Cigarette smoke exposure

  • 文章类型: Journal Article
    背景:吸烟可能会影响特发性肺纤维化(IPF)的进展,吸烟强度与IPF呈剂量反应相关。
    方法:回顾性分析我院2014年至2018年确诊的IPF患者,并进行随访以确认生存状况和持续时间。并确定吸烟对IPF预后的影响。我们从生物信息学数据库中检索了IPF的信息,以鉴定吸烟者中lncRNAs和蛋白质的差异表达。因此,我们通过细胞实验探索并验证了香烟烟雾暴露(CSE)调节LINC00665/XBP-1参与肺纤维化的机制.我们通过细胞和分子实验阐明了LINC00665和XBP-1之间的机制。并通过使用博来霉素(BLM)诱导的肺纤维化模型验证了沉默LINC00665对肺纤维化的抑制作用。
    结果:我们发现,与不吸烟者相比,患有IPF的吸烟者预后较差。LINC00665和XBP-1在IPF肺组织和吸烟者肺组织中的表达均显著上调,此外,LINC00665在吸烟者IPF肺组织中高于吸烟者健康人。暴露于CSE可以上调LINC00665/XBP-1在肺成纤维细胞到肌成纤维细胞的转变。细胞和分子实验表明LINC00665通过靶向miR-214-3p调节XBP-1的表达。LINC00665表达式,在BLM诱导的小鼠肺纤维化组织中显著上调,和LINC00665敲低抑制BLM诱导的肺纤维化中的纤维化。
    结论:我们的研究发现LINC00665的高表达与吸烟者IPF的发病机制有关,CSE可能正调控LINC00665/XBP-1参与肺成纤维细胞向肌成纤维细胞的转化。这些发现有助于阐明吸烟者IPF的发病机制,并可能有助于开发新的IPF治疗靶向药物。
    BACKGROUND: Cigarette smoking may impact the progression of idiopathic pulmonary fibrosis (IPF), and the intensity of smoking presents a dose-response association with IPF.
    METHODS: We retrospectively analyzed IPF patients diagnosed in our hospital from 2014 to 2018 and performed follow-up to confirm survival status and duration, and determine the effect of smoking on the prognosis of IPF. We retrieved information on IPF from a bioinformatics database to identify the differential expression of lncRNAs and proteins in smokers. Therefore, we explored and verified the mechanism by which cigarette smoke exposure (CSE) regulates LINC00665/XBP-1 involvement in pulmonary fibrosis through cell experiments. We clarified the mechanism between LINC00665 and XBP-1 through cellular and molecular experiments, and verified the inhibitory effect of silencing LINC00665 on pulmonary fibrosis by using a bleomycin (BLM)-induced pulmonary fibrosis model.
    RESULTS: We found that smokers with IPF had a poor prognosis compared with non-smokers. Both the expression of LINC00665 and XBP-1 in IPF lung tissue and smoker lung tissue were significantly upregulated, moreover, LINC00665 was higher in smoker IPF lung tissue than in smoker healthy people. Exposure to CSE could upregulate LINC00665/XBP-1 in lung fibroblast-to-myofibroblast transition. Cellular and molecular experiments showed that LINC00665 regulates the expression of XBP-1 by targeting miR-214-3p. LINC00665 expression, was significantly upregulated in BLM-induced mouse lung fibrosis tissues, and LINC00665 knockdown inhibited fibrogenesis in BLM-induced lung fibrosis.
    CONCLUSIONS: Our study found that the high expression of LINC00665 is involved in the pathogenesis of smoker IPF and that CSE may positively regulate LINC00665/XBP-1 to participate in lung fibroblast-to-myofibroblast transition. These findings help elucidate the pathogenesis of smoker IPF and may contribute to the development of new targeted drugs for IPF therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    慢性阻塞性肺疾病(COPD)是一种使人衰弱的肺部疾病,没有有效的治疗方法可以降低死亡率或减缓疾病进展。COPD是全球死亡的第三大原因,其特征在于由慢性支气管炎和肺泡损伤/肺气肿引起的气流受限。慢性香烟烟雾暴露(CS)会损害气道和肺泡上皮,并且仍然是COPD发病的主要危险因素。我们发现caveolin-1,肿瘤抑制蛋白,p53和纤溶酶原激活物抑制剂-1(PAI-1)的表达,来自COPD患者或CS诱导的肺损伤(CS-LI)的野生型小鼠的气道上皮细胞(AECs)以及II型肺泡上皮细胞(AT2)中p53的下游靶标之一显著增加.此外,p53和PAI-1缺陷型小鼠抵抗CS-LI。此外,AECs的治疗,来自COPD患者的AT2细胞或肺组织切片,或具有具有七个氨基酸的caveolin-1支架结构域肽(CSP7)的CS-LI小鼠可减少AECs中的粘液分泌过多并改善AT2细胞活力。值得注意的是,通过增加的caveolin-1和p53诱导PAI-1表达有助于AECs中的粘液细胞化生和粘液分泌过多,并降低AT2的活力,由于衰老和凋亡增加,已被CSP7废除。此外,用CSP7通过腹膜内注射或通过气道雾化治疗具有CS-LI的野生型小鼠,肺泡损伤,显著改善肺功能。本研究验证了CSP7治疗CS-LI和COPD的潜在治疗作用。
    Chronic obstructive pulmonary disease (COPD) is a debilitating lung disease with no effective treatment that can reduce mortality or slow the disease progression. COPD is the third leading cause of global death and is characterized by airflow limitations due to chronic bronchitis and alveolar damage/emphysema. Chronic cigarette smoke (CS) exposure damages airway and alveolar epithelium and remains a major risk factor for the pathogenesis of COPD. We found that the expression of caveolin-1, a tumor suppressor protein; p53; and plasminogen activator inhibitor-1 (PAI-1), one of the downstream targets of p53, was markedly increased in airway epithelial cells (AECs) as well as in type II alveolar epithelial (AT2) cells from the lungs of patients with COPD or wild-type mice with CS-induced lung injury (CS-LI). Moreover, p53- and PAI-1-deficient mice resisted CS-LI. Furthermore, treatment of AECs, AT2 cells, or lung tissue slices from patients with COPD or mice with CS-LI with a seven amino acid caveolin-1 scaffolding domain peptide (CSP7) reduced mucus hypersecretion in AECs and improved AT2 cell viability. Notably, induction of PAI-1 expression via increased caveolin-1 and p53 contributed to mucous cell metaplasia and mucus hypersecretion in AECs, and reduced AT2 viability, due to increased senescence and apoptosis, which was abrogated by CSP7. In addition, treatment of wild-type mice having CS-LI with CSP7 by intraperitoneal injection or nebulization via airways attenuated mucus hypersecretion, alveolar injury, and significantly improved lung function. This study validates the potential therapeutic role of CSP7 for treating CS-LI and COPD. NEW & NOTEWORTHY Chronic cigarette smoke (CS) exposure remains a major risk factor for the pathogenesis of COPD, a debilitating disease with no effective treatment. Increased caveolin-1 mediated induction of p53 and downstream plasminogen activator inhibitor-1 (PAI-1) expression contributes to CS-induced airway mucus hypersecretion and alveolar wall damage. This is reversed by caveolin-1 scaffolding domain peptide (CSP7) in preclinical models, suggesting the therapeutic potential of CSP7 for treating CS-induced lung injury (CS-LI) and COPD.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    慢性阻塞性肺疾病(COPD)通常与肠道合并症有关。在这项研究中,研究了香烟烟雾(CS)和脂多糖(LPS)诱导的COPD模型中肠道稳态和免疫力的变化。小鼠暴露于香烟烟雾或空气中72天,除了第42、52和62天,小鼠通过气管内滴注用盐水或LPS处理。香烟烟雾暴露增加了气道炎症细胞数量,粘液产生,和不同的炎症介质,包括C反应蛋白(CRP)和角质形成细胞来源的趋化因子(KC),支气管肺泡灌洗(BAL)液和血清。LPS不会进一步影响气道炎症细胞数量或粘液产生,但会降低BAL液中的炎症介质水平。香烟烟雾暴露后,脾脏中的T辅助(Th)1细胞增强;然而,结合LPS,香烟暴露导致Th1和Th2细胞增加。香烟烟雾暴露后,在近端小肠观察到组织形态学变化,而添加LPS则没有影响。香烟烟雾激活了小肠远端产生IgA的肠道免疫网络,这与粪便sIgA水平增加和Peyer斑块扩大有关。香烟烟雾加LPS降低了粪便sIgA水平和Peyer斑块的大小。总之,有或没有LPS的香烟烟雾会影响肠道健康,如通过肠道组织形态学和IgA产生的免疫网络的变化所观察到的。升高的全身介质可能在肺肠串扰中起作用。这些发现有助于更好地了解与COPD相关的肠道疾病。
    Chronic obstructive pulmonary disease (COPD) is often associated with intestinal comorbidities. In this study, changes in intestinal homeostasis and immunity in a cigarette smoke (CS)- and lipopolysaccharide (LPS)-induced COPD model were investigated. Mice were exposed to cigarette smoke or air for 72 days, except days 42, 52, and 62 on which the mice were treated with saline or LPS via intratracheal instillation. Cigarette smoke exposure increased the airway inflammatory cell numbers, mucus production, and different inflammatory mediators, including C-reactive protein (CRP) and keratinocyte-derived chemokine (KC), in bronchoalveolar lavage (BAL) fluid and serum. LPS did not further impact airway inflammatory cell numbers or mucus production but decreased inflammatory mediator levels in BAL fluid. T helper (Th) 1 cells were enhanced in the spleen after cigarette smoke exposure; however, in combination with LPS, cigarette exposure caused an increase in Th1 and Th2 cells. Histomorphological changes were observed in the proximal small intestine after cigarette smoke exposure, and addition of LPS had no effect. Cigarette smoke activated the intestinal immune network for IgA production in the distal small intestine that was associated with increased fecal sIgA levels and enlargement of Peyer\'s patches. Cigarette smoke plus LPS decreased fecal sIgA levels and the size of Peyer\'s patches. In conclusion, cigarette smoke with or without LPS affects intestinal health as observed by changes in intestinal histomorphology and immune network for IgA production. Elevated systemic mediators might play a role in the lung-gut cross talk. These findings contribute to a better understanding of intestinal disorders related to COPD.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    An electrothermal piezoresistive cantilever (EPC) sensor is a low-cost MEMS resonance sensor that provides self-actuating and self-sensing capabilities. In the platform, which is of MEMS-cantilever shape, the EPC sensor offers several advantages in terms of physical, chemical, and biological sensing, e.g., high sensitivity, low cost, simple procedure, and quick response. However, a crosstalk effect is generated by the coupling of parasitic elements from the actuation part to the sensing part. This study presents a parasitic feedthrough subtraction (PFS) method to mitigate a crosstalk effect in an electrothermal piezoresistive cantilever (EPC) resonance sensor. The PFS method is employed to identify a resonance phase that is, furthermore, deployed to a phase-locked loop (PLL)-based system to track and lock the resonance frequency of the EPC sensor under cigarette smoke exposure. The performance of the EPC sensor is further evaluated and compared to an AFM-microcantilever sensor and a commercial particle counter (DC1100-PRO). The particle mass-concentration measurement result generated from cigarette-smoke puffs shows a good agreement between these three detectors.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    烟草暴露对儿童健康的有害影响是众所周知的。尽管如此,在儿科人群中,二手烟或直接香烟烟雾暴露(CSE)的患病率并没有随着时间的推移而显著下降.相反,青少年使用电子烟的迅速倾向引起了公众的关注,因为越来越多的电子烟引起的急性肺损伤病例突显了这些新型“吸烟”装置的潜在危害。两个儿科人群特别容易受到香烟烟雾的有害影响。第一组是前早产儿,其风险由于其早产以及其他风险因素(例如氧气和机械通气)而升高,他们不成比例地暴露于其中。第二组是患有慢性呼吸道疾病的儿童和青少年,特别是哮喘和其他喘息疾病。2019年冠状病毒病(COVID-19)是由严重急性呼吸道综合症冠状病毒2(SARS-CoV-2)感染引起的一系列疾病,该疾病在过去一年中在全球蔓延。这里,从轻度到急性呼吸窘迫综合征(ARDS)的呼吸道症状在成人COVID-19病例中居于首位,在这个人群中,吸烟与更糟糕的结果有关,在这个人群中,吸烟与更糟糕的结果有关。有趣的是,SARS-CoV-2感染对儿童的感染易感性影响不同,疾病表现,和并发症。虽然儿童携带和传播病毒,症状性感染的可能性很低,与成年人相比,住院率和死亡率甚至更低。然而,在儿科人群中,多系统炎症综合征被认为是SARS-CoV-2感染的严重后果。此外,最近的数据表明,感染SARS-CoV-2的儿童发生多系统炎症综合征与严重的COVID-19。在这次审查中,在SARS-CoV-2大流行的背景下,我们强调了CSE对脆弱儿科人群的肺部影响.
    The detrimental effects of tobacco exposure on children\'s health are well known. Nonetheless, the prevalence of secondhand or direct cigarette smoke exposure (CSE) in the pediatric population has not significantly decreased over time. On the contrary, the rapid incline in use of e-cigarettes among adolescents has evoked public health concerns since increasing cases of vaping-induced acute lung injury have highlighted the potential harm of these new \"smoking\" devices. Two pediatric populations are especially vulnerable to the detrimental effects of cigarette smoke. The first group is former premature infants whose risk is elevated both due to their prematurity as well as other risk factors such as oxygen and mechanical ventilation to which they are disproportionately exposed. The second group is children and adolescents with chronic respiratory diseases, in particular asthma and other wheezing disorders. Coronavirus disease 2019 (COVID-19) is a spectrum of diseases caused by infection with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that has spread worldwide over the last year. Here, respiratory symptoms ranging from mild to acute respiratory distress syndrome (ARDS) are at the forefront of COVID-19 cases among adults, and cigarette smoking is associated with worse outcomes in this population, and cigarette smoking is associated with worse outcomes in this population. Interestingly, SARS-CoV-2 infection affects children differently in regard to infection susceptibility, disease manifestations, and complications. Although children carry and transmit the virus, the likelihood of symptomatic infection is low, and the rates of hospitalization and death are even lower when compared to the adult population. However, multisystem inflammatory syndrome is recognized as a serious consequence of SARS-CoV-2 infection in the pediatric population. In addition, recent data demonstrate specific clinical patterns in children infected with SARS-CoV-2 who develop multisystem inflammatory syndrome vs. severe COVID-19. In this review, we highlight the pulmonary effects of CSE in vulnerable pediatric populations in the context of the ongoing SARS-CoV-2 pandemic.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    我们通过测试原始量表的三部分结构,对适应的二手烟(SHS)问卷进行了心理测量分析,该量表用于测量家庭中的SHS暴露,工作和社会环境。
    对参加多站点随机对照试验的839名每日吸烟者进行了15项问卷。在并行分析之后,我们进行了确定三因素结构的验证性因素分析.计算克朗巴赫的阿尔法和拟合指数以评估内部一致性。通过将社会环境子量表与威斯康星州吸烟依赖动机简要清单社会/环境行为子量表进行比较来评估标准有效性。使用线性回归和危害的烟草生物标志物评估问卷的预测效度;NNAL,过期的一氧化碳和总的可替宁。
    五个项目没有加载到任何因素上,被丢弃了,产生10个项目的问卷。克朗巴赫的阿尔法是(0.86),(0.77)和(0.67)的工作,社会,和家庭分量表,分别。WISDM子量表与社会子量表的得分呈中度相关(r=0.57,p<0.001)。问卷显示,通过每天吸烟来衡量,烟雾暴露量高于个人报告的使用量。
    出现了三个结构;结果表明,在未来的研究中可以使用缩短的10项量表。
    We conducted a psychometric analysis of an adapted secondhand smoke (SHS) questionnaire by testing the three-component structure of the original scale that measures SHS exposure in home, work and social environments.
    The 15-item questionnaire was administered to 839 daily smokers participating in a multi-site randomized controlled trial. Following parallel analysis, we conducted a confirmatory factor analysis specifying a three-factor structure. Cronbach\'s alphas and fit indices were calculated to assess internal consistency. Criterion validity was assessed by comparing the Social environments subscale to the Brief Wisconsin Inventory of Smoking Dependence Motives Social/Environmental Goads subscale. Predicative validity of the questionnaire was assessed using linear regressions and tobacco biomarkers of harm; NNAL, expired carbon monoxide and total cotinine.
    Five items did not load onto any factor and were dropped, resulting in a 10-item questionnaire. The Cronbach\'s alphas were (0.86), (0.77) and (0.67) for the Work, Social, and Home subscales, respectively. The WISDM subscale was moderately correlated with scores on the Social subscale (r = 0.57, p < 0.001). The questionnaire demonstrated predictive validity of smoke exposure above individual\'s own reported use as measured by cigarettes smoked per day.
    Three constructs emerged; results indicate that a shortened 10-item scale could be used in future studies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Despite a decline in popularity over the past several decades, cigarette smoking remains a leading cause of cardiovascular morbidity and mortality. Yet, the effects of cigarette smoking on vascular structure and function are largely unknown. To evaluate changes in the mechanical properties of the aorta that occur with chronic smoking, we exposed female apolipoprotein E-deficient mice to mainstream cigarette smoke daily for 24 wk, with room air as control. By the time of euthanasia, cigarette-exposed mice had lower body mass but experienced larger systolic/diastolic blood pressure when compared with controls. Smoking was associated with significant wall thickening, reduced axial stretch, and circumferential material softening of the aorta. Although this contributed to maintaining intrinsic tissue stiffness at control levels despite larger pressure loads, the structural stiffness became significantly larger. Furthermore, the aorta from cigarette-exposed mice exhibited decreased ability to store elastic energy and augment diastolic blood flow. Histological analysis revealed a region-dependent increase in the cross-sectional area due to smoking. Increased smooth muscle and extracellular matrix content led to medial thickening in the ascending aorta, whereas collagen deposition increased the thickness of the descending thoracic and abdominal aorta. Atherosclerotic lesions were larger in exposed vessels and featured a necrotic core overlaid by a thinned fibrous cap and macrophage infiltration, consistent with a vulnerable phenotype. Collectively, our data indicate that cigarette smoking decreases the mechanical functionality of the aorta, inflicts morphometric alterations to distinct segments of the aorta, and accelerates the progression of atherosclerosis.NEW & NOTEWORTHY We studied the effects of chronic cigarette smoking on the structure and function of the aorta in a mouse model of nose-only aerosol inhalation. Our data indicated that exposure to cigarette smoke impairs vascular function by reducing the ability of the aorta to store elastic energy and by decreasing aortic distensibility. Combined with a more vulnerable atherosclerotic phenotype, these findings reveal the biomechanical mechanisms that support the development of cardiovascular disease due to cigarette smoking.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    OBJECTIVE: Patients with inflammatory bowel disease are at increased risk of colorectal neoplasia (CRN) due to mucosal inflammation. As current surveillance guidelines form a burden on patients and healthcare costs, stratification of high-risk patients is crucial. Cigarette smoke reduces inflammation in ulcerative colitis (UC) but not Crohn\'s disease (CD) and forms a known risk factor for CRN in the general population. Due to this divergent association, the effect of smoking on CRN in IBD is unclear and subject of this study.
    METHODS: In this retrospective cohort study, 1,386 IBD patients with previous biopsies analyzed and reported in the PALGA register were screened for development of CRN. Clinical factors and cigarette smoke were evaluated. Patients were stratified for guideline-based risk of CRN. Cox-regression modeling was used to estimate the effect of cigarette smoke and its additive effect within the current risk stratification for prediction of CRN.
    RESULTS: 153 (11.5%) patients developed CRN. Previously described risk factors, i.e. first-degree family member with CRN in CD (p-value=.001), presence of post-inflammatory polyps in UC (p-value=.005), were replicated. Former smoking increased risk of CRN in UC (HR 1.73; 1.05-2.85), whereas passive smoke exposure yielded no effect. For CD, active smoking (2.20; 1.02-4.76) and passive smoke exposure (1.87; 1.09-3.20) significantly increased CRN risk. Addition of smoke exposure to the current risk-stratification model significantly improved model fit for CD.
    CONCLUSIONS: This study is the first to describe the important role of cigarette smoke in CRN development in IBD patients. Adding this risk factor improves the current risk stratification for CRN surveillance strategies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Smokers with apparently \"healthy\" lungs suffer from more severe and frequent viral respiratory infections, but the mechanisms underlying this observation are still unclear. Epithelial cells and dendritic cells (DC) form the first line of defense against inhaled noxes such as smoke or viruses. We therefore aimed to obtain insight into how cigarette smoke affects DCs and epithelial cells and how this influences the response to viral infection. Female C57BL/6J mice were exposed to cigarette smoke (CS) for 1 h daily for 24 days and then challenged i.n. with the viral mimic and Toll-like receptor 3 (TLR3) ligand poly (I:C) after the last exposure. DC subpopulations were analyzed 24 h later in whole lung homogenates by flow cytometry. Calu-3 cells or human precision-cut lung slices (PCLS) cultured at air-liquid interface were exposed to CS or air and subsequently inoculated with influenza H1N1. At 48 h post infection cytokines were analyzed by multiplex technology. Cytotoxic effects were measured by release of lactate dehydrogenase (LDH) and confocal imaging. In Calu-3 cells the trans-epithelial electrical resistance (TEER) was assessed. Smoke exposure of mice increased numbers of inflammatory and plasmacytoid DCs in lung tissue. Additional poly (I:C) challenge further increased the population of inflammatory DCs and conventional DCs, especially CD11b+ cDCs. Smoke exposure led to a loss of the barrier function in Calu-3 cells, which was further exaggerated by additional influenza H1N1 infection. Influenza H1N1-induced secretion of antiviral cytokines (IFN-α2a, IFN-λ, interferon-γ-induced protein 10 [IP-10]), pro-inflammatory cytokine IL-6, as well as T cell-associated cytokines (e.g., I-TAC) were completely suppressed in both Calu-3 cells and human PCLS after smoke exposure. In summary, cigarette smoke exposure increased the number of inflammatory DCs in the lung and disrupted epithelial barrier functions, both of which was further enhanced by viral stimulation. Additionally, the antiviral immune response to influenza H1N1 was strongly suppressed by smoke. These data suggest that smoke impairs protective innate mechanisms in the lung, which could be responsible for the increased susceptibility to viral infections in \"healthy\" smokers.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    UNASSIGNED: Physiological rhythms in mammals are essential for maintaining health, whereas disruptions may cause or exacerbate disease pathogenesis. As such, our objective was to characterize how cigarette smoke exposure affects physiological rhythms of otherwise healthy mice using telemetry and cosinor analysis.
    UNASSIGNED: Female BALB/c mice were implanted with telemetry devices to measure body temperature, heart rate, systolic blood pressure (SBP), and activity. Following baseline measurements, mice were exposed to cigarette smoke for approximately 50 min twice daily during weekdays over 24 weeks. Physiological parameters were recorded after 1, 4, 8, and 24 weeks of exposure or after 4 weeks cessation following 4 weeks of cigarette smoke exposure.
    UNASSIGNED: Acute cigarette smoke exposure resulted in anapyrexia, and bradycardia, with divergent effects on SBP. Long term, cigarette smoke exposure disrupted physiological rhythms after just 1 week, which persisted across 24 weeks of exposure (as shown by mixed effects on mesor, amplitude, acrophase, and goodness-of-fit using cosinor analysis). Four weeks of cessation was insufficient to allow full recovery of rhythms.
    UNASSIGNED: Our characterization of the pathophysiology of cigarette smoke exposure on physiological rhythms of mice suggests that rhythm disruption may precede and contribute to disease pathogenesis. These findings provide a clear rationale and guide for the future use of chronotherapeutics.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号