Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.000
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
J Mol Model ; 30(6): 170, 2024 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-38753123

RESUMO

CONTEXT: In the pursuit of novel therapeutic possibilities, repurposing existing drugs has gained prominence as an efficient strategy. The findings from our study highlight the potential of repurposed drugs as promising candidates against receptor for advanced glycation endproducts (RAGE) that offer therapeutic implications in cancer, neurodegenerative conditions and metabolic syndromes. Through careful analyses of binding affinities and interaction patterns, we identified a few promising candidates, ultimately focusing on sertindole and temoporfin. These candidates exhibited exceptional binding affinities, efficacy, and specificity within the RAGE binding pocket. Notably, they displayed a pronounced propensity to interact with the active site of RAGE. Our investigation further revealed that sertindole and temoporfin possess desirable pharmacological properties that highlighted them as attractive candidates for targeted drug development. Overall, our integrated computational approach provides a comprehensive understanding of the interactions between repurposed drugs, sertindole and temoporfin and RAGE that pave the way for future experimental validation and drug development endeavors. METHODS: We present an integrated approach utilizing molecular docking and extensive molecular dynamics (MD) simulations to evaluate the potential of FDA-approved drugs, sourced from DrugBank, against RAGE. To gain deeper insights into the binding mechanisms of the elucidated candidate repurposed drugs, sertindole and temoporfin with RAGE, we conducted extensive all-atom MD simulations, spanning 500 nanoseconds (ns). These simulations elucidated the conformational dynamics and stability of the RAGE-sertindole and RAGE-temoporfin complexes.


Assuntos
Reposicionamento de Medicamentos , Imidazóis , Indóis , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Receptor para Produtos Finais de Glicação Avançada , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Receptor para Produtos Finais de Glicação Avançada/química , Humanos , Indóis/química , Indóis/farmacologia , Imidazóis/química , Imidazóis/farmacologia , Ligação Proteica , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Doenças Metabólicas/tratamento farmacológico , Doenças Metabólicas/metabolismo , Sítios de Ligação
2.
Metabolism ; 155: 155911, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38609037

RESUMO

BACKGROUND: The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) is increasing year by year and has become one of the leading causes of end-stage liver disease worldwide. Triggering Receptor Expressed on Myeloid Cells 2 (Trem2) has been confirmed to play an essential role in the progression of MASLD, but its specific mechanism still needs to be clarified. This study aims to explore the role and mechanism of Trem2 in MASLD. METHODS: Human liver tissues were obtained from patients with MASLD and controls. Myeloid-specific knockout mice (Trem2mKO) and myeloid-specific overexpression mice (Trem2TdT) were fed a high-fat diet, either AMLN or CDAHFD, to establish the MASLD model. Relevant signaling molecules were assessed through lipidomics and RNA-seq analyses after that. RESULTS: Trem2 is upregulated in human MASLD/MASH-associated macrophages and is associated with hepatic steatosis and inflammation progression. Hepatic steatosis and inflammatory responses are exacerbated with the knockout of myeloid Trem2 in MASLD mice, while mice overexpressing Trem2 exhibit the opposite phenomenon. Mechanistically, Trem2mKO can aggravate macrophage pyroptosis through the PI3K/AKT signaling pathway and amplify the resulting inflammatory response. At the same time, Trem2 promotes the inflammation resolution phenotype transformation of macrophages through TGFß1, thereby promoting tissue repair. CONCLUSIONS: Myeloid Trem2 ameliorates the progression of Metabolic dysfunction-associated steatotic liver disease by regulating macrophage pyroptosis and inflammation resolution. We believe targeting myeloid Trem2 could represent a potential avenue for treating MASLD.


Assuntos
Progressão da Doença , Fígado Gorduroso , Inflamação , Macrófagos , Glicoproteínas de Membrana , Camundongos Knockout , Piroptose , Receptores Imunológicos , Animais , Receptores Imunológicos/metabolismo , Receptores Imunológicos/genética , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Camundongos , Humanos , Macrófagos/metabolismo , Inflamação/metabolismo , Inflamação/patologia , Piroptose/fisiologia , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Fígado Gorduroso/genética , Masculino , Camundongos Endogâmicos C57BL , Doenças Metabólicas/metabolismo , Doenças Metabólicas/patologia , Doenças Metabólicas/genética , Fígado/metabolismo , Fígado/patologia
3.
Biomolecules ; 14(4)2024 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-38672429

RESUMO

In 1992, a transcendental report suggested that the receptor of advanced glycation end-products (RAGE) functions as a cell surface receptor for a wide and diverse group of compounds, commonly referred to as advanced glycation end-products (AGEs), resulting from the non-enzymatic glycation of lipids and proteins in response to hyperglycemia. The interaction of these compounds with RAGE represents an essential element in triggering the cellular response to proteins or lipids that become glycated. Although initially demonstrated for diabetes complications, a growing body of evidence clearly supports RAGE's role in human diseases. Moreover, the recognizing capacities of this receptor have been extended to a plethora of structurally diverse ligands. As a result, it has been acknowledged as a pattern recognition receptor (PRR) and functionally categorized as the RAGE axis. The ligation to RAGE leads the initiation of a complex signaling cascade and thus triggering crucial cellular events in the pathophysiology of many human diseases. In the present review, we intend to summarize basic features of the RAGE axis biology as well as its contribution to some relevant human diseases such as metabolic diseases, neurodegenerative, cardiovascular, autoimmune, and chronic airways diseases, and cancer as a result of exposure to AGEs, as well as many other ligands.


Assuntos
Produtos Finais de Glicação Avançada , Inflamação , Receptor para Produtos Finais de Glicação Avançada , Humanos , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Produtos Finais de Glicação Avançada/metabolismo , Inflamação/metabolismo , Transdução de Sinais , Neoplasias/metabolismo , Animais , Doenças Cardiovasculares/metabolismo , Doenças Neurodegenerativas/metabolismo , Doenças Metabólicas/metabolismo , Doenças Autoimunes/metabolismo
4.
J Agric Food Chem ; 72(15): 8632-8649, 2024 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-38577880

RESUMO

Our previous studies found that Sea Buckthorn polyphenols (SBP) extract inhibits fatty acid synthase (FAS) in vitro. Thus, we continued to explore possible effects and underlying mechanisms of SBP on complicated metabolic disorders in long-term high-fat-diet (HFD)-fed mice. To reveal that, an integrated approach was developed in this study. Targeted quantitative lipidomics with a total of 904 unique lipids mapping contributes to profiling the comprehensive features of disarranged hepatic lipid homeostasis and discovering a set of newfound lipid-based biomarkers to predict the occurrence and indicate the progression of metabolic disorders beyond current indicators. On the other hand, technologies of intermolecular interactions characterization, especially surface plasmon resonance (SPR) assay, contribute to recognizing targeted bioactive constituents present in SBP. Our findings highlight hepatic lipid homeostasis maintenance and constituent-FAS enzyme interactions, to provide new insights that SBP as a functional food alleviates HFD-induced metabolic disorders in mice via reprograming hepatic lipid homeostasis caused by targeting FAS, owing to four polyphenols directly interacting with FAS and cinaroside binding to FAS with good affinity.


Assuntos
Hippophae , Doenças Metabólicas , Camundongos , Animais , Polifenóis/metabolismo , Fígado/metabolismo , Dieta Hiperlipídica/efeitos adversos , Ácido Graxo Sintases/genética , Ácido Graxo Sintases/metabolismo , Lipídeos/farmacologia , Doenças Metabólicas/metabolismo , Homeostase , Camundongos Endogâmicos C57BL , Metabolismo dos Lipídeos
5.
Nat Rev Endocrinol ; 20(6): 366-378, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38519567

RESUMO

Protein tyrosine phosphatase 1B (PTP1B), a non-transmembrane phosphatase, has a major role in a variety of signalling pathways, including direct negative regulation of classic insulin and leptin signalling pathways, and is implicated in the pathogenesis of several cardiometabolic diseases and cancers. As such, PTP1B has been a therapeutic target for over two decades, with PTP1B inhibitors identified either from natural sources or developed throughout the years. Some of these inhibitors have reached phase I and/or II clinical trials in humans for the treatment of type 2 diabetes mellitus, obesity and/or metastatic breast cancer. In this Review, we summarize the cellular processes and regulation of PTP1B, discuss evidence from in vivo preclinical and human studies of the association between PTP1B and different disorders, and discuss outcomes of clinical trials. We outline challenges associated with the targeting of this phosphatase (which was, until the past few years, viewed as difficult to target), the current state of the field of PTP1B inhibitors (and dual phosphatase inhibitors) and future directions for manipulating the activity of this key metabolic enzyme.


Assuntos
Desenvolvimento de Medicamentos , Doenças Metabólicas , Proteína Tirosina Fosfatase não Receptora Tipo 1 , Humanos , Proteína Tirosina Fosfatase não Receptora Tipo 1/antagonistas & inibidores , Proteína Tirosina Fosfatase não Receptora Tipo 1/metabolismo , Doenças Metabólicas/tratamento farmacológico , Doenças Metabólicas/metabolismo , Animais , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Inibidores Enzimáticos/uso terapêutico , Inibidores Enzimáticos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Obesidade/tratamento farmacológico , Obesidade/metabolismo
6.
J Cell Physiol ; 239(5): e31229, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38426269

RESUMO

RNA-binding proteins (RBPs) play a crucial role in the regulation of posttranscriptional RNA networks, which can undergo dysregulation in many pathological conditions. Human antigen R (HuR) is a highly researched RBP that plays a crucial role as a posttranscriptional regulator. HuR plays a crucial role in the amplification of inflammatory signals by stabilizing the messenger RNA of diverse inflammatory mediators and key molecular players. The noteworthy correlations between HuR and its target molecules, coupled with the remarkable impacts reported on the pathogenesis and advancement of multiple diseases, position HuR as a promising candidate for therapeutic intervention in diverse inflammatory conditions. This review article examines the significance of HuR as a member of the RBP family, its regulatory mechanisms, and its implications in the pathophysiology of inflammation and cardiometabolic illnesses. Our objective is to illuminate potential directions for future research and drug development by conducting a comprehensive analysis of the existing body of research on HuR.


Assuntos
Doenças Cardiovasculares , Proteína Semelhante a ELAV 1 , Inflamação , Humanos , Proteína Semelhante a ELAV 1/metabolismo , Proteína Semelhante a ELAV 1/genética , Inflamação/genética , Inflamação/patologia , Doenças Cardiovasculares/genética , Doenças Cardiovasculares/imunologia , Doenças Cardiovasculares/metabolismo , Animais , Regulação da Expressão Gênica , Doenças Metabólicas/genética , Doenças Metabólicas/imunologia , Doenças Metabólicas/metabolismo , Transdução de Sinais , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo
7.
Cell Rep ; 43(3): 113900, 2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38460132

RESUMO

Iron overload is closely associated with metabolic dysfunction. However, the role of iron in the hypothalamus remains unclear. Here, we find that hypothalamic iron levels are increased, particularly in agouti-related peptide (AgRP)-expressing neurons in high-fat-diet-fed mice. Using pharmacological or genetic approaches, we reduce iron overload in AgRP neurons by central deferoxamine administration or transferrin receptor 1 (Tfrc) deletion, ameliorating diet-induced obesity and related metabolic dysfunction. Conversely, Tfrc-mediated iron overload in AgRP neurons leads to overeating and adiposity. Mechanistically, the reduction of iron overload in AgRP neurons inhibits AgRP neuron activity; improves insulin and leptin sensitivity; and inhibits iron-induced oxidative stress, endoplasmic reticulum stress, nuclear factor κB signaling, and suppression of cytokine signaling 3 expression. These results highlight the critical role of hypothalamic iron in obesity development and suggest targets for treating obesity and related metabolic disorders.


Assuntos
Sobrecarga de Ferro , Doenças Metabólicas , Camundongos , Animais , Proteína Relacionada com Agouti/metabolismo , Obesidade/metabolismo , Hipotálamo/metabolismo , Leptina/metabolismo , Neurônios/metabolismo , Dieta Hiperlipídica/efeitos adversos , Doenças Metabólicas/metabolismo , Ferro/metabolismo , Camundongos Endogâmicos C57BL
8.
Anim Sci J ; 95(1): e13935, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38471769

RESUMO

High-yielding dairy cows undergo various physiological stresses during the transitional phase of the calving cycle. In this period, they experience negative energy balance, subjecting the liver to significant metabolic stress from an influx of nonesterified fatty acids. This metabolic stress not only impairs liver function but also diminishes milk production. Early lactation dairy cows may develop endoplasmic reticulum (ER) stress in the liver, potentially leading to liver-related diseases and contributing to ER stress in mammary epithelial cells, resulting in decreased milk production. Natural products that alleviate ER stress have been identified, and if further in vivo studies confirm their efficacy, they have potential as feed additives to prevent disease and reduce milk yield. Conversely, physiological levels of ER stress play a role in mammary gland development and positively influence protein synthesis in milk. Understanding the threshold level of ER stress in mammary tissue and its detailed mechanisms will be crucial in dairy farming.


Assuntos
Doenças dos Bovinos , Hepatopatias , Doenças Metabólicas , Feminino , Bovinos , Animais , Glândulas Mamárias Animais/metabolismo , Leite/metabolismo , Lactação/fisiologia , Estresse do Retículo Endoplasmático , Hepatopatias/veterinária , Células Epiteliais , Doenças Metabólicas/metabolismo , Doenças Metabólicas/veterinária , Doenças dos Bovinos/metabolismo
9.
Biochem Pharmacol ; 222: 116108, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38438053

RESUMO

The primary role of adipose tissue stem cells (ADSCs) is to support the function and homeostasis of adipose tissue in physiological and pathophysiological conditions. However, when ADSCs become dysfunctional in diseases such as obesity and cancer, they become impaired, undergo signalling changes, and their epigenome is altered, which can have a dramatic effect on human health. In more recent years, the therapeutic potential of ADSCs in regenerative medicine, wound healing, and for treating conditions such as cancer and metabolic diseases has been extensively investigated with very promising results. ADSCs have also been used to generate two-dimensional (2D) and three-dimensional (3D) cellular and in vivo models to study adipose tissue biology and function as well as intracellular communication. Characterising the biology and function of ADSCs, how it is altered in health and disease, and its therapeutic potential and uses in cellular models is key for designing intervention strategies for complex metabolic diseases and cancer.


Assuntos
Doenças Metabólicas , Neoplasias , Humanos , Tecido Adiposo/metabolismo , Células-Tronco/metabolismo , Cicatrização/fisiologia , Doenças Metabólicas/terapia , Doenças Metabólicas/metabolismo , Neoplasias/metabolismo
10.
Br J Pharmacol ; 181(12): 1695-1719, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38528718

RESUMO

Cardio-cerebrovascular diseases encompass pathological changes in the heart, brain and vascular system, which pose a great threat to health and well-being worldwide. Moreover, metabolic diseases contribute to and exacerbate the impact of vascular diseases. Inflammation is a complex process that protects against noxious stimuli but is also dysregulated in numerous so-called inflammatory diseases, one of which is atherosclerosis. Inflammation involves multiple organ systems and a complex cascade of molecular and cellular events. Numerous studies have shown that inflammation plays a vital role in cardio-cerebrovascular diseases and metabolic diseases. The absent in melanoma 2 (AIM2) inflammasome detects and is subsequently activated by double-stranded DNA in damaged cells and pathogens. With the assistance of the mature effector molecule caspase-1, the AIM2 inflammasome performs crucial biological functions that underpin its involvement in cardio-cerebrovascular diseases and related metabolic diseases: The production of interleukin-1 beta (IL-1ß), interleukin-18 (IL-18) and N-terminal pore-forming Gasdermin D fragment (GSDMD-N) mediates a series of inflammatory responses and programmed cell death (pyroptosis and PANoptosis). Currently, several agents have been reported to inhibit the activity of the AIM2 inflammasome and have the potential to be evaluated for use in clinical settings. In this review, we systemically elucidate the assembly, biological functions, regulation and mechanisms of the AIM2 inflammasome in cardio-cerebrovascular diseases and related metabolic diseases and outline the inhibitory agents of the AIM2 inflammasome as potential therapeutic drugs.


Assuntos
Proteínas de Ligação a DNA , Inflamassomos , Doenças Metabólicas , Humanos , Inflamassomos/metabolismo , Animais , Proteínas de Ligação a DNA/metabolismo , Doenças Metabólicas/tratamento farmacológico , Doenças Metabólicas/metabolismo , Doenças Vasculares/tratamento farmacológico , Doenças Vasculares/metabolismo , Doenças Vasculares/imunologia , Inflamação/metabolismo , Inflamação/tratamento farmacológico
11.
Biochim Biophys Acta Mol Basis Dis ; 1870(4): 167063, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38360073

RESUMO

CONTEXT: Polycystic ovary syndrome (PCOS) is one of the most common endocrine disorders in women. Retinoid-interferon-induced mortality 19 (GRIM19) is a functional component of mitochondrial complex I that plays a role in cellular energy metabolism. However, the role of GRIM19 in the pathogenesis of PCOS is still unclear. OBJECTIVE: To investigate the role of GRIM19 in the pathogenesis of PCOS. DESIGN: We first measured the expression of GRIM19 in human granulosa cells (hGCs) from patients with and without PCOS (n = 16 per group), and then established a PCOS mouse model with WT and Grim19+/- mice for in vivo experiments. Glucose uptake-related genes RAC1 and GLUT4 and energy metabolism levels in KGN cells were examined in vitro by knocking down GRIM19 in the cell lines. Additionally, ovulation-related genes such as p-ERK1/2, HAS2, and PTX3 were also studied to determine their expression levels. RESULTS: GRIM19 expression was reduced in hGCs of PCOS patients, which was negatively correlated with BMI and serum testosterone level. Grim19+/- mice with PCOS exhibited a markedly anovulatory phenotype and disturbed glycolipid metabolism. In vitro experiments, GRIM19 deficiency inhibited the RAC1/GLUT4 pathway, reducing insulin-stimulated glucose uptake in KGN cells. Moreover, GRIM19 deficiency induced mitochondrial dysfunction, defective glucose metabolism, and apoptosis. In addition, GRIM19 deficiency suppressed the expression of ovulation-related genes in KGN cells, which was regulated by dihydrotestosterone mediated androgen receptor. CONCLUSIONS: GRIM19 deficiency may mediate ovulation and glucose metabolism disorders in PCOS patients. Our results suggest that GRIM19 may be a new target for diagnosis and treatment.


Assuntos
Doenças Metabólicas , Síndrome do Ovário Policístico , Animais , Feminino , Humanos , Camundongos , Linhagem Celular , Glucose/metabolismo , Células da Granulosa/metabolismo , Doenças Metabólicas/metabolismo , NADH NADPH Oxirredutases/metabolismo , Síndrome do Ovário Policístico/genética
12.
Proc Natl Acad Sci U S A ; 121(4): e2308960121, 2024 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-38232288

RESUMO

Metabolic disorders are characterized by an imbalance in muscle fiber composition, and a potential therapeutic approach involves increasing the proportion of oxidative muscle fibers. Prokineticin receptor 1 (PROKR1) is a G protein-coupled receptor that plays a role in various metabolic functions, but its specific involvement in oxidative fiber specification is not fully understood. Here, we investigated the functions of PROKR1 in muscle development to address metabolic disorders and muscular diseases. A meta-analysis revealed that the activation of PROKR1 upregulated exercise-responsive genes, particularly nuclear receptor subfamily 4 group A member 2 (NR4A2). Further investigations using ChIP-PCR, luciferase assays, and pharmacological interventions demonstrated that PROKR1 signaling enhanced NR4A2 expression by Gs-mediated phosphorylation of cyclic adenosine monophosphate (cAMP) response element-binding protein (CREB) in both mouse and human myotubes. Genetic and pharmacological interventions showed that the PROKR1-NR4A2 axis promotes the specification of oxidative muscle fibers in both myocytes by promoting mitochondrial biogenesis and metabolic function. Prokr1-deficient mice displayed unfavorable metabolic phenotypes, such as lower lean mass, enlarged muscle fibers, impaired glucose, and insulin tolerance. These mice also exhibited reduced energy expenditure and exercise performance. The deletion of Prokr1 resulted in decreased oxidative muscle fiber composition and reduced activity in the Prokr1-CREB-Nr4a2 pathway, which were restored by AAV-mediated Prokr1 rescue. In summary, our findings highlight the activation of the PROKR1-CREB-NR4A2 axis as a mechanism for increasing the oxidative muscle fiber composition, which positively impacts overall metabolic function. This study lays an important scientific foundation for the development of effective muscular-metabolic therapeutics with unique mechanisms of action.


Assuntos
Regulação da Expressão Gênica , Doenças Metabólicas , Camundongos , Animais , Humanos , Transdução de Sinais , Fibras Musculares Esqueléticas/metabolismo , Estresse Oxidativo , Doenças Metabólicas/metabolismo , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo
13.
Diabetes Metab J ; 48(1): 1-18, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38173375

RESUMO

Mitochondrial stress and the dysregulated mitochondrial unfolded protein response (UPRmt) are linked to various diseases, including metabolic disorders, neurodegenerative diseases, and cancer. Mitokines, signaling molecules released by mitochondrial stress response and UPRmt, are crucial mediators of inter-organ communication and influence systemic metabolic and physiological processes. In this review, we provide a comprehensive overview of mitokines, including their regulation by exercise and lifestyle interventions and their implications for various diseases. The endocrine actions of mitokines related to mitochondrial stress and adaptations are highlighted, specifically the broad functions of fibroblast growth factor 21 and growth differentiation factor 15, as well as their specific actions in regulating inter-tissue communication and metabolic homeostasis. Finally, we discuss the potential of physiological and genetic interventions to reduce the hazards associated with dysregulated mitokine signaling and preserve an equilibrium in mitochondrial stress-induced responses. This review provides valuable insights into the mechanisms underlying mitochondrial regulation of health and disease by exploring mitokine interactions and their regulation, which will facilitate the development of targeted therapies and personalized interventions to improve health outcomes and quality of life.


Assuntos
Doenças Metabólicas , Qualidade de Vida , Humanos , Mitocôndrias/metabolismo , Doenças Metabólicas/terapia , Doenças Metabólicas/metabolismo , Transdução de Sinais
14.
J Inherit Metab Dis ; 47(1): 22-40, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37254440

RESUMO

The liver is the metabolic center of the body and an ideal target for gene therapy of inherited metabolic disorders (IMDs). Adeno-associated viral (AAV) vectors can deliver transgenes to the liver with high efficiency and specificity and a favorable safety profile. Recombinant AAV vectors contain only the transgene cassette, and their payload is converted to non-integrating circular double-stranded DNA episomes, which can provide stable expression from months to years. Insights from cellular studies and preclinical animal models have provided valuable information about AAV capsid serotypes with a high liver tropism. These vectors have been applied successfully in the clinic, particularly in trials for hemophilia, resulting in the first approved liver-directed gene therapy. Lessons from ongoing clinical trials have identified key factors affecting efficacy and safety that were not readily apparent in animal models. Circumventing pre-existing neutralizing antibodies to the AAV capsid, and mitigating adaptive immune responses to transduced cells are critical to achieving therapeutic benefit. Combining the high efficiency of AAV delivery with genome editing is a promising path to achieve more precise control of gene expression. The primary safety concern for liver gene therapy with AAV continues to be the small risk of tumorigenesis from rare vector integrations. Hepatotoxicity is a key consideration in the safety of neuromuscular gene therapies which are applied at substantially higher doses. The current knowledge base and toolkit for AAV is well developed, and poised to correct some of the most severe IMDs with liver-directed gene therapy.


Assuntos
Fígado , Doenças Metabólicas , Animais , Fígado/metabolismo , Terapia Genética/métodos , Capsídeo/metabolismo , Transgenes/genética , Doenças Metabólicas/genética , Doenças Metabólicas/terapia , Doenças Metabólicas/metabolismo , Vetores Genéticos/genética , Dependovirus/genética
15.
Physiology (Bethesda) ; 39(2): 98-125, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38051123

RESUMO

The heart, once considered a mere blood pump, is now recognized as a multifunctional metabolic and endocrine organ. Its function is tightly regulated by various metabolic processes, at the same time it serves as an endocrine organ, secreting bioactive molecules that impact systemic metabolism. In recent years, research has shed light on the intricate interplay between the heart and other metabolic organs, such as adipose tissue, liver, and skeletal muscle. The metabolic flexibility of the heart and its ability to switch between different energy substrates play a crucial role in maintaining cardiac function and overall metabolic homeostasis. Gaining a comprehensive understanding of how metabolic disorders disrupt cardiac metabolism is crucial, as it plays a pivotal role in the development and progression of cardiac diseases. The emerging understanding of the heart as a metabolic and endocrine organ highlights its essential contribution to whole body metabolic regulation and offers new insights into the pathogenesis of metabolic diseases, such as obesity, diabetes, and cardiovascular disorders. In this review, we provide an in-depth exploration of the heart's metabolic and endocrine functions, emphasizing its role in systemic metabolism and the interplay between the heart and other metabolic organs. Furthermore, emerging evidence suggests a correlation between heart disease and other conditions such as aging and cancer, indicating that the metabolic dysfunction observed in these conditions may share common underlying mechanisms. By unraveling the complex mechanisms underlying cardiac metabolism, we aim to contribute to the development of novel therapeutic strategies for metabolic diseases and improve overall cardiovascular health.


Assuntos
Diabetes Mellitus , Doenças Metabólicas , Humanos , Diabetes Mellitus/metabolismo , Tecido Adiposo/metabolismo , Homeostase , Doenças Metabólicas/metabolismo , Transdução de Sinais
16.
J Physiol Biochem ; 80(2): 249-260, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38158555

RESUMO

N6-methyladenosine (m6A) is one of the most abundant epitranscriptomic modifications on eukaryotic mRNA. Evidence has highlighted that m6A is altered in response to inflammation-related factors and it is closely associated with various inflammation-related diseases. Multiple subpopulations of myeloid cells, such as macrophages, dendritic cells, and granulocytes, are crucial for the regulating of immune process in inflammation-related diseases. Recent studies have revealed that m6A plays an important regulatory role in the functional of multiple myeloid cells. In this review, we comprehensively summarize the function of m6A modification in myeloid cells from the perspective of myeloid cell production, activation, polarization, and migration. Furthermore, we discuss how m6A-mediated myeloid cell function affects the progression of inflammation-related diseases, including autoimmune diseases, chronic metabolic diseases, and malignant tumors. Finally, we discuss the challenges encountered in the study of m6A in myeloid cells, intended to provide a new direction for the study of the pathogenesis of inflammation-related diseases.


Assuntos
Adenosina , Adenosina/análogos & derivados , Inflamação , Células Mieloides , Adenosina/metabolismo , Humanos , Inflamação/metabolismo , Células Mieloides/metabolismo , Animais , Doenças Autoimunes/metabolismo , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Neoplasias/metabolismo , Neoplasias/patologia , Neoplasias/imunologia , Neoplasias/genética , Doenças Metabólicas/metabolismo , Doenças Metabólicas/imunologia , Doenças Metabólicas/patologia
17.
Obes Rev ; 25(3): e13677, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38114233

RESUMO

The induction of thermogenesis in brown adipose tissue is emerging as an attractive therapy for obesity and metabolic syndrome. However, the long-term efficacy and safety of clinical pharmaceutical agents have yet to be fully characterized. The transplantation of brown adipose tissue represents an alternative approach that might have a therapeutic effect by inducing a long-term increase in energy expenditure. However, limited tissue resources hinder the development of transplantation. Stem cell-based therapy and brown adipose tissue engineering, in addition to transplantation, represent alternative approaches that might resolve this problem. In this article, we discuss recent advances in understanding the mechanisms and applications of brown adipose tissue transplantation in the treatment of obesity and related metabolic disorders. Specifically, the induction of brown adipocytes and the fabrication of engineered brown adipose tissue as novel transplantation resources have long-term effects on ameliorating metabolic defects in rodent models. Additionally, we explore future prospects regarding the development of three-dimensional engineered brown adipose tissue and the associated challenges.


Assuntos
Tecido Adiposo Marrom , Doenças Metabólicas , Humanos , Tecido Adiposo Marrom/metabolismo , Obesidade/cirurgia , Obesidade/tratamento farmacológico , Adipócitos Marrons/metabolismo , Doenças Metabólicas/metabolismo , Termogênese , Metabolismo Energético
18.
Diabetes Obes Metab ; 26(3): 809-819, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38100156

RESUMO

Metabolic diseases have become a major threat to human health worldwide as a result of changing lifestyles. The exploration of the underlying molecular mechanisms of metabolic diseases and the development of improved therapeutic methods have been hindered by the lack of appropriate human experimental models. Organoids are three-dimensional in vitro models of self-renewing cells that spontaneously self-organize into structures similar to the corresponding in vivo tissues, recapitulating the original tissue function. Off-body organoid technology has been successfully applied to disease modelling, developmental biology, regenerative medicine, and tumour precision medicine. This new generation of biological models has received widespread attention. This article focuses on the construction process and research progress with regard to organoids related to metabolic diseases in recent years, and looks forward to their prospective applications.


Assuntos
Doenças Metabólicas , Neoplasias , Humanos , Organoides/metabolismo , Modelos Biológicos , Neoplasias/metabolismo , Medicina de Precisão , Doenças Metabólicas/terapia , Doenças Metabólicas/metabolismo
19.
Int J Mol Sci ; 24(23)2023 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-38068890

RESUMO

In recent years, the effects of androgens on metabolic and body weight regulation systems and their underlying mechanisms have been gradually revealed in females. In women and experimental animals of reproductive age, androgen excess can adversely affect metabolic functioning, appetite, and body weight regulation. In addition, excess androgens can increase the risk of metabolic disorders, such as obesity, insulin resistance, and diabetes. These unfavorable effects of androgens are induced by alterations in the actions of hypothalamic appetite-regulatory factors, reductions in energy expenditure, insulin resistance in skeletal muscle, and ß-cell dysfunction. Interestingly, these unfavorable effects of androgens on metabolic and body-weight regulation systems are neither observed nor evident in ovariectomized animals and post-menopausal women, indicating that the adverse effects of androgens might be dependent on the estrogen milieu. Recent findings may provide novel sex- and age-specific strategies for treating metabolic diseases.


Assuntos
Resistência à Insulina , Doenças Metabólicas , Síndrome do Ovário Policístico , Animais , Humanos , Feminino , Androgênios/farmacologia , Androgênios/metabolismo , Resistência à Insulina/fisiologia , Obesidade/metabolismo , Doenças Metabólicas/etiologia , Doenças Metabólicas/metabolismo , Animais de Laboratório/metabolismo , Síndrome do Ovário Policístico/metabolismo
20.
J Agric Food Chem ; 71(51): 20701-20712, 2023 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-38088361

RESUMO

Purple Pennisetum (Pennisetum purpureum Schumach), a hybrid between Taihucao No. 2 and the local wild species of purple Pennisetum, has dark red stems and leaves due to its anthocyanin content. This study explores the potential of purple napiergrass extracts (PNE) in alleviating obesity and metabolic disorders induced by a high-fat diet in mice, where 50% of the caloric content is derived from fat. Mice were orally administered low-dose or high-dose PNE alongside a high-fat diet. Experimental findings indicate that PNE attenuated weight gain, reduced liver, and adipose tissue weight, and lowered blood cholesterol, triglyceride, low-density lipoprotein, and blood sugar levels. Stained sections showed that PNE inhibited lipid accumulation and fat hypertrophy in the liver. Immunoblotting analysis suggested that PNE improved the inflammatory response associated with obesity, dyslipidemia, and hyperglycemia induced by a high-fat diet. Furthermore, PNE potentially functions as a PPAR-γ agonist, increasing the adiponectin (ADIPOQ) concentration and suppressing inflammatory factors, while elevating the anti-inflammatory factor interleukin-10 (IL-10) in the liver. PNE-treated mice showed enhanced activation of the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) and AMP-activated protein kinase (AMPK) pathways and increased fatty acid oxidation and liver lipolysis. In conclusion, this study elucidated the mechanisms underlying the anti-inflammatory, PI3K/Akt, and AMPK pathways in a high-fat diet-induced obesity model. These findings highlight the potential of PNE in reducing weight, inhibiting inflammation, and improving blood sugar and lipid levels, showing the potential for addressing obesity-related metabolic disorders in humans.


Assuntos
Doenças Metabólicas , Pennisetum , Humanos , Camundongos , Animais , Proteínas Proto-Oncogênicas c-akt/metabolismo , Pennisetum/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Dieta Hiperlipídica/efeitos adversos , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Glicemia/metabolismo , Extratos Vegetais/farmacologia , Obesidade/tratamento farmacológico , Obesidade/etiologia , Fígado/metabolismo , Triglicerídeos/metabolismo , Água/metabolismo , Doenças Metabólicas/tratamento farmacológico , Doenças Metabólicas/etiologia , Doenças Metabólicas/metabolismo , Anti-Inflamatórios/metabolismo , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA