Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 592(7854): 428-432, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33790465

RESUMO

Chronic, sustained exposure to stressors can profoundly affect tissue homeostasis, although the mechanisms by which these changes occur are largely unknown. Here we report that the stress hormone corticosterone-which is derived from the adrenal gland and is the rodent equivalent of cortisol in humans-regulates hair follicle stem cell (HFSC) quiescence and hair growth in mice. In the absence of systemic corticosterone, HFSCs enter substantially more rounds of the regeneration cycle throughout life. Conversely, under chronic stress, increased levels of corticosterone prolong HFSC quiescence and maintain hair follicles in an extended resting phase. Mechanistically, corticosterone acts on the dermal papillae to suppress the expression of Gas6, a gene that encodes the secreted factor growth arrest specific 6. Restoring Gas6 expression overcomes the stress-induced inhibition of HFSC activation and hair growth. Our work identifies corticosterone as a systemic inhibitor of HFSC activity through its effect on the niche, and demonstrates that the removal of such inhibition drives HFSCs into frequent regeneration cycles, with no observable defects in the long-term.


Assuntos
Corticosterona/farmacologia , Folículo Piloso/citologia , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Glândulas Suprarrenais/metabolismo , Glândulas Suprarrenais/cirurgia , Adrenalectomia , Animais , Divisão Celular/efeitos dos fármacos , Feminino , Folículo Piloso/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estresse Psicológico/metabolismo , Estresse Psicológico/patologia , Transcriptoma , Regulação para Cima
2.
Nature ; 577(7792): 676-681, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31969699

RESUMO

Empirical and anecdotal evidence has associated stress with accelerated hair greying (formation of unpigmented hairs)1,2, but so far there has been little scientific validation of this link. Here we report that, in mice, acute stress leads to hair greying through the fast depletion of melanocyte stem cells. Using a combination of adrenalectomy, denervation, chemogenetics3,4, cell ablation and knockout of the adrenergic receptor specifically in melanocyte stem cells, we find that the stress-induced loss of melanocyte stem cells is independent of immune attack or adrenal stress hormones. Instead, hair greying results from activation of the sympathetic nerves that innervate the melanocyte stem-cell niche. Under conditions of stress, the activation of these sympathetic nerves leads to burst release of the neurotransmitter noradrenaline (also known as norepinephrine). This causes quiescent melanocyte stem cells to proliferate rapidly, and is followed by their differentiation, migration and permanent depletion from the niche. Transient suppression of the proliferation of melanocyte stem cells prevents stress-induced hair greying. Our study demonstrates that neuronal activity that is induced by acute stress can drive a rapid and permanent loss of somatic stem cells, and illustrates an example in which the maintenance of somatic stem cells is directly influenced by the overall physiological state of the organism.


Assuntos
Vias Autônomas/fisiopatologia , Cor de Cabelo/fisiologia , Melanócitos/patologia , Nicho de Células-Tronco/fisiologia , Células-Tronco/patologia , Estresse Psicológico/fisiopatologia , Sistema Nervoso Simpático/fisiopatologia , Glândulas Suprarrenais/metabolismo , Adrenalectomia , Animais , Vias Autônomas/patologia , Proliferação de Células , Células Cultivadas , Denervação , Feminino , Humanos , Masculino , Melanócitos/citologia , Melanócitos/metabolismo , Camundongos , Norepinefrina/metabolismo , Trauma Psicológico/patologia , Trauma Psicológico/fisiopatologia , Receptores Adrenérgicos beta 2/deficiência , Receptores Adrenérgicos beta 2/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Estresse Psicológico/patologia , Sistema Nervoso Simpático/patologia
3.
Exp Dermatol ; 30(4): 448-456, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33739490

RESUMO

Hair follicle stem cells (HFSCs) are known to be responsible for the initiation of a new hair cycle, but typically remain quiescent for very long periods. In alopecia, or hair loss disorders, follicles can be refractory to activation for years or even permanently. Alopecia can be triggered by autoimmunity, age, chemotherapeutic treatment, stress, disrupted circadian rhythm or other environmental insults. We previously showed that hair follicle stem cells and the hair cycle can be manipulated by regulation of pyruvate entry into mitochondria for subsequent oxidation to fuel the TCA cycle in normal adult mice with typical hair cycling. Here, we present new data from our efforts to develop murine models of alopecia based on environmental triggers that have been shown to do the same in human skin. We found that inhibition of pyruvate transport into mitochondria can accelerate the hair cycle even during refractory hair cycling due to age, repeated chemotherapeutic treatment and stress. Hair cycle acceleration in these alopecia models led to the formation of histologically normal hair follicles within 30-40 days of treatment without any overt signs of toxicity or deleterious effects. Therefore, we propose inhibition of pyruvate entry into mitochondria as a versatile treatment strategy for alopecia in humans.


Assuntos
Alopecia/terapia , Folículo Piloso/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Piruvatos/antagonistas & inibidores , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco/efeitos dos fármacos
4.
J Neurosci ; 38(14): 3571-3583, 2018 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-29530986

RESUMO

Mutations in the cereblon (CRBN) gene cause human intellectual disability, one of the most common cognitive disorders. However, the molecular mechanisms of CRBN-related intellectual disability remain poorly understood. We investigated the role of CRBN in synaptic function and animal behavior using male mouse and Drosophila models. Crbn knock-out (KO) mice showed normal brain and spine morphology as well as intact synaptic plasticity; however, they also exhibited decreases in synaptic transmission and presynaptic release probability exclusively in excitatory synapses. Presynaptic function was impaired not only by loss of CRBN expression, but also by expression of pathogenic CRBN mutants (human R419X mutant and Drosophila G552X mutant). We found that the BK channel blockers paxilline and iberiotoxin reversed this decrease in presynaptic release probability in Crbn KO mice. In addition, paxilline treatment also restored normal cognitive behavior in Crbn KO mice. These results strongly suggest that increased BK channel activity is the pathological mechanism of intellectual disability in CRBN mutations.SIGNIFICANCE STATEMENTCereblon (CRBN), a well known target of the immunomodulatory drug thalidomide, was originally identified as a gene that causes human intellectual disability when mutated. However, the molecular mechanisms of CRBN-related intellectual disability remain poorly understood. Based on the idea that synaptic abnormalities are the most common factor in cognitive dysfunction, we monitored the synaptic structure and function of Crbn knock-out (KO) animals to identify the molecular mechanisms of intellectual disability. Here, we found that Crbn KO animals showed cognitive deficits caused by enhanced BK channel activity and reduced presynaptic glutamate release. Our findings suggest a physiological pathomechanism of the intellectual disability-related gene CRBN and will contribute to the development of therapeutic strategies for CRBN-related intellectual disability.


Assuntos
Cognição , Deficiência Intelectual/genética , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Transmissão Sináptica , Proteínas Adaptadoras de Transdução de Sinal , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Células Cultivadas , Drosophila , Ácido Glutâmico/metabolismo , Indóis/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/antagonistas & inibidores , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/genética , Peptídeos/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Sinapses/fisiologia
5.
J Biol Chem ; 292(35): 14473-14485, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28726639

RESUMO

Mitochondrial calcium plays critical roles in diverse cellular processes ranging from energy metabolism to cell death. Previous studies have demonstrated that mitochondrial calcium uptake is mainly mediated by the mitochondrial calcium uniporter (MCU) complex. However, the roles of the MCU complex in calcium transport, signaling, and dysregulation by oxidative stress still remain unclear. Here, we confirmed that Drosophila MCU contains evolutionarily conserved structures and requires essential MCU regulator (EMRE) for its calcium channel activities. We generated Drosophila MCU loss-of-function mutants, which lacked mitochondrial calcium uptake in response to caffeine stimulation. Basal metabolic activities were not significantly affected in these MCU mutants, as observed in examinations of body weight, food intake, body sugar level, and starvation-induced autophagy. However, oxidative stress-induced increases in mitochondrial calcium, mitochondrial membrane potential depolarization, and cell death were prevented in these mutants. We also found that inositol 1,4,5-trisphosphate receptor genetically interacts with Drosophila MCU and effectively modulates mitochondrial calcium uptake upon oxidative stress. Taken together, these results support the idea that Drosophila MCU is responsible for endoplasmic reticulum-to-mitochondrial calcium transfer and for cell death due to mitochondrial dysfunction under oxidative stress.


Assuntos
Apoptose , Canais de Cálcio/metabolismo , Sinalização do Cálcio , Proteínas de Transporte de Cátions/metabolismo , Proteínas de Drosophila/metabolismo , Retículo Endoplasmático/metabolismo , Mitocôndrias Musculares/metabolismo , Estresse Oxidativo , Animais , Animais Geneticamente Modificados , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Cafeína/farmacologia , Canais de Cálcio/química , Canais de Cálcio/genética , Sinalização do Cálcio/efeitos dos fármacos , Proteínas de Transporte de Cátions/química , Proteínas de Transporte de Cátions/genética , Linhagem Celular , Estimulantes do Sistema Nervoso Central/farmacologia , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster/efeitos dos fármacos , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Retículo Endoplasmático/efeitos dos fármacos , Inativação Gênica , Receptores de Inositol 1,4,5-Trifosfato/antagonistas & inibidores , Receptores de Inositol 1,4,5-Trifosfato/química , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Larva/efeitos dos fármacos , Larva/genética , Larva/crescimento & desenvolvimento , Larva/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias Musculares/efeitos dos fármacos , Mutação , Estresse Oxidativo/efeitos dos fármacos , Sinais Direcionadores de Proteínas/efeitos dos fármacos , Interferência de RNA , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
6.
PLoS Genet ; 11(5): e1005263, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25996931

RESUMO

LKB1 plays important roles in governing energy homeostasis by regulating AMP-activated protein kinase (AMPK) and other AMPK-related kinases, including the salt-inducible kinases (SIKs). However, the roles and regulation of LKB1 in lipid metabolism are poorly understood. Here we show that Drosophila LKB1 mutants display decreased lipid storage and increased gene expression of brummer, the Drosophila homolog of adipose triglyceride lipase (ATGL). These phenotypes are consistent with those of SIK3 mutants and are rescued by expression of constitutively active SIK3 in the fat body, suggesting that SIK3 is a key downstream kinase of LKB1. Using genetic and biochemical analyses, we identify HDAC4, a class IIa histone deacetylase, as a lipolytic target of the LKB1-SIK3 pathway. Interestingly, we found that the LKB1-SIK3-HDAC4 signaling axis is modulated by dietary conditions. In short-term fasting, the adipokinetic hormone (AKH) pathway, related to the mammalian glucagon pathway, inhibits the kinase activity of LKB1 as shown by decreased SIK3 Thr196 phosphorylation, and consequently induces HDAC4 nuclear localization and brummer gene expression. However, under prolonged fasting conditions, AKH-independent signaling decreases the activity of the LKB1-SIK3 pathway to induce lipolytic responses. We also identify that the Drosophila insulin-like peptides (DILPs) pathway, related to mammalian insulin pathway, regulates SIK3 activity in feeding conditions independently of increasing LKB1 kinase activity. Overall, these data suggest that fasting stimuli specifically control the kinase activity of LKB1 and establish the LKB1-SIK3 pathway as a converging point between feeding and fasting signals to control lipid homeostasis in Drosophila.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Ingestão de Alimentos , Jejum , Lipólise , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Drosophila/genética , Proteínas de Drosophila/genética , Regulação da Expressão Gênica , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Homeostase , Lipase/genética , Lipase/metabolismo , Fosforilação , Proteínas Quinases/genética , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais
7.
J Biol Chem ; 291(4): 1803-1816, 2016 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-26631732

RESUMO

Parkin is an E3 ligase that contains a ubiquitin-like (UBL) domain in the N terminus and an R1-in-between-ring-RING2 motif in the C terminus. We showed that the UBL domain specifically interacts with the R1 domain and negatively regulates Parkin E3 ligase activity, Parkin-dependent mitophagy, and Parkin translocation to the mitochondria. The binding between the UBL domain and the R1 domain was suppressed by carbonyl cyanide m-chlorophenyl hydrazone treatment or by expression of PTEN-induced putative kinase 1 (PINK1), an upstream kinase that phosphorylates Parkin at the Ser-65 residue of the UBL domain. Moreover, we demonstrated that phosphorylation of the UBL domain at Ser-65 prevents its binding to the R1 domain and promotes Parkin activities. We further showed that mitochondrial translocation of Parkin, which depends on phosphorylation at Ser-65, and interaction between the R1 domain and a mitochondrial outer membrane protein, VDAC1, are suppressed by binding of the UBL domain to the R1 domain. Interestingly, Parkin with missense mutations associated with Parkinson disease (PD) in the UBL domain, such as K27N, R33Q, and A46P, did not translocate to the mitochondria and induce E3 ligase activity by m-chlorophenyl hydrazone treatment, which correlated with the interaction between the R1 domain and the UBL domain with those PD mutations. These findings provide a molecular mechanism of how Parkin recruitment to the mitochondria and Parkin activation as an E3 ubiquitin ligase are regulated by PINK1 and explain the previously unknown mechanism of how Parkin mutations in the UBL domain cause PD pathogenesis.


Assuntos
Doença de Parkinson/enzimologia , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/metabolismo , Motivos de Aminoácidos , Humanos , Mitocôndrias/enzimologia , Doença de Parkinson/genética , Fosforilação , Complexo Repressor Polycomb 1/genética , Complexo Repressor Polycomb 1/metabolismo , Ligação Proteica , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Estrutura Terciária de Proteína , Transporte Proteico , Ubiquitina-Proteína Ligases/genética , Ubiquitinação
8.
J Biol Chem ; 291(4): 1841-1853, 2016 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-26631731

RESUMO

TRAP1 (tumor necrosis factor receptor-associated protein 1), a mitochondrial Hsp90 family chaperone, has been identified as a critical regulator of cell survival and bioenergetics in tumor cells. To discover novel signaling networks regulated by TRAP1, we generated Drosophila TRAP1 mutants. The mutants successfully developed into adults and produced fertile progeny, showing that TRAP1 is dispensable in development and reproduction. Surprisingly, mutation or knockdown of TRAP1 markedly enhanced Drosophila survival under oxidative stress. Moreover, TRAP1 mutation ameliorated mitochondrial dysfunction and dopaminergic (DA) neuron loss induced by deletion of a familial Parkinson disease gene PINK1 (Pten-induced kinase 1) in Drosophila. Gamitrinib-triphenylphosphonium, a mitochondria-targeted Hsp90 inhibitor that increases cell death in HeLa and MCF7 cells, consistently inhibited cell death induced by oxidative stress and mitochondrial dysfunction induced by PINK1 mutation in mouse embryonic fibroblast cells and DA cell models such as SH-SY5Y and SN4741 cells. Additionally, gamitrinib-triphenylphosphonium also suppressed the defective locomotive activity and DA neuron loss in Drosophila PINK1 null mutants. In further genetic analyses, we showed enhanced expression of Thor, a downstream target gene of transcription factor FOXO, in TRAP1 mutants. Furthermore, deletion of FOXO almost nullified the protective roles of TRAP1 mutation against oxidative stress and PINK1 mutation. These results strongly suggest that inhibition of the mitochondrial chaperone TRAP1 generates a retrograde cell protective signal from mitochondria to the nucleus in a FOXO-dependent manner.


Assuntos
Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Guanidinas/farmacologia , Proteínas de Choque Térmico HSP90/genética , Lactamas Macrocíclicas/farmacologia , Mitocôndrias/metabolismo , Compostos Organosselênicos/farmacologia , Doença de Parkinson/metabolismo , Animais , Sobrevivência Celular , Modelos Animais de Doenças , Neurônios Dopaminérgicos/citologia , Neurônios Dopaminérgicos/metabolismo , Drosophila/efeitos dos fármacos , Drosophila/genética , Drosophila/crescimento & desenvolvimento , Proteínas de Drosophila/antagonistas & inibidores , Feminino , Fatores de Transcrição Forkhead/genética , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/genética , Mutação , Estresse Oxidativo , Doença de Parkinson/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo
9.
J Biol Chem ; 288(8): 5660-72, 2013 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-23303188

RESUMO

Mutations in PINK1 (PTEN-induced putative kinase 1) are tightly linked to autosomal recessive Parkinson disease (PD). Although more than 50 mutations in PINK1 have been discovered, the role of these mutations in PD pathogenesis remains poorly understood. Here, we characterized 17 representative PINK1 pathogenic mutations in both mammalian cells and Drosophila. These mutations did not affect the typical cleavage patterns and subcellular localization of PINK1 under both normal and damaged mitochondria conditions in mammalian cells. However, PINK1 mutations in the kinase domain failed to translocate Parkin to mitochondria and to induce mitochondrial aggregation. Consistent with the mammalian data, Drosophila PINK1 mutants with mutations in the kinase domain (G426D and L464P) did not genetically interact with Parkin. Furthermore, PINK1-null flies expressing the transgenic G426D mutant displayed defective phenotypes with increasing age, whereas L464P mutant-expressing flies exhibited the phenotypes at an earlier age. Collectively, these results strongly support the hypothesis that the kinase activity of PINK1 is essential for its function and for regulating downstream Parkin functions in mitochondria. We believe that this study provides the basis for understanding the molecular and physiological functions of various PINK1 mutations and provides insights into the pathogenic mechanisms of PINK1-linked PD.


Assuntos
Mutação , Doença de Parkinson/metabolismo , Proteínas Quinases/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Carbonil Cianeto m-Clorofenil Hidrazona/farmacologia , Drosophila melanogaster , Fibroblastos/citologia , Células HEK293 , Células HeLa , Humanos , Imuno-Histoquímica/métodos , Masculino , Potencial da Membrana Mitocondrial , Camundongos , Mitocôndrias/metabolismo , Neurônios/metabolismo , Fenótipo , Proteínas Quinases/metabolismo , Transgenes , Ubiquitina-Proteína Ligases/metabolismo
10.
Exp Mol Med ; 56(1): 110-117, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38182654

RESUMO

Hair follicles, which are connected to sebaceous glands in the skin, undergo cyclic periods of regeneration, degeneration, and rest throughout adult life in mammals. The crucial function of hair follicle stem cells is to maintain these hair growth cycles. Another vital aspect is the activity of melanocyte stem cells, which differentiate into melanin-producing melanocytes, contributing to skin and hair pigmentation. Sebaceous gland stem cells also have a pivotal role in maintaining the skin barrier by regenerating mature sebocytes. These stem cells are maintained in a specialized microenvironment or niche and are regulated by internal and external signals, determining their dynamic behaviors in homeostasis and hair follicle regeneration. The activity of these stem cells is tightly controlled by various factors secreted by the niche components around the hair follicles, as well as immune-mediated damage signals, aging, metabolic status, and stress. In this study, we review these diverse stem cell regulatory and related molecular mechanisms of hair regeneration and disease conditions. Molecular insights would provide new perspectives on the disease mechanisms as well as hair and skin disorder treatment.


Assuntos
Folículo Piloso , Cabelo , Animais , Folículo Piloso/fisiologia , Pele , Melanócitos/metabolismo , Células-Tronco/metabolismo , Mamíferos
11.
Nat Commun ; 15(1): 468, 2024 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-38212312

RESUMO

Diabetic sensory neuropathy (DSN) is one of the most common complications of type 2 diabetes (T2D), however the molecular mechanistic association between T2D and DSN remains elusive. Here we identify ubiquitin C-terminal hydrolase L1 (UCHL1), a deubiquitinase highly expressed in neurons, as a key molecule underlying T2D and DSN. Genetic ablation of UCHL1 leads to neuronal insulin resistance and T2D-related symptoms in Drosophila. Furthermore, loss of UCHL1 induces DSN-like phenotypes, including numbness to external noxious stimuli and axonal degeneration of sensory neurons in flies' legs. Conversely, UCHL1 overexpression improves DSN-like defects of T2D model flies. UCHL1 governs insulin signaling by deubiquitinating insulin receptor substrate 1 (IRS1) and antagonizes an E3 ligase of IRS1, Cullin 1 (CUL1). Consistent with these results, genetic and pharmacological suppression of CUL1 activity rescues T2D- and DSN-associated phenotypes. Therefore, our findings suggest a complete set of genetic factors explaining T2D and DSN, together with potential remedies for the diseases.


Assuntos
Diabetes Mellitus Tipo 2 , Resistência à Insulina , Animais , Resistência à Insulina/genética , Ubiquitina Tiolesterase/genética , Diabetes Mellitus Tipo 2/genética , Drosophila , Neurônios
12.
PLoS One ; 19(5): e0303433, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38743676

RESUMO

Triple-negative breast cancer (TNBC) demands urgent attention for the development of effective treatment strategies due to its aggressiveness and limited therapeutic options [1]. This research is primarily focused on identifying new biomarkers vital for immunotherapy, with the aim of developing tailored treatments specifically for TNBC, such as those targeting the PD-1/PD-L1 pathway. To achieve this, the study places a strong emphasis on investigating Ig genes, a characteristic of immune checkpoint inhibitors, particularly genes expressing Ig-like domains with altered expression levels induced by "cancer deformation," a condition associated with cancer malignancy. Human cells can express approximately 800 Ig family genes, yet only a few Ig genes, including PD-1 and PD-L1, have been developed into immunotherapy drugs thus far. Therefore, we investigated the Ig genes that were either upregulated or downregulated by the artificial metastatic environment in TNBC cell line. As a result, we confirmed the upregulation of approximately 13 Ig genes and validated them using qPCR. In summary, our study proposes an approach for identifying new biomarkers applicable to future immunotherapies aimed at addressing challenging cases of TNBC where conventional treatments fall short.


Assuntos
Biomarcadores Tumorais , Imunoterapia , Neoplasias de Mama Triplo Negativas , Humanos , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/imunologia , Neoplasias de Mama Triplo Negativas/terapia , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Imunoterapia/métodos , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica , Antígeno B7-H1/metabolismo , Antígeno B7-H1/genética , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/metabolismo
13.
Mol Cells ; 46(10): 573-578, 2023 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-37650216

RESUMO

The mammalian skin contains hair follicles, which are epidermal appendages that undergo periodic cycles and exhibit mini-organ features, such as discrete stem cell compartments and different cellular components. Wound-induced hair follicle neogenesis (WIHN) is the remarkable ability to regenerate hair follicles after large-scale wounding and occurs in several adult mammals. WIHN is comparable to embryonic hair follicle development in its processes. Researchers are beginning to identify the stem cells that, in response to wounding, develop into neogenic hair follicles, as well as to understand the functions of immune cells, mesenchymal cells, and several signaling pathways that are essential for this process. WIHN represents a promising therapeutic approach to the reprogramming of cellular states for promoting hair follicle regeneration and preventing scar formation. In the scope of this review, we investigate the contribution of several cell types and molecular mechanisms to WIHN.


Assuntos
Folículo Piloso , Cicatrização , Camundongos , Animais , Folículo Piloso/metabolismo , Cicatrização/fisiologia , Camundongos Endogâmicos C57BL , Cabelo , Pele/metabolismo , Mamíferos
14.
BMB Rep ; 56(1): 2-9, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36379515

RESUMO

Hair follicles in the skin undergo cyclic rounds of regeneration, degeneration, and rest throughout life. Stem cells residing in hair follicles play a pivotal role in maintaining tissue homeostasis and hair growth cycles. Research on hair follicle aging and age-related hair loss has demonstrated that a decline in hair follicle stem cell (HFSC) activity with aging can decrease the regeneration capacity of hair follicles. This review summarizes our understanding of how age-associated HFSC intrinsic and extrinsic mechanisms can induce HFSC aging and hair loss. In addition, we discuss approaches developed to attenuate ageassociated changes in HFSCs and their niches, thereby promoting hair regrowth. [BMB Reports 2023; 56(1): 2-9].


Assuntos
Folículo Piloso , Cabelo , Humanos , Envelhecimento , Alopecia , Células-Tronco
15.
J Biol Chem ; 286(4): 2658-64, 2011 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-21127058

RESUMO

Salt-inducible kinase (SIK), one of the AMP-activated kinase (AMPK)-related kinases, has been suggested to play important functions in glucose homeostasis by inhibiting the cAMP-response element-binding protein (CREB)-regulated transcription coactivator (CRTC). To examine the role of SIK in vivo, we generated Drosophila SIK mutant and found that the mutant flies have higher amounts of lipid and glycogen stores and are resistant to starvation. Interestingly, SIK transcripts are highly enriched in the brain, and we found that neuron-specific expression of exogenous SIK fully rescued lipid and glycogen storage phenotypes as well as starvation resistance of the mutant. Using genetic and biochemical analyses, we demonstrated that CRTC Ser-157 phosphorylation by SIK is critical for inhibiting CRTC activity in vivo. Furthermore, double mutants of SIK and CRTC became sensitive to starvation, and the Ser-157 phosphomimetic mutation of CRTC reduced lipid and glycogen levels in the SIK mutant, suggesting that CRTC mediates the effects of SIK signaling. Collectively, our results strongly support the importance of the SIK-CRTC signaling axis that functions in the brain to maintain energy homeostasis in Drosophila.


Assuntos
AMP Cíclico , Proteínas de Drosophila/metabolismo , Proteínas Serina-Treonina Quinases/biossíntese , Elementos de Resposta/fisiologia , Inanição/metabolismo , Fatores de Transcrição/metabolismo , Animais , Proteínas de Drosophila/genética , Drosophila melanogaster , Regulação Enzimológica da Expressão Gênica , Mutação , Especificidade de Órgãos/fisiologia , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais , Inanição/genética , Fatores de Transcrição/genética
16.
Sci Adv ; 7(28)2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34244144

RESUMO

The role of ubiquitin carboxyl-terminal hydrolase L1 (UCHL1; also called PARK5) in the pathogenesis of Parkinson's disease (PD) has been controversial. Here, we find that the loss of UCHL1 destabilizes pyruvate kinase (PKM) and mitigates the PD-related phenotypes induced by PTEN-induced kinase 1 (PINK1) or Parkin loss-of-function mutations in Drosophila and mammalian cells. In UCHL1 knockout cells, cellular pyruvate production and ATP levels are diminished, and the activity of AMP-activated protein kinase (AMPK) is highly induced. Consequently, the activated AMPK promotes the mitophagy mediated by Unc-51-like kinase 1 (ULK1) and FUN14 domain-containing 1 (FUNDC1), which underlies the effects of UCHL1 deficiency in rescuing PD-related defects. Furthermore, we identify tripartite motif-containing 63 (TRIM63) as a previously unknown E3 ligase of PKM and demonstrate its antagonistic interaction with UCHL1 to regulate PD-related pathologies. These results suggest that UCHL1 is an integrative factor for connecting glycolysis and PD pathology.

17.
JCI Insight ; 4(13)2019 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-31292296

RESUMO

Ubiquitin-conjugating enzyme E2O (UBE2O) is expressed preferentially in metabolic tissues, but its role in regulating energy homeostasis has yet to be defined. Here we find that UBE2O is markedly upregulated in obese subjects with type 2 diabetes and show that whole-body disruption of Ube2o in mouse models in vivo results in improved metabolic profiles and resistance to high-fat diet-induced (HFD-induced) obesity and metabolic syndrome. With no difference in nutrient intake, Ube2o-/- mice were leaner and expended more energy than WT mice. In addition, hyperinsulinemic-euglycemic clamp studies revealed that Ube2o-/- mice were profoundly insulin sensitive. Through phenotype analysis of HFD mice with muscle-, fat-, or liver-specific knockout of Ube2o, we further identified UBE2O as an essential regulator of glucose and lipid metabolism programs in skeletal muscle, but not in adipose or liver tissue. Mechanistically, UBE2O acted as a ubiquitin ligase and targeted AMPKα2 for ubiquitin-dependent degradation in skeletal muscle; further, muscle-specific heterozygous knockout of Prkaa2 ablated UBE2O-controlled metabolic processes. These results identify the UBE2O/AMPKα2 axis as both a potent regulator of metabolic homeostasis in skeletal muscle and a therapeutic target in the treatment of diabetes and metabolic disorders.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Resistência à Insulina , Síndrome Metabólica/metabolismo , Obesidade/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo , Animais , Linhagem Celular , Diabetes Mellitus Tipo 2/complicações , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Feminino , Glucose/metabolismo , Humanos , Insulina/metabolismo , Metabolismo dos Lipídeos , Masculino , Síndrome Metabólica/etiologia , Camundongos , Camundongos Knockout , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Mioblastos Esqueléticos , Obesidade/etiologia , Cultura Primária de Células , Proteólise , Enzimas de Conjugação de Ubiquitina/análise , Enzimas de Conjugação de Ubiquitina/genética , Ubiquitinação , Regulação para Cima
18.
Curr Biol ; 26(6): 814-20, 2016 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-26948873

RESUMO

Although several neural pathways have been implicated in feeding behaviors in mammals [1-7], it remains unclear how the brain coordinates feeding motivations to maintain a constant body weight (BW). Here, we identified a neuropeptide pathway important for the satiety and BW control in Drosophila. Silencing of myoinhibitory peptide (MIP) neurons significantly increased BW through augmented food intake and fat storage. Likewise, the loss-of-function mutation of mip also increased feeding and BW. Suppressing the MIP pathway induced satiated flies to behave like starved ones, with elevated sensitivity toward food. Conversely, activating MIP neurons greatly decreased food intake and BW and markedly blunted the sensitivity of starved flies toward food. Upon terminating the activation protocol of MIP neurons, the decreased BW reverts rapidly to the normal level through a strong feeding rebound, indicating the switch-like role of MIP pathway in feeding. Surprisingly, the MIP-mediated BW decrease occurred independently of sex peptide receptor (SPR), the only known receptor for MIP, suggesting the presence of a yet-unknown MIP receptor. Together, our results reveal a novel anorexigenic pathway that controls satiety in Drosophila and provide a new avenue to study how the brain actively maintains a constant BW.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/fisiologia , Resposta de Saciedade/fisiologia , Animais , Animais Geneticamente Modificados , Peso Corporal , Encéfalo/fisiologia , Proteínas de Drosophila/genética , Ingestão de Alimentos , Comportamento Alimentar , Feminino , Regulação da Expressão Gênica , Canais Iônicos , Masculino , Neurônios/metabolismo , Peptídeos/metabolismo , Receptores de Peptídeos , Canal de Cátion TRPA1 , Canais de Cátion TRPC/metabolismo
19.
Curr Biol ; 25(6): 790-797, 2015 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-25702579

RESUMO

In polyandrous females, sperm storage permits competition between sperm of different mates, and in some species females influence the relative fertilization success of competing sperm in favor of a preferred mate [1, 2]. In female Drosophila melanogaster, sperm competition is strongly influenced by the timing of sperm ejection from the uterus [3, 4]. Understanding how female behavior influences sperm competition requires knowledge of the neuronal mechanisms controlling sperm retention and storage, which is currently lacking. Here, we show that D. melanogaster females eject male ejaculates from the uterus 1-6 hr after mating with a stereotypic behavior regulated by a brain signaling pathway composed of diuretic hormone 44 (Dh44), a neuropeptide related to vertebrate corticotropin-releasing factor (CRF), and its receptor, Dh44R1. Suppression of Dh44 signals in the brain expedites sperm ejection from the uterus, resulting in marked reduction of sperm in the storage organs and decreased fecundity, whereas enhancement of Dh44 signals delays sperm expulsion. The Dh44 function was mapped to six neurons located in the pars intercerebralis of the brain together with a small subset of Dh44R1 neurons that express the sex-specific transcription factor doublesex. This study identifies a neuronal pathway by which females can control sperm retention and storage and provides new insight into how the female might exercise post-copulatory sexual selection.


Assuntos
Drosophila melanogaster/fisiologia , Comportamento Sexual Animal/fisiologia , Espermatozoides/fisiologia , Animais , Animais Geneticamente Modificados , Encéfalo/fisiologia , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/genética , Feminino , Fertilização/genética , Fertilização/fisiologia , Hormônios de Inseto/antagonistas & inibidores , Hormônios de Inseto/genética , Hormônios de Inseto/fisiologia , Masculino , Vias Neurais/fisiologia , Neuropeptídeos/antagonistas & inibidores , Neuropeptídeos/genética , Neuropeptídeos/fisiologia , Interferência de RNA , Receptores de Superfície Celular/antagonistas & inibidores , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/fisiologia , Reprodução/genética , Reprodução/fisiologia
20.
Mol Cell Biol ; 34(6): 926-38, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24379443

RESUMO

SREBP1c is a key transcription factor that regulates de novo lipogenesis during anabolic periods. However, the molecular mechanisms involved in the suppression of SREBP1c under nutritional deprivation are largely unknown. In this study, we demonstrate that the small ubiquitin-related modifier (SUMO) E3 ligase, a protein inhibitor of activated STAT Y (PIASy), sumoylates SREBP1c at Lys98, leading to suppression of the hepatic lipogenic program upon fasting-induced signals. In primary hepatocytes, ablation of PIASy stimulated intracellular lipid accumulation through the induction of SREBP1c and its target genes. Given that protein kinase A (PKA) plays important roles in catabolic responses, activated PKA enhances the sumoylation of SREBP1c and potentiates the interaction between SREBP1c and PIASy. Notably, overexpression of PIASy in obese db/db mice ameliorated hepatic steatosis, while suppression of PIASy in lean (wild-type) mice stimulated hepatic lipogenesis with increased expression of SREBP1c target genes. Furthermore, PKA-mediated SREBP1c phosphorylation augmented SREBP1c sumoylation, subsequently leading to degradation of SREBP1c via ubiquitination. Together, these data suggest that PKA-induced SREBP1c sumoylation by PIASy is a key regulatory mechanism to turn off hepatic lipogenesis during nutritional deprivation.


Assuntos
Jejum/metabolismo , Metabolismo dos Lipídeos/fisiologia , Proteínas Inibidoras de STAT Ativados/metabolismo , Transdução de Sinais/fisiologia , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Sumoilação/fisiologia , Animais , Células COS , Células Cultivadas , Chlorocebus aethiops , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Jejum/fisiologia , Hepatócitos/metabolismo , Hepatócitos/fisiologia , Metabolismo dos Lipídeos/genética , Lipogênese/genética , Lipogênese/fisiologia , Fígado/metabolismo , Fígado/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Fosforilação/genética , Fosforilação/fisiologia , Proteínas Inibidoras de STAT Ativados/genética , Proteína SUMO-1/genética , Proteína SUMO-1/metabolismo , Transdução de Sinais/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Sumoilação/genética , Transcrição Gênica/genética , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação/genética , Ubiquitinação/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA