de Monossacarídeos/genética , Multimerização Proteica/efeitos dos fármacos , Estabilidade Proteica/efeitos dos fármacos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Bancos de Tecidos , Técnicas de Cultura de Tecidos , Ubiquitina Tiolesterase/antagonistas & inibidores , Ubiquitina Tiolesterase/química , Ubiquitina Tiolesterase/genética , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/genética , Ubiquitinação/efeitos dos fármacos
17.
Mol Metab ; 5(6): 404-414, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27257600

RESUMO

OBJECTIVE: Glucose promotes lipid remodelling in pancreatic ß-cells, and this is thought to contribute to the regulation of insulin secretion, but the metabolic pathways and potential signalling intermediates have not been fully elaborated. METHODS: Using mass spectrometry (MS) we quantified changes in approximately 300 lipid metabolites in MIN6 ß-cells and isolated mouse islets following 1 h stimulation with glucose. Flux through sphingolipid pathways was also assessed in (3)H-sphinganine-labelled cells using TLC. RESULTS: Glucose specifically activates the conversion of triacylglycerol (TAG) to diacylglycerol (DAG). This leads indirectly to the formation of 18:1 monoacylglycerol (MAG), via degradation of saturated/monounsaturated DAG species, such as 16:0_18:1 DAG, which are the most abundant, immediate products of glucose-stimulated TAG hydrolysis. However, 16:0-containing, di-saturated DAG species are a better direct marker of TAG hydrolysis since, unlike the 18:1-containing DAGs, they are predominately formed via this route. Using multiple reaction monitoring, we confirmed that in islets under basal conditions, 18:1 MAG is the most abundant species. We further demonstrated a novel site of glucose to enhance the conversion of ceramide to sphingomyelin (SM) and galactosylceramide (GalCer). Flux and product:precursor analyses suggest regulation of the enzyme SM synthase, which would constitute a separate mechanism for localized generation of DAG in response to glucose. Phosphatidylcholine (PC) plasmalogen (P) species, specifically those containing 20:4, 22:5 and 22:6 side chains, were also diminished in the presence of glucose, whereas the more abundant phosphatidylethanolamine plasmalogens were unchanged. CONCLUSION: Our results highlight 18:1 MAG, GalCer, PC(P) and DAG/SM as potential contributors to metabolic stimulus-secretion coupling.

18.
Vet J ; 202(3): 533-5, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25241949

RESUMO

Expression of several putative markers of pluripotency (OCT4, SOX2, NANOG, LIN28A, REX1, DNMT3B and TERT) was examined in a range of equine tissues, including early embryos, induced pluripotent stem cells (iPSCs), testis, adipose- and bone marrow-derived mesenchymal stromal cells (MSCs), and keratinocytes. Transcript levels of all markers were highest in embryos and iPSCs and, except for SOX2, were very low or undetectable in keratinocytes. Mean expression levels of all markers were lower in testis than in embryos or iPSCs and, except for DNMT3B, were higher in testis than in MSCs. Expression of OCT4, NANOG and DNMT3B, but not the other markers, was detected in MSCs. Of all markers analysed, only LIN28A, REX1 and TERT were associated exclusively with pluripotent cells in the horse.


Assuntos
Diferenciação Celular , Regulação da Expressão Gênica no Desenvolvimento , Cavalos/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Queratinócitos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Testículo/metabolismo , Animais , Biomarcadores/metabolismo , Embrião de Mamíferos/embriologia , Desenvolvimento Embrionário , Cavalos/embriologia , Cavalos/metabolismo , Masculino , Especificidade de Órgãos
19.
Diabetes ; 58(8): 1826-34, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19401415

RESUMO

OBJECTIVE: Insufficient insulin secretion is a hallmark of type 2 diabetes, and exposure of beta-cells to elevated lipid levels (lipotoxicity) contributes to secretory dysfunction. Functional ablation of protein kinase C epsilon (PKCepsilon) has been shown to improve glucose homeostasis in models of type 2 diabetes and, in particular, to enhance glucose-stimulated insulin secretion (GSIS) after lipid exposure. Therefore, we investigated the lipid-dependent mechanisms responsible for the enhanced GSIS after inactivation of PKCepsilon. RESEARCH DESIGN AND METHODS: We cultured islets isolated from PKCepsilon knockout (PKCepsilonKO) mice in palmitate prior to measuring GSIS, Ca(2+) responses, palmitate esterification products, lipolysis, lipase activity, and gene expression. RESULTS: The enhanced GSIS could not be explained by increased expression of another PKC isoform or by alterations in glucose-stimulated Ca(2+) influx. Instead, an upregulation of the amplifying pathways of GSIS in lipid-cultured PKCepsilonKO beta-cells was revealed under conditions in which functional ATP-sensitive K(+) channels were bypassed. Furthermore, we showed increased esterification of palmitate into triglyceride pools and an enhanced rate of lipolysis and triglyceride lipase activity in PKCepsilonKO islets. Acute treatment with the lipase inhibitor orlistat blocked the enhancement of GSIS in lipid-cultured PKCepsilonKO islets, suggesting that a lipolytic product mediates the enhancement of glucose-amplified insulin secretion after PKCepsilon deletion. CONCLUSIONS: Our findings demonstrate a mechanistic link between lipolysis and the amplifying pathways of GSIS in murine beta-cells, and they suggest an interaction between PKCepsilon and lipolysis. These results further highlight the therapeutic potential of PKCepsilon inhibition to enhance GSIS from the beta-cell under conditions of lipid excess.


Assuntos
Glucose/farmacologia , Células Secretoras de Insulina/fisiologia , Insulina/metabolismo , Lipólise/fisiologia , Proteína Quinase C-épsilon/deficiência , Animais , Cruzamentos Genéticos , Feminino , Deleção de Genes , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Lipólise/genética , Masculino , Camundongos , Camundongos Knockout , Proteína Quinase C-épsilon/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA