Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Am J Physiol Endocrinol Metab ; 326(3): E245-E257, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38265287

RESUMO

Delayed Golgi export of proinsulin has recently been identified as an underlying mechanism leading to insulin granule loss and ß-cell secretory defects in type 2 diabetes (T2D). Because acidification of the Golgi lumen is critical for proinsulin sorting and delivery into the budding secretory granule, we reasoned that dysregulation of Golgi pH may contribute to proinsulin trafficking defects. In this report, we examined pH regulation of the Golgi and identified a partial alkalinization of the Golgi lumen in a diabetes model. To further explore this, we generated a ß-cell specific knockout (KO) of the v0a2 subunit of the v-ATPase pump, which anchors the v-ATPase to the Golgi membrane. Although loss of v0a2 partially neutralized Golgi pH and was accompanied by distension of the Golgi cisternae, proinsulin export from the Golgi and insulin granule formation were not affected. Furthermore, ß-cell function was well preserved. ß-cell v0a2 KO mice exhibited normal glucose tolerance in both sexes, no genotypic difference to diet-induced obesity, and normal insulin secretory responses. Collectively, our data demonstrate the v0a2 subunit contributes to ß-cell Golgi pH regulation but suggest that additional disturbances to Golgi structure and function contribute to proinsulin trafficking defects in diabetes.NEW & NOTEWORTHY Delayed proinsulin export from the Golgi in diabetic ß-cells contributes to decreased insulin granule formation, but the underlying mechanisms are not clear. Here, we explored if dysregulation of Golgi pH can alter Golgi function using ß-cell specific knockout (KO) of the Golgi-localized subunit of the v-ATPase, v0a2. We show that partial alkalinization of the Golgi dilates the cisternae, but does not affect proinsulin export, insulin granule formation, insulin secretion, or glucose homeostasis.


Assuntos
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Animais , Feminino , Masculino , Camundongos , Adenosina Trifosfatases , Diabetes Mellitus Tipo 2/genética , Glucose , Insulina , Proinsulina/genética
2.
Proc Natl Acad Sci U S A ; 113(20): 5622-7, 2016 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-27140606

RESUMO

Melanin is responsible for pigmentation of skin and hair and is synthesized in a specialized organelle, the melanosome, in melanocytes. A genome-wide association study revealed that the two pore segment channel 2 (TPCN2) gene is strongly linked to pigmentation variations. TPCN2 encodes the two-pore channel 2 (TPC2) protein, a cation channel. Nevertheless, how TPC2 regulates pigmentation remains unknown. Here, we show that TPC2 is expressed in melanocytes and localizes to the melanosome-limiting membrane and, to a lesser extent, to endolysosomal compartments by confocal fluorescence and immunogold electron microscopy. Immunomagnetic isolation of TPC2-containing organelles confirmed its coresidence with melanosomal markers. TPCN2 knockout by means of clustered regularly interspaced short palindromic repeat/CRISPR-associated 9 gene editing elicited a dramatic increase in pigment content in MNT-1 melanocytic cells. This effect was rescued by transient expression of TPC2-GFP. Consistently, siRNA-mediated knockdown of TPC2 also caused a substantial increase in melanin content in both MNT-1 cells and primary human melanocytes. Using a newly developed genetically encoded pH sensor targeted to melanosomes, we determined that the melanosome lumen in TPC2-KO MNT-1 cells and primary melanocytes subjected to TPC2 knockdown is less acidic than in control cells. Fluorescence and electron microscopy analysis revealed that TPC2-KO MNT-1 cells have significantly larger melanosomes than control cells, but the number of organelles is unchanged. TPC2 likely regulates melanosomes pH and size by mediating Ca(2+) release from the organelle, which is decreased in TPC2-KO MNT-1 cells, as determined with the Ca(2+) sensor tyrosinase-GCaMP6. Thus, our data show that TPC2 regulates pigmentation through two fundamental determinants of melanosome function: pH and size.


Assuntos
Canais de Cálcio/fisiologia , Tamanho Celular , Melanossomas/metabolismo , Pigmentação , Cálcio/metabolismo , Canais de Cálcio/análise , Humanos , Concentração de Íons de Hidrogênio , Melaninas/análise , Melanossomas/química
3.
Front Pharmacol ; 14: 1339518, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38269286

RESUMO

pH-sensitive fluorescent proteins have revolutionized the field of cellular imaging and physiology, offering insight into the dynamic pH changes that underlie fundamental cellular processes. This comprehensive review explores the diverse applications and recent advances in the use of pH-sensitive fluorescent proteins. These remarkable tools enable researchers to visualize and monitor pH variations within subcellular compartments, especially mitochondria, shedding light on organelle-specific pH regulation. They play pivotal roles in visualizing exocytosis and endocytosis events in synaptic transmission, monitoring cell death and apoptosis, and understanding drug effects and disease progression. Recent advancements have led to improved photostability, pH specificity, and subcellular targeting, enhancing their utility. Techniques for multiplexed imaging, three-dimensional visualization, and super-resolution microscopy are expanding the horizon of pH-sensitive protein applications. The future holds promise for their integration into optogenetics and drug discovery. With their ever-evolving capabilities, pH-sensitive fluorescent proteins remain indispensable tools for unravelling cellular dynamics and driving breakthroughs in biological research. This review serves as a comprehensive resource for researchers seeking to harness the potential of pH-sensitive fluorescent proteins.

4.
Methods Enzymol ; 654: 315-344, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34120720

RESUMO

Melanocytes are specialized cells that produce melanin pigments responsible for skin, hair, and eye pigmentation. The synthesis and storage of melanin occurs in unique lysosome-related organelles called melanosomes, which regulate melanin production via complex regulatory mechanisms. Maintenance of the melanosome luminal ionic environment and pH is crucial for proper function of the main melanogenic enzymes. Defects in genes encoding pH-regulating melanosomal proteins result in oculocutaneous albinism, which is characterized by hypopigmentation, impaired vision, and increased susceptibility to skin and eye cancers. We recently uncovered several ion channels and transporters that modulate melanin synthesis by acidifying or neutralizing the luminal pH of melanosomes. However, our understanding of how melanosomes and other related organelles maintain their luminal pH is far from complete. The study of melanosome pH regulation requires robust imaging and quantification tools. Despite recent advances in the development of such methods, many limitations remain, particularly for quantitative analysis of individual organelle pH. In this chapter, we will provide an overview of the available methods used for melanosome pH determination, including their advantages, limitations, and challenges. To address the critical, unmet need for reliable melanosome pH quantification tools, we engineered a novel genetically encoded, ratiometric pH sensor for melanosomes that we named RpHiMEL. Here, we describe the design and optimization of RpHiMEL, and provide a pH quantification method for individual melanosomes in live cells. We demonstrate that RpHiMEL is a highly versatile tool with the potential to advance our understanding of pH regulation in melanosomes and related organelles.


Assuntos
Melanócitos , Melanossomas , Concentração de Íons de Hidrogênio , Melaninas , Pigmentação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA