Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Nat Commun ; 15(1): 4755, 2024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38834568

RESUMO

Non-alcoholic steatohepatitis (NASH) is a severe type of the non-alcoholic fatty liver disease (NAFLD). NASH is a growing global health concern due to its increasing morbidity, lack of well-defined biomarkers and lack of clinically effective treatments. Using metabolomic analysis, the most significantly changed active lipid sphingosine d18:1 [So(d18:1)] is selected from NASH patients. So(d18:1) inhibits macrophage HIF-2α as a direct inhibitor and promotes the inflammatory factors secretion. Male macrophage-specific HIF-2α knockout and overexpression mice verified the protective effect of HIF-2α on NASH progression. Importantly, the HIF-2α stabilizer FG-4592 alleviates liver inflammation and fibrosis in NASH, which indicated that macrophage HIF-2α is a potential drug target for NASH treatment. Overall, this study confirms that So(d18:1) promotes NASH and clarifies that So(d18:1) inhibits the transcriptional activity of HIF-2α in liver macrophages by suppressing the interaction of HIF-2α with ARNT, suggesting that macrophage HIF-2α may be a potential target for the treatment of NASH.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos , Macrófagos , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica , Esfingosina , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Hepatopatia Gordurosa não Alcoólica/patologia , Hepatopatia Gordurosa não Alcoólica/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Masculino , Macrófagos/metabolismo , Macrófagos/efeitos dos fármacos , Humanos , Camundongos , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Fígado/metabolismo , Fígado/patologia , Fígado/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Translocador Nuclear Receptor Aril Hidrocarboneto/metabolismo , Translocador Nuclear Receptor Aril Hidrocarboneto/genética , Cirrose Hepática/metabolismo , Cirrose Hepática/tratamento farmacológico , Cirrose Hepática/patologia , Cirrose Hepática/genética , Modelos Animais de Doenças
2.
J Ovarian Res ; 17(1): 100, 2024 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-38734641

RESUMO

BACKGROUND: Polycystic ovary syndrome (PCOS) is a reproductive endocrine disorder with multiple metabolic abnormalities. Most PCOS patients have concomitant metabolic syndromes such as insulin resistance and obesity, which often lead to the development of type II diabetes and cardiovascular disease with serious consequences. Current treatment of PCOS with symptomatic treatments such as hormone replacement, which has many side effects. Research on its origin and pathogenesis is urgently needed. Although improving the metabolic status of the body can alleviate reproductive function in some patients, there is still a subset of patients with metabolically normal PCOS that lacks therapeutic tools to address ovarian etiology. METHODS: The effect of IL-22 on PCOS ovarian function was verified in a non-metabolic PCOS mouse model induced by dehydroepiandrosterone (DHEA) and rosiglitazone, as well as granulosa cell -specific STAT3 knockout (Fshrcre+Stat3f/f) mice (10 groups totally and n = 5 per group). Mice were maintained under controlled temperature and lighting conditions with free access to food and water in a specific pathogen-free (SPF) facility. Secondary follicles separated from Fshrcre+Stat3f/f mice were cultured in vitro with DHEA to mimic the hyperandrogenic environment in PCOS ovaries (4 groups and n = 7 per group) and then were treated with IL-22 to investigate the specific role of IL-22 on ovarian function. RESULTS: We developed a non-metabolic mice model with rosiglitazone superimposed on DHEA. This model has normal metabolic function as evidenced by normal glucose tolerance without insulin resistance and PCOS-like ovarian function as evidenced by irregular estrous cycle, polycystic ovarian morphology (PCOM), abnormalities in sex hormone level. Supplementation with IL-22 improved these ovarian functions in non-metabolic PCOS mice. Application of DHEA in an in vitro follicular culture system to simulate PCOS follicular developmental block and ovulation impairment. Follicles from Fshrcre+Stat3f/f did not show improvement in POCS follicle development with the addition of IL-22. In DHEA-induced PCOS mice, selective ablation of STAT3 in granulosa cells significantly reversed the ameliorative effect of IL-22 on ovarian function. CONCLUSION: IL-22 can improve non-metabolic PCOS mice ovarian function. Granulosa cells deficient in STAT3 reverses the role of IL-22 in alleviating ovary dysfunction in non-metabolic PCOS mice.


Assuntos
Modelos Animais de Doenças , Interleucina 22 , Interleucinas , Ovário , Síndrome do Ovário Policístico , Feminino , Animais , Síndrome do Ovário Policístico/metabolismo , Camundongos , Interleucinas/metabolismo , Interleucinas/genética , Ovário/metabolismo , Ovário/patologia , Desidroepiandrosterona/farmacologia , Fator de Transcrição STAT3/metabolismo , Rosiglitazona/farmacologia , Rosiglitazona/uso terapêutico , Células da Granulosa/metabolismo , Camundongos Knockout
3.
Nat Metab ; 6(5): 947-962, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38769396

RESUMO

Polycystic ovary syndrome (PCOS), an endocrine disorder afflicting 6-20% of women of reproductive age globally, has been linked to alterations in the gut microbiome. We previously showed that in PCOS, elevation of Bacteroides vulgatus in the gut microbiome was associated with altered bile acid metabolism. Here we show that B. vulgatus also induces a PCOS-like phenotype in female mice via an alternate mechanism independent of bile acids. We find that B. vulgatus contributes to PCOS-like symptoms through its metabolite agmatine, which is derived from arginine by arginine decarboxylase. Mechanistically, agmatine activates the farnesoid X receptor (FXR) pathway to subsequently inhibit glucagon-like peptide-1 (GLP-1) secretion by L cells, which leads to insulin resistance and ovarian dysfunction. Critically, the GLP-1 receptor agonist liraglutide and the arginine decarboxylase inhibitor difluoromethylarginine ameliorate ovarian dysfunction in a PCOS-like mouse model. These findings reveal that agmatine-FXR-GLP-1 signalling contributes to ovarian dysfunction, presenting a potential therapeutic target for PCOS management.


Assuntos
Agmatina , Microbioma Gastrointestinal , Síndrome do Ovário Policístico , Receptores Citoplasmáticos e Nucleares , Síndrome do Ovário Policístico/tratamento farmacológico , Síndrome do Ovário Policístico/metabolismo , Animais , Feminino , Camundongos , Agmatina/farmacologia , Agmatina/metabolismo , Agmatina/uso terapêutico , Receptores Citoplasmáticos e Nucleares/agonistas , Receptores Citoplasmáticos e Nucleares/metabolismo , Microbioma Gastrointestinal/efeitos dos fármacos , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Transdução de Sinais/efeitos dos fármacos , Modelos Animais de Doenças , Resistência à Insulina , Bacteroides/efeitos dos fármacos , Humanos , Carboxiliases/metabolismo
4.
Cell ; 187(11): 2717-2734.e33, 2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38653239

RESUMO

The gut microbiota has been found to play an important role in the progression of metabolic dysfunction-associated steatohepatitis (MASH), but the mechanisms have not been established. Here, by developing a click-chemistry-based enrichment strategy, we identified several microbial-derived bile acids, including the previously uncharacterized 3-succinylated cholic acid (3-sucCA), which is negatively correlated with liver damage in patients with liver-tissue-biopsy-proven metabolic dysfunction-associated fatty liver disease (MAFLD). By screening human bacterial isolates, we identified Bacteroides uniformis strains as effective producers of 3-sucCA both in vitro and in vivo. By activity-based protein purification and identification, we identified an enzyme annotated as ß-lactamase in B. uniformis responsible for 3-sucCA biosynthesis. Furthermore, we found that 3-sucCA is a lumen-restricted metabolite and alleviates MASH by promoting the growth of Akkermansia muciniphila. Together, our data offer new insights into the gut microbiota-liver axis that may be leveraged to augment the management of MASH.


Assuntos
Akkermansia , Bacteroides , Ácidos e Sais Biliares , Microbioma Gastrointestinal , Hepatopatia Gordurosa não Alcoólica , Simbiose , Animais , Humanos , Masculino , Camundongos , Akkermansia/metabolismo , Bacteroides/metabolismo , beta-Lactamases/metabolismo , Ácidos e Sais Biliares/metabolismo , Vias Biossintéticas/genética , Fígado Gorduroso/metabolismo , Fígado/metabolismo , Camundongos Endogâmicos C57BL , Verrucomicrobia/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/microbiologia
5.
Science ; 381(6657): eadd5787, 2023 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-37535747

RESUMO

A mechanistic understanding of how microbial proteins affect the host could yield deeper insights into gut microbiota-host cross-talk. We developed an enzyme activity-screening platform to investigate how gut microbiota-derived enzymes might influence host physiology. We discovered that dipeptidyl peptidase 4 (DPP4) is expressed by specific bacterial taxa of the microbiota. Microbial DPP4 was able to decrease the active glucagon like peptide-1 (GLP-1) and disrupt glucose metabolism in mice with a leaky gut. Furthermore, the current drugs targeting human DPP4, including sitagliptin, had little effect on microbial DPP4. Using high-throughput screening, we identified daurisoline-d4 (Dau-d4) as a selective microbial DPP4 inhibitor that improves glucose tolerance in diabetic mice.


Assuntos
Bacteroides , Diabetes Mellitus Tipo 2 , Dipeptidil Peptidase 4 , Inibidores da Dipeptidil Peptidase IV , Microbioma Gastrointestinal , Interações entre Hospedeiro e Microrganismos , Hipoglicemiantes , Animais , Humanos , Camundongos , Bacteroides/efeitos dos fármacos , Bacteroides/enzimologia , Bacteroides/genética , Diabetes Mellitus Tipo 2/enzimologia , Diabetes Mellitus Tipo 2/microbiologia , Dipeptidil Peptidase 4/metabolismo , Inibidores da Dipeptidil Peptidase IV/farmacologia , Inibidores da Dipeptidil Peptidase IV/uso terapêutico , Fezes/microbiologia , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Glucose/metabolismo , Isoenzimas/metabolismo , Fosfato de Sitagliptina/farmacologia , Fosfato de Sitagliptina/uso terapêutico , Hipoglicemiantes/farmacologia , Hipoglicemiantes/uso terapêutico
6.
Acta Pharm Sin B ; 12(4): 1899-1912, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35847503

RESUMO

Atherosclerosis is a chronic multifactorial cardiovascular disease. Western diets have been reported to affect atherosclerosis through regulating adipose function. In high cholesterol diet-fed ApoE -/- mice, adipocyte HIF-1α deficiency or direct inhibition of HIF-1α by the selective pharmacological HIF-1α inhibitor PX-478 alleviates high cholesterol diet-induced atherosclerosis by reducing adipose ceramide generation, which lowers cholesterol levels and reduces inflammatory responses, resulting in improved dyslipidemia and atherogenesis. Smpd3, the gene encoding neutral sphingomyelinase, is identified as a new target gene directly regulated by HIF-1α that is involved in ceramide generation. Injection of lentivirus-SMPD3 in epididymal adipose tissue reverses the decrease in ceramides in adipocytes and eliminates the improvements on atherosclerosis in the adipocyte HIF-1α-deficient mice. Therefore, HIF-1α inhibition may constitute a novel approach to slow atherosclerotic progression.

7.
J Reprod Immunol ; 151: 103628, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35472833

RESUMO

Polycystic ovary syndrome (PCOS) is a common endocrine/reproductive/metabolic disorder. The etiology of PCOS is complex and has been linked to low-grade chronic inflammation. Local inflammation of the ovary affects ovulation and induces or aggravates systemic inflammation. PCOS patients demonstrated significantly higher concentrations of circulating inflammatory cells, such as lymphocytes, neutrophils, eosinophilic granulocytes, monocytes and Th17 cells than women without PCOS, while the percentage of Treg cells was lower. Inflammatory factors, such as serum CRP, hs-CRP, IL-1Ra, IL-6, IL-17A, IL-17F, IL-18, IL-23, TNF-α, α-1 acid glycoprotein,monocyte chemoattractant protein-1 (MCP-1) and adipokines and their paralogs, including chemerin, C1q and TNF-related 6 (C1QTNF6), were also found to be significantly increased in the peripheral blood of PCOS patients. Levels of anti-inflammatory cytokines, such as IL-10, IL-17E, IL-27, IL-35 and IL-37, TGF-ß, omentin-1, Secreted frizzled-related protein5 (SFRP5) were significantly lower. An analogous situation occurs locally in the ovary. Some vital inflammatory cells and cytokines may initially be released from the ovary and then enter the circulation. The systemic inflammation underlying PCOS is thought to interact with obesity, insulin resistance (IR) and hyperandrogenism. Traditional Chinese medicine, multitargeted treatment, anti-inflammatory and antioxidant medicine, and lifestyle modification can benefit PCOS women by alleviating inflammatory responses.


Assuntos
Hiperandrogenismo , Resistência à Insulina , Síndrome do Ovário Policístico , Anti-Inflamatórios/uso terapêutico , Colágeno/uso terapêutico , Citocinas , Feminino , Humanos , Inflamação
8.
Front Endocrinol (Lausanne) ; 13: 815968, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35237237

RESUMO

Polycystic ovary syndrome (PCOS) is a common reproductive endocrine disease in women of reproductive age. Ovarian dysfunction including abnormal steroid hormone synthesis and follicular arrest play a vital role in PCOS pathogenesis. Hyperandrogenemia is one of the important characteristics of PCOS. However, the mechanism of regulation and interaction between hyperandrogenism and ovulation abnormalities are not clear. To investigate androgen-related metabolic state in granulosa cells of PCOS patients, we identified the transcriptome characteristics of PCOS granulosa cells by RNA-seq. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis of differentially expressed genes (DEGs) revealed that genes enriched in lipid metabolism pathway, fatty acid biosynthetic process and ovarian steroidogenesis pathway were abnormally expressed in PCOS granulosa cells in comparison with that in control. There are close interactions among these three pathways as identified by analysis of the protein-protein interaction (PPI) network of DEGs. Furthermore, in vitro mouse follicle culture system was established to explore the effect of high androgen and its related metabolic dysfunction on follicular growth and ovulation. RT-qPCR results showed that follicles cultured with dehydroepiandrosterone (DHEA) exhibited decreased expression levels of cumulus expansion-related genes (Has2, Ptx3, Tnfaip6 and Adamts1) and oocyte maturation-related genes (Gdf9 and Bmp15), which may be caused by impaired steroid hormone synthesis and lipid metabolism, thus inhibited follicular development and ovulation. Furthermore, the inhibition effect of DHEA on follicle development and ovulation was ameliorated by flutamide, an androgen receptor (AR) antagonist, suggesting the involvement of AR signaling. In summary, our study offers new insights into understanding the role of androgen excess induced granulosa cell metabolic disorder in ovarian dysfunction of PCOS patients.


Assuntos
Androgênios , Síndrome do Ovário Policístico , Androgênios/farmacologia , Animais , Desidroepiandrosterona , Feminino , Células da Granulosa , Humanos , Camundongos , Síndrome do Ovário Policístico/metabolismo , Esteroides
9.
Am J Obstet Gynecol ; 226(6): 835.e1-835.e17, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-34919894

RESUMO

BACKGROUND: Postpartum hemorrhage remains the leading cause of maternal mortality. However, there is an insufficient understanding of atonic postpartum hemorrhage. Uterine atony is the most common cause of postpartum hemorrhage. Although an association between myometrium inflammatory cytokines and atonic postpartum hemorrhage has been demonstrated preliminarily, it is not clinically useful in predicting postpartum hemorrhage. Plasma is more readily available, and the assessment of its inflammatory status is more relevant to biological markers of postpartum hemorrhage and might explain the pathophysiology of atonic postpartum hemorrhage. OBJECTIVE: Our objective was to examine changes in maternal plasma cytokines in women with atonic postpartum hemorrhage. STUDY DESIGN: This was a retrospective longitudinal case-control study of pregnant women with singleton gestations at term undergoing vaginal delivery. Cases were women with atonic postpartum hemorrhage, and 1:1 propensity-score matching was used to match the control group. Maternal plasma was collected in the first trimester, early third trimester, and late third trimester, and multiplex Luminex assay was used to determine the cytokine concentrations. Multivariate logistic regressions were used to determine the association between maternal cytokines at different stages of pregnancy and atonic postpartum hemorrhage. RESULTS: A total of 36 pregnant women met the clinical diagnostic criteria for atonic postpartum hemorrhage, and 36 patients without postpartum hemorrhage were matched as the control group. Concentrations were lower for most cytokines in the atonic postpartum hemorrhage group in the first and early third trimester. However, in the late third trimester, higher plasma concentrations of basic fibroblast growth factor, interleukin-1 alpha, interleukin-1 beta, interleukin-1 receptor antagonist, interleukin-2 receptor alpha, interleukin-16, interleukin-18, macrophage colony stimulating factor, macrophage inflammatory protein-1 alpha, beta-nerve growth factor, tumor necrosis factor-related apoptosis-induced ligand, and stem cell factor were significantly associated with increased risk of atonic postpartum hemorrhage. Multiple testing correction showed that basic fibroblast growth factor (P<.001; fold change [FC]=1.16), macrophage inflammatory protein-1 alpha (P<.001; FC=1.15), and stem cell factor (P=.001; FC=1.25) had the most significant difference (P<.001). The prediction model of atonic postpartum hemorrhage constructed by these significantly changed cytokines had a high predictive efficiency (area under the curve, 0.84; sensitivity, 0.78; specificity, 0.83; +likelihood ratio, 4.66; -likelihood ratio, 0.27). CONCLUSION: Higher concentrations of maternal plasma cytokines in the late third trimester are associated with high risk of subsequent atonic postpartum hemorrhage. These indicators may be potential biomarkers for predicting atonic postpartum hemorrhage.


Assuntos
Citocinas , Hemorragia Pós-Parto , Biomarcadores , Estudos de Casos e Controles , Citocinas/sangue , Feminino , Fator 2 de Crescimento de Fibroblastos , Humanos , Estudos Longitudinais , Hemorragia Pós-Parto/epidemiologia , Gravidez , Estudos Retrospectivos , Fator de Células-Tronco
11.
Cell Rep ; 36(8): 109607, 2021 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-34433035

RESUMO

The interrelation between hypoxia and immune response has pivotal roles in the pathogenesis of chronic metabolic diseases. However, the role of macrophage HIF-2α in NLRP3 inflammasome activation remains unclear. Here, we show that deficiency of HIF-2α in macrophages results in excessive activation of the NLRP3 inflammasome in a manner dependent on CPT1A-mediated enhancement of fatty acid oxidation (FAO). Mechanistically, HIF-2α binds directly to the Cpt1a promoter and is involved in the regulation of H3K27me3 methylation during NLRP3 inflammasome activation. Myeloid-specific Hif2α knockout mice exhibit exacerbated insulin resistance and increased activation of NLRP3 inflammasome in macrophages. Overexpression of the Hif2α gene or stabilization of the protein by FG-4592 ameliorates insulin resistance and reduces NLRP3 inflammasome activation in macrophages. Taken together, our results suggest that macrophage HIF-2α inhibits FAO-mediated activation of the NLRP3 inflammasome and alleviates insulin resistance.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Inflamassomos/metabolismo , Resistência à Insulina/fisiologia , Macrófagos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Animais , Humanos , Metabolismo dos Lipídeos/fisiologia , Camundongos
12.
Front Endocrinol (Lausanne) ; 12: 667422, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34122341

RESUMO

Polycystic ovary syndrome (PCOS) is a common reproductive endocrine disease. PCOS patients are characterized by hyperandrogenemia, anovulation, and metabolic dysfunction. Hypothalamus-pituitary-ovary axis imbalance is considered as an important pathophysiology underlying PCOS, indicating that central modulation, especially the abnormal activation of hypothalamic GnRH neurons plays a vital role in PCOS development. Increased GnRH pulse frequency can promote LH secretion, leading to ovarian dysfunction and abnormal sex steroids synthesis. By contrast, peripheral sex steroids can modulate the action of GnRH neurons through a feedback effect, which is impaired in PCOS, thus forming a vicious cycle. Additionally, hypothalamic GnRH neurons not only serve as the final output pathway of central control of reproductive axis, but also as the central connection point where reproductive function and metabolic state inter-regulate with each other. Metabolic factors, such as insulin resistance and obesity in PCOS patients can regulate GnRH neurons activity, and ultimately regulate reproductive function. Besides, gut hormones act on both brain and peripheral organs to modify metabolic state. Gut microbiota disturbance is also related to many metabolic diseases and has been reported to play an essential part in PCOS development. This review concludes with the mechanism of central modulation and the interaction between neuroendocrine factors and reproductive or metabolic disorders in PCOS development. Furthermore, the role of the gut microenvironment as an important part involved in the abnormal neuronal-reproductive-metabolic circuits that contribute to PCOS is discussed, thus offering possible central and peripheral therapeutic targets for PCOS patients.


Assuntos
Doenças Metabólicas/fisiopatologia , Neurônios/patologia , Síndrome do Ovário Policístico/patologia , Reprodução , Feminino , Humanos , Síndrome do Ovário Policístico/etiologia
13.
Gut Microbes ; 13(1): 1-21, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33722164

RESUMO

As the gut microbiota exerts various effects on the intestinal milieu which influences distant organs and pathways, it is considered to be a full-fledged endocrine organ. The microbiota plays a major role in the reproductive endocrine system throughout a woman's lifetime by interacting with estrogen, androgens, insulin, and other hormones. Imbalance of the gut microbiota composition can lead to several diseases and conditions, such as pregnancy complications, adverse pregnancy outcomes, polycystic ovary syndrome (PCOS), endometriosis, and cancer; however, research on the mechanisms is limited. More effort should be concentrated on exploring the potential causes and underlying the mechanisms of microbiota-hormone-mediated disease, and providing novel therapeutic and preventive strategies.As the gut microbiota exerts various effects on the intestinal milieu which influences distant organs and pathways, it is considered to be a full-fledged endocrine organ. The microbiota plays a major role in the reproductive endocrine system throughout a woman's lifetime by interacting with estrogen, androgens, insulin, and other hormones. Imbalance of the gut microbiota composition can lead to several diseases and conditions, such as pregnancy complications, adverse pregnancy outcomes, polycystic ovary syndrome (PCOS), endometriosis, and cancer; however, research on the mechanisms is limited. More effort should be concentrated on exploring the potential causes and underlying the mechanisms of microbiota-hormone-mediated disease, and providing novel therapeutic and preventive strategies.


Assuntos
Sistema Endócrino/fisiologia , Microbioma Gastrointestinal/fisiologia , Doenças dos Genitais Femininos/microbiologia , Genitália Feminina/fisiologia , Complicações na Gravidez/microbiologia , Androgênios/metabolismo , Sistema Endócrino/metabolismo , Estrogênios/metabolismo , Feminino , Hormônios/metabolismo , Humanos , Insulina/metabolismo , Gravidez
14.
J Endocrinol ; 245(2): 281-289, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32163914

RESUMO

Polycystic ovary syndrome (PCOS) is a complex syndrome involving both endocrine and metabolic disorders. Gut microbiota and the intestinal immune factor IL-22 play an important role in the pathogenesis of PCOS. However, the therapeutic role of IL-22 in high androgen-induced PCOS mice is not clear. We aimed to determine the therapeutic effects of IL-22 on the DHEA-induced PCOS mouse model and to explore the possible mechanism of IL-22 in regulating hyperandrogenism-associated PCOS. Insulin resistance levels and ovarian functions were investigated in DHEA-induced PCOS mice with or without additional IL-22 treatment. We found that IL-22 could reverse insulin resistance, disturbed estrous cycle, abnormal ovary morphology, and decreased embryo number in DHEA mice. Mechanistically, IL-22 upregulated the browning of white adipose tissue in DHEA mice. This study demonstrated that IL-22-associated browning of white adipose tissue regulated insulin sensitivity and ovarian functions in PCOS, suggesting that IL-22 may be of value for the treatment of PCOS with a hyperandrogenism phenotype.


Assuntos
Interleucinas/farmacologia , Síndrome do Ovário Policístico/tratamento farmacológico , Androgênios , Animais , Modelos Animais de Doenças , Ciclo Estral/efeitos dos fármacos , Feminino , Hiperandrogenismo/induzido quimicamente , Hiperandrogenismo/complicações , Resistência à Insulina/fisiologia , Camundongos , Ovário/fisiopatologia , Síndrome do Ovário Policístico/induzido quimicamente , Síndrome do Ovário Policístico/fisiopatologia , Interleucina 22
15.
Mol Carcinog ; 59(7): 679-690, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-31912614

RESUMO

The immune-suppressive tumor microenvironment promotes metastatic spread and outgrowth. One of the major contributors is tumor-associated myeloid cells. However, the molecular mechanisms regulating their differentiation and function are not well understood. Here we report lamin A/C, a nuclear lamina protein associated with chromatin remodeling, was one of the critical regulators in cellular reprogramming of tumor-associated myeloid cells. Using myeloid-specific lamin A/C knockout mice and triple-negative breast cancer (TNBC) mouse models, we discovered that the loss of lamin A/C drives CD11b+ Ly6G+ granulocytic lineage differentiation, alters the production of inflammatory chemokines, decreases host antitumor immunity, and increases metastasis. The underlying mechanisms involve an increased H3K4me3 leading to the upregulation of transcription factors (TFs) Gfi-1 and C/EBPε. Decreased lamin A/C and increased Gfi-1 and C/EBPε were also found in the granulocytic subset in the peripheral blood of human cancer patients. Our data provide a mechanistic understanding of myeloid lineage differentiation and function in the immune-suppressive microenvironment in TNBC metastasis.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/genética , Diferenciação Celular/genética , Lamina Tipo A/genética , Neoplasias Pulmonares/genética , Células Mieloides/patologia , Metástase Neoplásica/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Repressoras/genética , Animais , Células Cultivadas , Quimiocinas/genética , Modelos Animais de Doenças , Feminino , Granulócitos/patologia , Humanos , Inflamação/genética , Inflamação/patologia , Leucócitos Mononucleares/patologia , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Metástase Neoplásica/patologia , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia , Microambiente Tumoral/genética , Regulação para Cima/genética
16.
EBioMedicine ; 49: 291-304, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31640947

RESUMO

BACKGROUND: Aortic dissection is a severe inflammatory vascular disease with high mortality and limited therapeutic options. The hallmarks of aortic dissection comprise aortic inflammatory cell infiltration and elastic fiber disruption, highlighting the involvement of macrophage. Here a role for macrophage hypoxia-inducible factor 1-alpha (HIF-1α) in aortic dissection was uncovered. METHODS: Immunochemistry, immunofluorescence, western blot and qPCR were performed to test the change of macrophage HIF-1α in two kinds of aortic dissection models and human tissues. Metabolomics and Seahorse extracellular flux analysis were used to detect the metabolic state of macrophages involved in the development of aortic dissection. Chromatin Immunoprecipitation (ChIP), enzyme-linked immunosorbent assay (ELISA) and cytometric bead array (CBA) were employed for mechanistic studies. FINDINGS: Macrophages involved underwent distinct metabolic reprogramming, especially fumarate accumulation, thus inducing HIF-1α activation in the development of aortic dissection in human and mouse models. Mechanistic studies revealed that macrophage HIF-1α activation triggered vascular inflammation, extracellular matrix degradation and elastic plate breakage through increased a disintegrin and metallopeptidase domain 17 (ADAM17), identified as a novel target gene of HIF-1α. A HIF-1α specific inhibitor acriflavine elicited protective effects on aortic dissection dependent on macrophage HIF-1α. INTERPRETATION: This study reveals that macrophage metabolic reprogramming activates HIF-1α and subsequently promotes aortic dissection progression, suggesting that macrophage HIF-1α inhibition might be a potential therapeutic target for treating aortic dissection.


Assuntos
Proteína ADAM17/metabolismo , Dissecção Aórtica/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Macrófagos/metabolismo , Acriflavina/farmacologia , Acriflavina/uso terapêutico , Aminopropionitrilo , Dissecção Aórtica/tratamento farmacológico , Dissecção Aórtica/patologia , Angiotensina II/farmacologia , Animais , Modelos Animais de Doenças , Progressão da Doença , Células HEK293 , Humanos , Macrófagos/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Regulação para Cima/efeitos dos fármacos
17.
Nat Med ; 25(8): 1225-1233, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31332392

RESUMO

Polycystic ovary syndrome (PCOS) is characterized by androgen excess, ovulatory dysfunction and polycystic ovaries1, and is often accompanied by insulin resistance2. The mechanism of ovulatory dysfunction and insulin resistance in PCOS remains elusive, thus limiting the development of therapeutics. Improved metabolic health is associated with a relatively high microbiota gene content and increased microbial diversity3,4. This study aimed to investigate the impact of the gut microbiota and its metabolites on the regulation of PCOS-associated ovarian dysfunction and insulin resistance. Here, we report that Bacteroides vulgatus was markedly elevated in the gut microbiota of individuals with PCOS, accompanied by reduced glycodeoxycholic acid and tauroursodeoxycholic acid levels. Transplantation of fecal microbiota from women with PCOS or B. vulgatus-colonized recipient mice resulted in increased disruption of ovarian functions, insulin resistance, altered bile acid metabolism, reduced interleukin-22 secretion and infertility. Mechanistically, glycodeoxycholic acid induced intestinal group 3 innate lymphoid cell IL-22 secretion through GATA binding protein 3, and IL-22 in turn improved the PCOS phenotype. This finding is consistent with the reduced levels of IL-22 in individuals with PCOS. This study suggests that modifying the gut microbiota, altering bile acid metabolism and/or increasing IL-22 levels may be of value for the treatment of PCOS.


Assuntos
Ácidos e Sais Biliares/metabolismo , Fator de Transcrição GATA3/fisiologia , Microbioma Gastrointestinal , Interleucinas/fisiologia , Síndrome do Ovário Policístico/etiologia , Animais , Feminino , Humanos , Inflamação/complicações , Resistência à Insulina , Camundongos , Camundongos Endogâmicos C57BL , Ovário/fisiopatologia , Síndrome do Ovário Policístico/fisiopatologia , Interleucina 22
18.
Sci Rep ; 8(1): 12949, 2018 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-30154492

RESUMO

Human polycystic ovary syndrome (PCOS) is a highly heritable disease regulated by genetic and environmental factors. Identifying PCOS genes is time consuming and costly in wet-lab. Developing an algorithm to predict PCOS candidates will be helpful. In this study, for the first time, we systematically analyzed properties of human PCOS genes. Compared with genes not yet known to be involved in PCOS regulation, known PCOS genes display distinguishing characteristics: (i) they tend to be located at network center; (ii) they tend to interact with each other; (iii) they tend to enrich in certain biological processes. Based on these features, we developed a machine-learning algorithm to predict new PCOS genes. 233 PCOS candidates were predicted with a posterior probability >0.9. Evidence supporting 7 of the top 10 predictions has been found.


Assuntos
Bases de Dados de Ácidos Nucleicos , Redes Reguladoras de Genes , Aprendizado de Máquina , Síndrome do Ovário Policístico/genética , Feminino , Humanos , Síndrome do Ovário Policístico/metabolismo
19.
Reprod Sci ; 25(12): 1629-1636, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29439619

RESUMO

BACKGROUND: Hyperhomocysteinemia, chronic low-grade inflammation, and insulin resistance are predominant features in women with polycystic ovary syndrome (PCOS). CD14++CD16+ inflammatory monocytes are elevated in cardiovascular diseases and diabetes, in which inflammation is one of the pathogenesis. We aimed to determine whether homocysteine levels are associated with monocyte subtypes or insulin resistance in women with PCOS. METHODS: A cross-sectional study was conducted between December 2014 and June 2015 at Peking University Third Hospital. Among 196 Chinese patients with PCOS enrolled, 102 had homocysteine levels ≥10 µmol/L and 94 exhibited normal homocysteine levels. Monocyte surface markers and related cytokines were detected in peripheral blood samples using a flow cytometry, and data on insulin resistance and homocysteine levels were collected. RESULTS: Our results showed that fasting insulin and homeostasis model assessment indices that reflect the severity of insulin resistance were increased in the hyperhomocysteinemia PCOS group. Simple linear regression analysis revealed that homocysteine level is one of the influence factors of insulin resistance in PCOS. Compared with the normal homocysteine group, patients with hyperhomocysteinemia had increased numbers of CD14++CD16+ inflammatory monocyte in their peripheral blood and elevated plasma levels of interleukin-1ß and interleukin-6, 2 typical cytokines secreted by the inflammatory monocytes. Unlike CD14+CD16++ nonclassical monocytes, CD14++CD16+ inflammatory monocytes are characterized by high expression of Human leukocyte antigen-antigen D related (HLA-DR). CONCLUSIONS: Our findings indicate that CD14++CD16+ inflammatory monocytes are associated with hyperhomocysteinemia and insulin resistance in patients with PCOS. Notably, CD14++CD16+ monocytes may contribute to the pathogenesis of insulin resistance in women with PCOS.


Assuntos
Hiper-Homocisteinemia/sangue , Resistência à Insulina/fisiologia , Receptores de Lipopolissacarídeos/metabolismo , Monócitos/metabolismo , Síndrome do Ovário Policístico/sangue , Receptores de IgG/metabolismo , Adulto , Biomarcadores/sangue , Estudos Transversais , Citocinas/metabolismo , Feminino , Humanos , Hiper-Homocisteinemia/complicações , Síndrome do Ovário Policístico/complicações , Adulto Jovem
20.
Endocrinology ; 158(5): 1181-1193, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28323956

RESUMO

It has been shown that serum homocysteine (Hcy) levels are higher in women with polycystic ovary syndrome (PCOS). However, the specific role of hyperhomocysteinemia (HHcy) in the development of PCOS has never been reported. Adipose tissue inflammation is featured by the infiltration of macrophages, which plays a critical role in the pathogenesis of glucose and insulin intolerance. In this study, C57BL/6 mice were treated with dehydroepiandrosterone (DHEA) and/or a high methionine diet to induce PCOS and HHcy mice models. We showed that DHEA induced a PCOS-like phenotypes, irregular estrous cycles, weight gain, abnormal sex hormone production, glucose and insulin resistance, and polycystic ovaries. HHcy further intensified the effects DHEA on the metabolic, endocrinal, hormonal, and morphological changes in PCOS-like mice. In addition, HHcy attenuated the DHEA-induced increase in serum estrogen levels in mice. Furthermore, HHcy may exacerbate the insulin resistance in PCOS-like mice, most likely through modulating the macrophage M1/M2 polarization pathways via the suppression of estrogen. Most important, our clinical data showed that there were increases in serum Hcy levels in patients with PCOS. These findings deepen our understanding of the pathological roles of HHcy in the development of PCOS and provide a promising target for PCOS therapy in clinical application.


Assuntos
Estrogênios/metabolismo , Hiper-Homocisteinemia/complicações , Resistência à Insulina , Macrófagos/fisiologia , Paniculite/etiologia , Síndrome do Ovário Policístico/complicações , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Animais , Polaridade Celular/imunologia , Células Cultivadas , Regulação para Baixo , Estrogênios/sangue , Feminino , Hiper-Homocisteinemia/metabolismo , Inflamação/metabolismo , Inflamação/patologia , Ativação de Macrófagos , Camundongos , Camundongos Endogâmicos C57BL , Paniculite/metabolismo , Síndrome do Ovário Policístico/imunologia , Síndrome do Ovário Policístico/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA