Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Environ Geochem Health ; 46(7): 238, 2024 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-38849627

RESUMO

Microplastics (MPs) are defined as plastic particles or fragments with a diameter of less than 5 mm. These particles have been identified as causing male reproductive toxicity, although the precise mechanism behind this association is yet to be fully understood. Recent research has found that exposure to polystyrene microplastics (PS-MPs) can disrupt spermatogenesis by impacting the integrity of the blood-testis barrier (BTB), a formidable barrier within mammalian blood tissues. The BTB safeguards germ cells from harmful substances and infiltration by immune cells. However, the disruption of the BTB leads to the entry of environmental pollutants and immune cells into the seminiferous tubules, resulting in adverse reproductive effects. Additionally, PS-MPs induce reproductive damage by generating oxidative stress, inflammation, autophagy, and alterations in the composition of intestinal flora. Despite these findings, the precise mechanism by which PS-MPs disrupt the BTB remains inconclusive, necessitating further investigation into the underlying processes. This review aims to enhance our understanding of the pernicious effects of PS-MP exposure on the BTB and explore potential mechanisms to offer novel perspectives on BTB damage caused by PS-MPs.


Assuntos
Barreira Hematotesticular , Microplásticos , Poliestirenos , Microplásticos/toxicidade , Poliestirenos/toxicidade , Masculino , Humanos , Barreira Hematotesticular/efeitos dos fármacos , Animais , Espermatogênese/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Poluentes Ambientais/toxicidade
2.
Free Radic Biol Med ; 221: 40-51, 2024 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-38759901

RESUMO

Fine particulate matter (PM2.5), a significant component of air pollution particulate matter, is inevitable and closely associated with increasing male reproductive disorder. However, the testicular targets of PM2.5 and its toxicity related molecular mechanisms are still not fully understood. In this study, the conditional knockout (cKO) mice and primary Leydig cells were used to explore the testicular targets of PM2.5 and the related underlying mechanisms. First, apparent the structure impairment of seminiferous tubules, Leydig cells vacuolization, decline of serum testosterone and sperm quality reduction were found in male wild-type (WT) and Sirt1 knockout mice after exposure to PM2.5. Enrichment analyses revealed that differentially expressed genes (DEGs) were enriched in steroid hormone biosynthesis, ferroptosis, and HIF-1 signaling pathway in the mice testes after exposure to PM2.5, which were subsequently verified by the molecular biological analyses. Notably, similar enrichment analyses results were also observed in primary Leydig cells after treatment with PM2.5. In addition, Knockdown of Sirt1 significantly increased PM2.5-induced expression and activation of HIF-1α, which was in parallel to the changes of cellular iron levels, oxidative stress indicators and the ferroptosis markers. In conclusion, this highlights that PM2.5 triggers ferroptosis via SIRT1/HIF-1α signaling pathway to inhibit testosterone synthesis in males. These findings provide a novel research support for the study that PM2.5 causes male reproductive injury.


Assuntos
Ferroptose , Subunidade alfa do Fator 1 Induzível por Hipóxia , Células Intersticiais do Testículo , Camundongos Knockout , Material Particulado , Transdução de Sinais , Sirtuína 1 , Testosterona , Animais , Masculino , Testosterona/metabolismo , Testosterona/sangue , Material Particulado/toxicidade , Material Particulado/efeitos adversos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Camundongos , Sirtuína 1/metabolismo , Sirtuína 1/genética , Transdução de Sinais/efeitos dos fármacos , Ferroptose/efeitos dos fármacos , Ferroptose/genética , Células Intersticiais do Testículo/metabolismo , Células Intersticiais do Testículo/efeitos dos fármacos , Células Intersticiais do Testículo/patologia , Testículo/metabolismo , Testículo/patologia , Testículo/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos
3.
Sci Total Environ ; 918: 170701, 2024 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-38325452

RESUMO

Epidemiological studies have found that long-term inhalation of PM2.5 is closely related to spermatogenesis disorders and infertility, but the underlying molecular mechanism is still unidentified. Testosterone, an essential reproductive hormone produced by Leydig cells, whose synthesis is disrupted by multiple environmental pollutants. In the current study, we explored the role of METTL3-m6A-SIRT1 axis-mediated abnormal autophagy in PM2.5-induced inhibition of testosterone production in in vivo and in vitro models. Our in vivo findings shown that long-term inhalation of PM2.5 decreased sperm count, increased sperm deformity rates, and altered testicular interstitial morphology accompanied by reduced testosterone in serum and testes. Further, data from the in vitro model displayed that exposure to PM2.5 caused an increase in m6A modification and METTL3 levels, followed by a decrease in testosterone levels and autophagy dysfunction in Leydig cells. The knockdown of METTL3 promotes autophagy flux and testosterone production in Leydig cells. Mechanistically, PM2.5 increased METTL3-induced m6A modification of SIRT1 mRNA in Leydig cells, bringing about abnormal autophagy. Subsequently, administration of SRT1720 (a SIRT1 activator) enhanced autophagy and further promoted testosterone biosynthesis. Collectively, our discoveries indicate that METTL3-m6A-SIRT1 axis-mediated autophagic flux contributes to PM2.5-induced inhibition of testosterone biosynthesis. This research offers a novel viewpoint on the mechanism of male reproductive injury following PM2.5 exposure.


Assuntos
Adenina/análogos & derivados , Células Intersticiais do Testículo , Testosterona , Masculino , Humanos , Sirtuína 1 , Sêmen , Material Particulado/toxicidade , Autofagia/fisiologia
4.
Chemosphere ; 345: 140428, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37858765

RESUMO

Persistent organic pollutant perfluorooctane sulfonate (PFOS) is strongly associated with male reproductive disorders, but the related mechanisms are still not fully understood. In this study, we used in vivo and in vitro models to explore the role of organic anion transporting polypeptide 3a1 (Oatp3a1) on PFOS-induced male reproductive injury. Thirty male C57BL/6 (B6) mice were orally given PFOS (0-10 mg/kg/bw) for 28 days. Body weight, organ index, sperm count, histology, and blood-testis barrier (BTB) integrity were evaluated. Primary Sertoli cells were used to describe the related molecular mechanisms of male reproductive injury caused by PFOS. Our results showed that PFOS induced a decrease in sperm count, morphological damage to testicular Sertoli cells, and disruption of BTB. In the in vitro model, exposure to PFOS significantly increased Oatp3a1 mRNA and protein levels and decreased miR-23a-3p expression in Sertoli cells, accompanied by reduced trans-epithelial electrical resistance (TEER) value. By performing the 14C-PFOS uptake experiment, we showed that 14C-PFOS uptake in HEK293-Oatp3a1 cells was apparently higher than in HEK293-MOCK cells. Meanwhile, treating Sertoli cells with Oatp3a1 siRNA significantly decreased Oatp3a1 expression and rescued PFOS-induced decreases in TEER value. As such, the present study highlights that Oatp3a1 may play an important role in the toxic effect of PFOS on Sertoli cells, advancing our understanding of molecular mechanisms for PFOS-induced male reproductive disorders.


Assuntos
Ácidos Alcanossulfônicos , Fluorocarbonos , Transportadores de Ânions Orgânicos , Masculino , Humanos , Camundongos , Animais , Células de Sertoli , Células HEK293 , Camundongos Endogâmicos C57BL , Sêmen , Ácidos Alcanossulfônicos/metabolismo , Fluorocarbonos/metabolismo , Transportadores de Ânions Orgânicos/genética , Transportadores de Ânions Orgânicos/metabolismo , Transportadores de Ânions Orgânicos/farmacologia
5.
Cancer Biomark ; 38(2): 261-272, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37599523

RESUMO

OBJECTIVE: Testicular germ cell tumors (TGCTs), containing pure seminoma and non-seminoma, occupy the most majority of testicular cancers in adolescents and young men, which has increased dramatically in recent decades. Therefore, it is important to find crucial genes for improving diagnosis and prognosis in TGCTs. However, the diagnostic and prognostic markers of TGCTs are limited. METHODS: In this study, our main objective is to explore novel potential genes that can be used as diagnostic and prognostic biomarkers in TGCTs. Our study detected 732 differentially expressed genes (DEGs) using three microarray expression profiling datasets from Gene Expression Omnibus (GEO). Multiple analysis was performed to identify the roles of DEGs, including pathway and functional enrichment analysis, protein-protein interaction (PPI) network analysis, module analysis, and survival analysis. RESULT: In total, 322 upregulated genes and 406 downregulated genes were identified as DEGs The functional and pathway enrichment analysis shows that DEGs were highly enriched in multiple biological attributes such as T cell activation, reproduction in multicellular organism, sperm flagellum, antigen processing and presentation Then, seven potential crucial genes were identified via PPI network analysis, module analysis, and survival analysis. Furthermore, 7 potential crucial genes had shown to play a key role in regulating immune cell infiltration level in patients with TGCTs. CONCLUSION: We identified seven potential crucial genes (LAPTM5, NCF2, PECAM1, CD14, COL4A2, ANPEP and RGS1), which may be molecular markers in improving the way of diagnosis and prognosis in TGCTs.

6.
Rev Environ Health ; 2023 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-37651650

RESUMO

Exposure to PM2.5 is the most significant air pollutant for health risk. The testosterone level in male is vulnerable to environmental toxicants. In the past, researchers focused more attention on the impacts of PM2.5 on respiratory system, cardiovascular system, and nervous system, and few researchers focused attention on the reproductive system. Recent studies have reported that PM2.5 involved in male testosterone biosynthesis disruption, which is closely associated with male reproductive health. However, the underlying mechanisms by which PM2.5 causes testosterone biosynthesis disruption are still not clear. To better understand its potential mechanisms, we based on the existing scientific publications to critically and comprehensively reviewed the role and potential mechanisms of PM2.5 that are participated in testosterone biosynthesis in male. In this review, we summarized the potential mechanisms of PM2.5 triggering the change of testosterone level in male, which involve in oxidative stress, inflammatory response, ferroptosis, pyroptosis, autophagy and mitophagy, microRNAs (miRNAs), endoplasmic reticulum (ER) stress, and N6-methyladenosine (m6A) modification. It will provide new suggestions and ideas for prevention and treatment of testosterone biosynthesis disruption caused by PM2.5 for future research.

7.
Environ Pollut ; 301: 118960, 2022 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-35150797

RESUMO

Perfluorooctane sulfonate (PFOS) is associated with male reproductive disorder, but the related mechanisms are still unclear. In this study, we used in vivo and in vitro models to explore the role of Sertoli cell-derived exosomes (SC-Exo)/miR-9-3p/StAR signaling pathway on PFOS-induced suppression of testosterone biosynthesis. Forty male ICR mice were orally administrated PFOS (0.5-10 mg/kg/bw) for 4 weeks. Bodyweight, organ index, sperm count, reproductive hormones were evaluated. Primary Sertoli cells and Leydig cells were used to delineate the molecular mechanisms that mediate the effects of PFOS on testosterone biosynthesis. Our results demonstrated that PFOS dose-dependently induced a decrease in sperm count, low levels of testosterone, and damage in testicular interstitium morphology. In vitro models, PFOS significantly increased miR-9-3p levels in Sertoli cells and SC-Exo, accompanied by a decrease in testosterone secretion and StAR expression in Leydig cells when Leydig cells were exposed to SC-Exo. Meanwhile, inhibition of SC-Exo or miR-9-3p by their inhibitors significantly rescued PFOS-induced decreases in testosterone secretion and the mRNA and protein expression of the StAR gene in Leydig cells. In summary, the present study highlights the role of the SC-Exo/miR-9-3p/StAR signaling pathway in PFOS-induced suppression of testosterone biosynthesis, advancing our understanding of molecular mechanisms for PFOS-induced male reproductive disorders.


Assuntos
Células Intersticiais do Testículo , MicroRNAs , Ácidos Alcanossulfônicos , Animais , Fluorocarbonos , Masculino , Camundongos , Camundongos Endogâmicos ICR , MicroRNAs/genética , MicroRNAs/metabolismo , Células de Sertoli , Testosterona/metabolismo
8.
Rev Environ Health ; 2022 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-36863426

RESUMO

With the rapid expansion of industrial scale, an increasing number of fine particulate matter (PM2.5) has bringing health concerns. Although exposure to PM2.5 has been clearly associated with male reproductive toxicity, the exact mechanisms are still unclear. Recent studies demonstrated that exposure to PM2.5 can disturb spermatogenesis through destroying the blood-testis barrier (BTB), consisting of different junction types, containing tight junctions (TJs), gap junctions (GJs), ectoplasmic specialization (ES) and desmosomes. The BTB is one of the tightest blood-tissue barriers among mammals, which isolating germ cells from hazardous substances and immune cell infiltration during spermatogenesis. Therefore, once the BTB is destroyed, hazardous substances and immune cells will enter seminiferous tubule and cause adversely reproductive effects. In addition, PM2.5 also has shown to cause cells and tissues injury via inducing autophagy, inflammation, sex hormones disorder, and oxidative stress. However, the exact mechanisms of the disruption of the BTB, induced by PM2.5, are still unclear. It is suggested that more research is required to identify the potential mechanisms. In this review, we aim to understand the adverse effects on the BTB after exposure to PM2.5 and explore its potential mechanisms, which provides novel insight into accounting for PM2.5-induced BTB injury.

9.
Environ Health Perspect ; 129(9): 97006, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34495743

RESUMO

BACKGROUND: Epidemiological studies have shown that exposure to ambient fine particulate matter with aerodynamic diameter less than or equal to 2.5 µm (PM2.5) correlates with a decrease in sperm count, but the biological mechanism remains elusive. OBJECTIVES: This study tested whether hypothalamic inflammation, an emerging pathophysiological mediator, mediates the development of lower epididymal sperm count due to PM2.5 exposure. METHODS: Inhibitor κB kinase 2 (IKK2) was conditionally knocked out either in all neurons or subtypes of hypothalamic neurons of mice. Effects of concentrated ambient PM2.5 (CAP) exposure on hypothalamic inflammation, the hypothalamic-pituitary-gonadal (HPG) axis, and epididymal sperm count of these mouse models were then assessed. Furthermore, to test whether hypothalamic inflammation is sufficient to decrease sperm production, we overexpressed constitutively active IKK2 (IKK2ca) either in all neurons or subtypes of hypothalamic neurons and assessed hypothalamic inflammation, the HPG axis, and sperm production of these overexpression mouse models. RESULTS: CAP-exposed wild-type control mice vs. filtered air (FA)-exposed wild-type control mice had a higher expression of hypothalamic inflammatory markers, lower functional indexes of the HPG axis, and a lower epididymal sperm count. In contrast, all these measurements for CAP- vs. FA-exposed mice deficient of IKK2 in all neurons were comparable. We also found that overexpression of IKK2ca in either all neurons or pro-opiomelanocortin (POMC) neurons only, but not in Agouti-related protein (AgRP) neurons only, resulted in lower functional indexes of the HPG axis and a lower epididymal sperm count. Moreover, we showed that CAP- vs. FA-exposed mice deficient of IKK2 in POMC neurons had a comparable expression of hypothalamic inflammatory markers, comparable functional indexes of the HPG axis, and a comparable epididymal sperm count. DISCUSSION: This mouse model study shows a causal role of IKK2 of POMC neurons in the development of lower epididymal sperm count due to PM2.5 exposure, providing a mechanistic insight into this emerging pathogenesis. https://doi.org/10.1289/EHP8868.


Assuntos
Material Particulado , Pró-Opiomelanocortina , Animais , Hipotálamo , Masculino , Camundongos , Neurônios , Material Particulado/metabolismo , Material Particulado/toxicidade , Pró-Opiomelanocortina/metabolismo , Pró-Opiomelanocortina/farmacologia , Espermatogênese
10.
Toxicology ; 460: 152871, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34303733

RESUMO

Perfluorooctane sulfonate (PFOS) is a widespread persistent organic pollutant. Both epidemiological survey and our previous in vivo study have revealed the associations between PFOS exposure and spermatogenesis disorder, while the underlying mechanisms are far from clear. In the present study, GC-2 cells, a mouse spermatocyte-derived cell line, was used to investigate the toxic effects of PFOS and its hypothetical mechanism of action. GC-2 cells were treated with PFOS (0, 50, 100 and 150 µM) for 24 h or 48 h. Results demonstrated that PFOS dose-dependently inhibited cell viability, induced G0/G1 cell cycle arrest and triggered apoptosis, which might be partly explained by the decrease in cyclin D1, PCNA and Bcl-2 protein expression; increase in Bax protein expression; and activation of caspase-9, -3. In addition, PFOS did not directly transactivate or repress estrogen receptors (ERs) in gene reporter assays, whereas the protein levels of both ERα and ERß were significantly altered and the downstream ERK1/2 phosphorylation was inhibited by PFOS. Furthermore, pretreatment with specific ERα agonist PPT (1 µM) significantly attenuated the above PFOS-induced effects while specific ERß agonist DPN (1 µM) accelerated them. These results suggest that PFOS may induce growth inhibition and apoptosis via non-genomic estrogen receptor/ERK1/2 signaling pathway in GC-2 cells, which provides a novel insight regarding the potential role of ERs in mediating PFOS-triggered spermatocyte toxicity.


Assuntos
Ácidos Alcanossulfônicos/toxicidade , Apoptose/efeitos dos fármacos , Fluorocarbonos/toxicidade , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Receptores de Estrogênio/antagonistas & inibidores , Espermatócitos/efeitos dos fármacos , Animais , Apoptose/fisiologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Chlorocebus aethiops , Relação Dose-Resposta a Droga , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Camundongos , Receptores de Estrogênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Espermatócitos/metabolismo
11.
Environ Sci Technol ; 55(9): 6097-6106, 2021 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-33825453

RESUMO

Considerable studies show that maternal exposure to ambient fine particulate matter (PM2.5) programs offspring's susceptibility to obesity. However, few studies have investigated the effect of paternal PM2.5 exposure on offspring's energy homeostasis. This study thus tested whether paternal PM2.5 exposure programs offspring's energy homeostasis. Male C57Bl/6J mice were exposed to filtered air or concentrated ambient PM2.5 (CAP) for 12 weeks and then mated with normal female C57Bl/6J mice. The offspring were assessed for growth trajectories, food intakes, and body compositions, and the sperm miRNAs of those sires were profiled by microarray. Zygotic injection was used to test whether the miRNA identified by the microarray mediates the impact of paternal PM2.5 exposure on offspring's energy homeostasis. Paternal CAP exposure resulted in significant hypophagia and weight loss in male, but not female, offspring. The weight loss of male offspring was accompanied by decreases in the liver and kidney masses and paradoxically an increase in the adipose mass. Without further exposure to CAP, this programming was three-generationally transmitted along the paternal line. The sperm miRNA profiling revealed that mmu-mir6909-5p was the sole differentially expressed sperm miRNA due to PM2.5 exposure, and zygotic injection of mmu-mir6909-5p mimicked all the effects of paternal PM2.5 exposure on offspring's energy homeostasis. Paternal PM2.5 exposure programs offspring's energy homeostasis through increasing paternal sperm mmu-mir6909-5p.


Assuntos
Exposição Materna , Exposição Paterna , Animais , Feminino , Homeostase , Humanos , Masculino , Camundongos , Obesidade , Material Particulado/efeitos adversos
12.
Toxicology ; 449: 152663, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33359577

RESUMO

Perfluorooctane sulfonate (PFOS), a stable end-product of perfluorinated compounds (PFCs), is associated with male reproductive disorders, but its underlying mechanisms are still unclear. We used in vivo and in vitro models to investigate the effects of PFOS on testosterone biosynthesis and related mechanisms. First, male ICR mice were orally administered PFOS (0-10 mg/kg/bw) for 4 weeks. Bodyweight, sperm count, reproductive hormones, mRNA expression of the genes related to testosterone biosynthesis, and the protein expression of protein kinase A (PKA), p38 mitogen-activated protein kinase (MAPK), cAMP-response element binding protein (CREB), CREB regulated transcription coactivator 2 (CRTC2) and steroidogenic acute regulatory protein (StAR) were evaluated. Furthermore, mouse primary Leydig cells were used to delineate the molecular mechanisms that mediate the effects of PFOS on testosterone biosynthesis. Our results demonstrated that PFOS dose-dependently decreased sperm count, testosterone level, CRTC2/StAR expression, and damaged testicular interstitium morphology, paralleled by increase in phosphorylated PKA, CREB and p38 in testes. Additionally, similar to the in vivo results, PFOS significantly decreased testosterone secretion, CRTC2/StAR expression, interaction between CREB and CRTC2 and binding of CREB/CRTC2 to StAR promoter region, paralleled by increase in phosphorylated-p38, PKA, and CREB expression. Meanwhile, inhibition of p38 by SB203580, or inhibition of PKA by H89 can significantly alleviate the above PFOS-induced effects. As such, the present study highlights a role of the CREB/CRTC2/StAR signaling pathway in PFOS-induced suppression of testosterone biosynthesis, advancing our understanding of molecular mechanisms for PFOS-induced male reproductive disorders.


Assuntos
Ácidos Alcanossulfônicos/toxicidade , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Fluorocarbonos/toxicidade , Células Intersticiais do Testículo/metabolismo , Fosfoproteínas/metabolismo , Testosterona/biossíntese , Fatores de Transcrição/metabolismo , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/antagonistas & inibidores , Relação Dose-Resposta a Droga , Células Intersticiais do Testículo/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Fosfoproteínas/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Testosterona/antagonistas & inibidores , Fatores de Transcrição/antagonistas & inibidores
13.
Ecotoxicol Environ Saf ; 208: 111464, 2021 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-33075589

RESUMO

Ambient fine particulate matter (PM2.5) exposure correlates with adverse cardiometabolic effects. The underlying mechanisms have not yet been fully understood. Hypothalamic-pituitary-adrenal (HPA) axis, as the central stress response system, regulates cardiometabolic homeostasis and is implicated in the progression of various adverse health effects caused by inhalational airborne pollutant exposure. In this study, we investigated whether ambient PM2.5 exposure activates HPA axis and its effect mediating PM2.5-induced pulmonary inflammation. C57Bl/6 J mice were intratracheally instilled with different concentrations of diesel exhaust PM2.5 (DEP), and plasma was harvested at different times. Assessments of plasma stress hormones revealed that DEP instillation dose- and time-dependently increased mouse circulating corticosterone and adrenocorticotropic hormone (ACTH) levels, strongly supporting that DEP instillation activates HPA axis. To determine which components of DEP activate HPA axis, C57Bl/6J mice were intratracheally instilled with water-soluble and -insoluble fractions of DEP. Plasma analyses showed that water-insoluble but not -soluble fraction of DEP increased circulating corticosterone and ACTH levels. Consistently, concentrated ambient PM2.5 (CAP) exposure significantly increased mouse urine and hair corticosterone levels, corroborating the activation of HPA axis by ambient PM2.5. Furthermore, deletion of stress hormones by total bilateral adrenalectomy alleviated PM2.5-induced pulmonary inflammation, providing insights into the contribution of central neurohormonal mechanisms in modulating adverse health effects caused by exposure to PM2.5.


Assuntos
Poluentes Atmosféricos/toxicidade , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Material Particulado/toxicidade , Pneumonia/induzido quimicamente , Animais , Corticosterona/sangue , Exposição por Inalação/análise , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Emissões de Veículos/análise
14.
Environ Health Perspect ; 127(5): 57009, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31095431

RESUMO

BACKGROUND: Pulmonary inflammation is believed to be central to the pathogenesis due to exposure to fine particulate matter with aerodynamic diameter [Formula: see text] ([Formula: see text]). This central role, however, has not yet been systemically examined. OBJECTIVE: In the present study, we exploited a lung epithelial cell-specific inhibitor [Formula: see text] kinase 2 (IKK2) knockout mouse model to determine the role of pulmonary inflammation in the pathophysiology due to exposure to diesel exhaust particulate matter (DEP). METHODS: [Formula: see text] (lung epithelial cell-specific IKK2 knockout, KO) and [Formula: see text] (wild-type, tgWT) mice were intratracheally instilled with either vehicle or DEP for 4 months, and their inflammatory response and glucose homeostasis were then assessed. RESULTS: In comparison with tgWT mice, lung epithelial cell-specific IKK2-deficient mice had fewer DEP exposure-induced bronchoalveolar lavage fluid immune cells and proinflammatory cytokines as well as fewer DEP exposure-induced circulating proinflammatory cytokines. Glucose and insulin tolerance tests revealed that lung epithelial cell-specific IKK2 deficiency resulted in markedly less DEP exposure-induced insulin resistance and greater glucose tolerance. Akt phosphorylation analyses of insulin-responsive tissues showed that DEP exposure primarily targeted hepatic insulin sensitivity. Lung epithelial cell-specific IKK2-deficient mice had significantly lower hepatic insulin resistance than tgWT mice had. Furthermore, this difference in insulin resistance was accompanied by consistent differences in hepatic insulin receptor substrate 1 serine phosphorylation and inflammatory marker expression. DISCUSSION: Our findings suggest that in a tissue-specific knockout mouse model, an IKK2-dependent pulmonary inflammatory response was essential for the development of abnormal glucose homeostasis due to exposure to DEP. https://doi.org/10.1289/EHP4591.


Assuntos
Poluentes Atmosféricos/toxicidade , Glucose/fisiologia , Homeostase , Quinase I-kappa B/deficiência , Material Particulado/toxicidade , Emissões de Veículos/toxicidade , Animais , Modelos Animais de Doenças , Células Epiteliais/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Masculino , Camundongos , Camundongos Transgênicos
15.
FASEB J ; 33(2): 2782-2795, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30307764

RESUMO

Fenvalerate (FEN), a mainstream pyrethroid pesticide, was initially recommended as a low-toxicity agent for controlling agricultural and domestic pests. Despite the widespread use of FEN worldwide, little data are available on FEN-induced hepatic lesions and molecular mechanisms. In the present study, we first performed an occupational cross-sectional study on FEN factory workers and found that the levels of serum alanine aminotransferase (ALT) and total antioxidant capacity increased, whereas malondialdehyde decreased in laborers in the working areas where the levels of airborne FEN were much higher compared with the office area. The results were then confirmed by animal experiments that abnormal hepatic histology, increased ALT level, and compromised hepatic oxidative capability were observed in rats exposed to a high concentration of FEN. Furthermore, the bioinformatics analysis of gene microarray in rat liver tissue showed that FEN significantly changed the expressions of genes related to the regulation of intracellular calcium ion homeostasis and the calcium signal pathway. Finally, the functional experiments in Buffalo rat liver (BRL) cells demonstrated that FEN first activated ERK MAPK, followed by IKK and NF-κB, which triggered the transcription of genes responsible for accelerating an overload of intracellular calcium ions, prompted reactive oxygen species generation in the mitochondria, and finally, induced hepatic cellular apoptosis. The calcium signaling pathway and in particular, an overload of intracellular calcium play a critical role in this pathophysiological process via the ERK/IKK/NF-κB pathway. Our study furthers the understanding of the mechanism of FEN-induced hepatic injuries and may have implications in the prevention and control of liver diseases induced by environmental pesticides.-Qiu, L.-L., Wang, C., Yao, S., Li, N., Hu, Y., Yu, Y., Xia, R., Zhu, J., Ji, M., Zhang, Z., Wang S.-L. Fenvalerate induces oxidative hepatic lesions through an overload of intracellular calcium triggered by the ERK/IKK/NF-κB pathway.


Assuntos
Cálcio/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Nitrilas/efeitos adversos , Exposição Ocupacional/efeitos adversos , Piretrinas/efeitos adversos , Transdução de Sinais/efeitos dos fármacos , Animais , Apoptose , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Estudos Transversais , Feminino , Perfilação da Expressão Gênica , Humanos , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , Inseticidas/efeitos adversos , Masculino , NF-kappa B/genética , NF-kappa B/metabolismo , Estresse Oxidativo , Ratos , Ratos Sprague-Dawley
16.
J Mol Endocrinol ; 61(4): 153-161, 2018 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-30021757

RESUMO

Numerous studies have implicated tumor necrosis factor α (TNFα) in the pathogenesis of type 2 diabetes. However, the role of its primary receptor, TNF receptor 1 (TNFR1), in homeostatic regulation of glucose metabolism is still controversial. In addition to TNFα, lymphotoxin α (LTα) binds to and activates TNFR1. Thus, TNFα and LTα together are known as TNF. To delineate the role of TNF signaling in glucose homeostasis, the present study ascertained how TNF signaling deficiency affects major regulatory components of glucose homeostasis. To this end, normal diet-fed male TNFR1 deficient mice (TNFR1-/-), TNFα/LTα/LTß triple deficient mice (TNF/LT∆3), and their littermate controls were subjected to intraperitoneal glucose tolerance test, insulin tolerance test, and oral glucose tolerance test. The present results showed that TNFR1-/- and TNF/LT∆3 mice versus their controls had comparable body weight, tolerance to intraperitoneal glucose, and sensitivity to insulin. However, their tolerance to oral glucose was significantly increased. Additionally, glucose-induced insulin secretion assessments revealed that TNFR1 or TNF/LT deficiency significantly increased oral but not intraperitoneal glucose-induced insulin secretion. Consistently, qPCR and immunohistochemistry analyses showed that TNFR1-/- and TNF/LT∆3 mice versus their controls had significantly increased ileal expression of glucagon-like peptide-1 (GLP-1), one of the primary incretins. Their oral glucose-induced secretion of GLP-1 was also significantly increased. These data collectively suggest that physiological TNF signaling regulates glucose metabolism primarily through effects on GLP-1 expression and secretion and subsequently insulin secretion.


Assuntos
Peptídeo 1 Semelhante ao Glucagon/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Peptídeo 1 Semelhante ao Glucagon/genética , Insulina/genética , Insulina/metabolismo , Masculino , Camundongos , Camundongos Knockout , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Fator de Necrose Tumoral alfa/genética
17.
Environ Health Perspect ; 126(2): 027003, 2018 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-29410383

RESUMO

BACKGROUND: Exposure to ambient fine particulate matter (PM2.5) is associated with cardiovascular mortality, but underlying pathophysiologic mechanisms are not fully understood. Hypothalamic inflammation, characterized by the activation of Inhibitor kappaB kinase 2/Nuclear factor kappaB (IKK2/NF-κB) signaling pathway, may play an important role in the pathogenesis of cardiovascular diseases. We recently demonstrated that hypothalamic inflammation is increased in mice exposed to concentrated ambient PM2.5 (CAP). OBJECTIVES: In the present study, we used a neuron-specific IKK2 knockout mouse model to examine the role of neural IKK2 expression and hypothalamic inflammation in the pathophysiologic effects of PM2.5. METHODS: We assessed inflammatory and vascular responses in Nestin-creIKK2flox/flox (IKK2Neu-KO) and littermate Nestin-creIKK2flox/+ (control) mice after 4 mo of exposure to filtered air (FA) or CAP. RESULTS: CAP exposure was associated with significantly higher tumor necrosis factor-α (TNFα) and interleukin (IL)-6 mRNA in the hypothalamus of control mice, but not IKK2Neu-KO mice. In addition, CAP exposure-induced increases in bronchoalveolar lavage fluid (BALF) leukocytes, pulmonary macrophage infiltration and IL-6 expression, plasma TNFα and IL-1ß levels, adipose macrophage infiltration and IL-1ß expression, and endothelial dysfunction were reduced or absent in IKK2Neu-KO mice compared with controls. CONCLUSIONS: Our findings support a role of neural IKK2 in CAP exposure-induced local and systemic pro-inflammatory cytokine expression, pulmonary and adipose inflammation, and endothelial dysfunction, thus providing insight into pathophysiologic mechanisms that may mediate effects of PM2.5 exposure. https://doi.org/10.1289/EHP2311.


Assuntos
Endotélio Vascular/efeitos dos fármacos , Quinase I-kappa B/genética , Inflamação/induzido quimicamente , Material Particulado/toxicidade , Tecido Adiposo/patologia , Poluentes Atmosféricos/toxicidade , Animais , Líquido da Lavagem Broncoalveolar , Citocinas/metabolismo , Modelos Animais de Doenças , Endotélio Vascular/fisiopatologia , Exposição Ambiental/efeitos adversos , Hipotálamo/efeitos dos fármacos , Hipotálamo/fisiopatologia , Inflamação/fisiopatologia , Pulmão/efeitos dos fármacos , Pulmão/patologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tamanho da Partícula
18.
Am J Physiol Endocrinol Metab ; 315(1): E72-E80, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29351483

RESUMO

Environmental stressors that encounter in early-life and cause abnormal fetal and/or neonatal development may increase susceptibility to non-communicable diseases such as diabetes. Maternal exposure to ambient fine particulate matter (PM2.5) is associated with various fetal abnormalities, suggesting that it may program offspring's susceptibility to diabetes. In the present study, we therefore examined whether maternal exposure to diesel exhaust PM2.5 (DEP), one of the major sources of ambient PM2.5 in urban areas, programs adult offspring's glucose metabolism. Female C57Bl/6J mice were intratracheally instilled with DEP or vehicle throughout a 7-wk preconceptional period, gestation, and lactation, and the glucose homeostasis of their adult male offspring was assessed. Intraperitoneal glucose tolerance test (IPGTT) revealed that the maternal exposure to DEP significantly impaired adult male offspring's glucose tolerance. Unexpectedly, it did not influence their insulin sensitivity, whereas it significantly decreased their glucose-induced insulin secretion (GIIS). This deficit in insulin secretion was corroborated by their significant decrease in arginine-induced insulin secretion. Histological analysis demonstrated that the deficit in insulin secretion was accompanied by the decrease in pancreatic islet and ß cell sizes. To differentiate the effects of maternal exposure to DEP before birth and during lactation, some offspring were cross-fostered once born. We did not observe any significant effect of cross-fostering on the glucose homeostasis of adult male offspring and the function and morphology of their ß cells. Prenatal exposure to DEP programs the morphology and function of ß cells and thus homeostatic regulation of glucose metabolism in adult male offspring.


Assuntos
Poluentes Atmosféricos/toxicidade , Células Secretoras de Insulina/efeitos dos fármacos , Material Particulado/toxicidade , Emissões de Veículos/toxicidade , Animais , Tamanho Celular/efeitos dos fármacos , Feminino , Glucose/metabolismo , Teste de Tolerância a Glucose , Resistência à Insulina , Secreção de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Masculino , Exposição Materna , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo
19.
Toxicol Sci ; 162(1): 318-326, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29165613

RESUMO

Epidemiological studies link ambient fine particulate matter (PM2.5) pollution to abnormalities in the male reproductive system. However, few toxicological studies have investigated this potentially important adverse effect of PM2.5 pollution. Therefore, in the present study, we analyzed the effects of PM2.5 exposure on spermatogenesis and hypothalamic-pituitary-gonadal (HPG) axis in a murine model. Fourteen male C57BL/6J mice were subjected to a 4-month exposure to filtered air or concentrated ambient PM2.5 (CAP). Their sperm count, testicular histology, spermatogenic parameters, and the major components of HPG axis were assessed. Exposure to CAP significantly reduced sperm count in the epididymis. This was accompanied by Sertoli cell vacuolization, immature germ cell dislocation, and decreases in pachytene spermatocytes and round spermatids of stage VII seminiferous tubules, suggesting a marked impairment of spermatogenesis in these mice. This impairment of spermatogenesis appeared to be attributable to a suppression of HPG axis subsequent to CAP exposure-induced hypothalamic inflammation, as exposure to CAP significantly increased TNFα and IL1b mRNA levels and meanwhile decreased gonadotropin-releasing hormone mRNA expression in the hypothalamus. Moreover, CAP exposure significantly reduced circulating testosterone and follicle-stimulating hormone, testicular testosterone and mRNA expression of follicle-stimulating hormone target gene SHBG and luteinizing hormone target genes P450scc, 17ßHSD, and StAR. The present data demonstrate that exposure to ambient PM2.5 impairs spermatogenesis in murine model, raising the concern over effects of ambient PM2.5 pollution on the male reproductive function.


Assuntos
Poluentes Atmosféricos/toxicidade , Genitália Masculina/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Material Particulado/toxicidade , Espermatogênese/efeitos dos fármacos , Animais , Citocinas/genética , Relação Dose-Resposta a Droga , Hormônio Foliculoestimulante/sangue , Hormônio Foliculoestimulante/genética , Genitália Masculina/metabolismo , Genitália Masculina/patologia , Hormônio Liberador de Gonadotropina/genética , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipotálamo-Hipofisário/fisiopatologia , Exposição por Inalação/efeitos adversos , Exposição por Inalação/análise , Masculino , Camundongos Endogâmicos C57BL , Tamanho do Órgão , Contagem de Espermatozoides , Testosterona/sangue , Testosterona/genética , Fatores de Tempo
20.
Toxicol Sci ; 160(1): 4-14, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-29036520

RESUMO

The lung is constantly exposed to ambient pollutants such as ambient fine particulate matter (PM2.5), making it one of the most frequent locations of inflammation in the body. Given the establishment of crucial role of inflammation in the pathogenesis of cardiometabolic diseases, pulmonary inflammation is thus widely believed to be an important risk factor for cardiometabolic diseases. However, the causality between them has not yet been well established. To determine if pulmonary inflammation is sufficient to cause adverse cardiometabolic effects, SFTPC-rtTA+/-tetO-cre+/-pROSA-inhibitor κB kinase 2(IKK2)ca+/- (LungIKK2ca) and littermate SFTPC-rtTA+/-tetO-cre-/-pROSA-IKK2ca+/- wildtype (WT) mice were fed with doxycycline diet to induce constitutively active Ikk2 (Ikk2ca) overexpression in the lung and their pulmonary, systemic, adipose, and hypothalamic inflammations, vascular function, and glucose homeostasis were assessed. Feeding with doxycycline diet resulted in IKK2ca overexpression in the lungs of LungIKK2ca but not WT mice. This induction of IKK2ca was accompanied by marked pulmonary inflammation as evidenced by significant increases in bronchoalveolar lavage fluid leukocytes, pulmonary macrophage infiltration, and pulmonary mRNA expression of tumor necrosis factor α (Tnfα) and interleukin-6 (Il-6). This pulmonary inflammation due to lung-specific overexpression of IKK2ca was sufficient to increase circulating TNFα and IL-6 levels, adipose expression of Tnfα and Il-6 mRNA, aortic endothelial dysfunction, and systemic insulin resistance. Unexpectedly, no significant alteration in hypothalamic expression of Tnfα and Il-6 mRNA and glucose intolerance were observed in these mice. Pulmonary inflammation is sufficient to induce systemic inflammation, endothelial dysfunction, and insulin resistance, but not hypothalamic inflammation and glucose intolerance.


Assuntos
Tecido Adiposo/enzimologia , Encefalite/enzimologia , Intolerância à Glucose/enzimologia , Hipotálamo/enzimologia , Quinase I-kappa B/metabolismo , Pulmão/enzimologia , Paniculite/enzimologia , Pneumonia/enzimologia , Animais , Aorta/enzimologia , Aorta/fisiopatologia , Glicemia/metabolismo , Encefalite/genética , Endotélio Vascular/enzimologia , Endotélio Vascular/fisiopatologia , Ativação Enzimática , Predisposição Genética para Doença , Intolerância à Glucose/sangue , Intolerância à Glucose/genética , Quinase I-kappa B/genética , Resistência à Insulina , Interleucina-6/genética , Interleucina-6/metabolismo , Pulmão/patologia , Camundongos Transgênicos , Paniculite/genética , Fenótipo , Pneumonia/genética , Pneumonia/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...