Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Biochem Pharmacol ; 222: 116109, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38458330

RESUMO

Angiotensin (Ang)-(1-7) stimulates vasoprotective functions of diabetic (DB) CD34+ hematopoietic stem/progenitor cells partly by decreasing reactive oxygen species (ROS), increasing nitric oxide (NO) levels and decreasing TGFß1 secretion. Telomerase reverse transcriptase (TERT) translocates to mitochondria and regulates ROS generation. Alternative splicing of TERT results in variants α-, ß- and α-ß-TERT, which may oppose functions of full-length (FL) TERT. This study tested if the protective functions of Ang-(1-7) or TGFß1-silencing are mediated by mitoTERT and that diabetes decreases FL-TERT expression by inducing splicing. CD34+ cells were isolated from the peripheral blood mononuclear cells of nondiabetic (ND, n = 68) or DB (n = 74) subjects. NO and mitoROS levels were evaluated by flow cytometry. TERT splice variants and mitoDNA-lesions were characterized by qPCR. TRAP assay was used for telomerase activity. Decoy peptide was used to block mitochondrial translocation (mitoXTERT). TERT inhibitor or mitoXTERT prevented the effects of Ang-(1-7) on NO or mitoROS levels in DB-CD34+ cells. FL-TERT expression and telomerase activity were lower and mitoDNA-lesions were higher in DB cells compared to ND and were reversed by Ang-(1-7) or TGFß1-silencing. The prevalence of TERT splice variants, with predominant ß-TERT expression, was higher and the expression of FL-TERT was lower in DB cells (n = 25) compared to ND (n = 30). Ang-(1-7) or TGFß1-silencing decreased TERT-splicing and increased FL-TERT. Blocking of ß-splicing increased FL-TERT and protected mitoDNA in DB-cells. The findings suggest that diabetes induces TERT-splicing in CD34+ cells and that ß-TERT splice variant largely contributes to the mitoDNA oxidative damage.


Assuntos
Angiotensina I , Diabetes Mellitus , Fragmentos de Peptídeos , Telomerase , Humanos , Telomerase/genética , Telomerase/metabolismo , Telomerase/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Leucócitos Mononucleares , Mitocôndrias/metabolismo , Diabetes Mellitus/metabolismo
2.
Heart Lung Circ ; 33(5): 576-604, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38184426

RESUMO

BACKGROUND: Cancer and cardiovascular disease (CVD) are major causes of morbidity and mortality in the United States (US). Cancer survivors have increased risks for CVD and CVD-related mortality due to multiple factors including cancer treatment-related cardiotoxicity. Disparities are rooted in differential exposure to risk factors and social determinants of health (SDOH), including systemic racism. This review aimed to assess SDOH's role in disparities, document CVD-related disparities among US cancer survivors, and identify literature gaps for future research. METHODS: Following the Peer Review of Electronic Search Strategies (PRESS) guidelines, MEDLINE, PsycINFO, and Scopus were searched on March 15, 2021, with an update conducted on September 26, 2023. Articles screening was performed using the Preferred Reporting Items for Systematic Review and Meta-Analysis (PRISMA) 2020, a pre-defined Population, Exposure, Comparison, Outcomes, and Settings (PECOS) framework, and the Rayyan platform. A modified version of the Newcastle-Ottawa Scale was used to assess the risk of bias, and RAW Graphs for alluvial charts. This review is registered with PROSPERO under ID #CRD42021236460. RESULTS: Out of 7,719 retrieved articles, 24 were included, and discussed diverse SDOH that contribute to CVD-related disparities among cancer survivors. The 24 included studies had a large combined total sample size (n=7,704,645; median=19,707). While various disparities have been investigated, including rural-urban, sex, socioeconomic status, and age, a notable observation is that non-Hispanic Black cancer survivors experience disproportionately adverse CVD outcomes when compared to non-Hispanic White survivors. This underscores historical racism and discrimination against non-Hispanic Black individuals as fundamental drivers of CVD-related disparities. CONCLUSIONS: Stakeholders should work to eliminate the root causes of disparities. Clinicians should increase screening for risk factors that exacerbate CVD-related disparities among cancer survivors. Researchers should prioritise the investigation of systemic factors driving disparities in cancer and CVD and develop innovative interventions to mitigate risk in cancer survivors.


Assuntos
Sobreviventes de Câncer , Doenças Cardiovasculares , Neoplasias , Humanos , Sobreviventes de Câncer/estatística & dados numéricos , Doenças Cardiovasculares/epidemiologia , Doenças Cardiovasculares/etnologia , Estados Unidos/epidemiologia , Neoplasias/epidemiologia , Neoplasias/terapia , Fatores de Risco , Disparidades nos Níveis de Saúde
3.
Life Sci Alliance ; 7(2)2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38052461

RESUMO

Gleason grading is an important prognostic indicator for prostate adenocarcinoma and is crucial for patient treatment decisions. However, intermediate-risk patients diagnosed in the Gleason grade group (GG) 2 and GG3 can harbour either aggressive or non-aggressive disease, resulting in under- or overtreatment of a significant number of patients. Here, we performed proteomic, differential expression, machine learning, and survival analyses for 1,348 matched tumour and benign sample runs from 278 patients. Three proteins (F5, TMEM126B, and EARS2) were identified as candidate biomarkers in patients with biochemical recurrence. Multivariate Cox regression yielded 18 proteins, from which a risk score was constructed to dichotomize prostate cancer patients into low- and high-risk groups. This 18-protein signature is prognostic for the risk of biochemical recurrence and completely independent of the intermediate GG. Our results suggest that markers generated by computational proteomic profiling have the potential for clinical applications including integration into prostate cancer management.


Assuntos
Neoplasias da Próstata , Proteômica , Masculino , Humanos , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Fatores de Risco , Gradação de Tumores
4.
Nat Aging ; 3(11): 1345-1357, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37783816

RESUMO

In humans, aggregation of polyglutamine repeat (polyQ) proteins causes disorders such as Huntington's disease. Although plants express hundreds of polyQ-containing proteins, no pathologies arising from polyQ aggregation have been reported. To investigate this phenomenon, we expressed an aggregation-prone fragment of human huntingtin (HTT) with an expanded polyQ stretch (Q69) in Arabidopsis thaliana plants. In contrast to animal models, we find that Arabidopsis sp. suppresses Q69 aggregation through chloroplast proteostasis. Inhibition of chloroplast proteostasis diminishes the capacity of plants to prevent cytosolic Q69 aggregation. Moreover, endogenous polyQ-containing proteins also aggregate on chloroplast dysfunction. We find that Q69 interacts with the chloroplast stromal processing peptidase (SPP). Synthetic Arabidopsis SPP prevents polyQ-expanded HTT aggregation in human cells. Likewise, ectopic SPP expression in Caenorhabditis elegans reduces neuronal Q67 aggregation and subsequent neurotoxicity. Our findings suggest that synthetic plant proteins, such as SPP, hold therapeutic potential for polyQ disorders and other age-related diseases involving protein aggregation.


Assuntos
Arabidopsis , Agregados Proteicos , Animais , Humanos , Arabidopsis/genética , Peptídeos/genética , Neurônios/metabolismo , Caenorhabditis elegans/genética
5.
Eur J Radiol Open ; 10: 100470, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36590327

RESUMO

Rationale and objectives: Patients receiving high cumulative effective doses (CED) from recurrent computed tomography (CT) in a real-life setting are not well identified. Evaluation of causes and patient characteristics may help to define individuals potentially at risk of radiation-induced secondary malignancies. Materials and methods: Patients who received a CED > 100 mSv from CT scans during October 2012 and April 2020 at a tertiary university center were identified with the help of a radiological radiation dose monitoring system. The primary disease and referral diagnosis, number of CT exams, time period, age, BMI and gender distribution of the 1000 patients with the highest CED were analysed. Results: 3431 patients had a CED of more than 100 mSv, which corresponded to 2.75% of all patients who received a CT exam. From the 1000 patients with the highest CED, mean number of CT exams per patient was 14.6, mean CED was 257 mSv (SD 98, range 177-1339). Mean age of patients was 63.9 years (SD 10.6), male to female ratio 3:2, and mean BMI 28.7 kg/m2 (SD 5.5). 728 (72.9%) patients had cancer. The leading primary diagnosis was liver cirrhosis in 197 patients and 103 patients had a liver transplantation. In patients with liver cirrhosis, 750 exams were indicated for the follow-up of the disease, 662 for the clarification of an acute clinical condition, and 202 for CT-guided stereotactic radiofrequency ablation. Conclusion: Recurrent CT scans of patients with cancer, liver cirrhosis and liver transplantation may lead to critically high CED.

6.
PLoS One ; 17(11): e0276517, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36417344

RESUMO

SIGNIFICANCE: Globally, cardiovascular disease (CVD) and cancer are leading causes of morbidity and mortality. While having different etiologies, CVD and cancer are linked by multiple shared risk factors, the presence of which exacerbate adverse outcomes for individuals with either disease. For both pathologies, factors such as poverty, lack of physical activity (PA), poor dietary intake, and climate change increase risk of adverse outcomes. Prior research has shown that greenspaces and other nature-based interventions (NBIs) contribute to improved health outcomes and climate change resilience. OBJECTIVE: To summarize evidence on the impact of greenspaces or NBIs on cardiovascular health and/or cancer-related outcomes and identify knowledge gaps to inform future research. METHODS: Following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) 2020 and Peer Review of Electronic Search Strategies (PRESS) guidelines, we searched five databases: Web of Science, Scopus, Medline, PsycINFO and GreenFile. Two blinded reviewers used Rayyan AI and a predefined criteria for article inclusion and exclusion. The risk of bias was assessed using a modified version of the Newcastle-Ottawa Scale (NOS). This review is registered with PROSPERO, ID # CRD42021231619. RESULTS & DISCUSSION: Of 2565 articles retrieved, 31 articles met the inclusion criteria, and overall had a low risk of bias. 26 articles studied cardiovascular related outcomes and 5 studied cancer-related outcomes. Interventions were coded into 4 categories: forest bathing, green exercise, gardening, and nature viewing. Outcomes included blood pressure (BP), cancer-related quality of life (QoL) and (more infrequently) biomarkers of CVD risk. Descriptions of findings are presented as well as visual presentations of trends across the findings using RAW graphs. Overall studies included have a low risk of bias; and alluvial chart trends indicated that NBIs may have beneficial effects on CVD and cancer-related outcomes. CONCLUSIONS & IMPLICATIONS: (1) Clinical implication: Healthcare providers should consider the promotion of nature-based programs to improve health outcomes. (2) Policy implication: There is a need for investment in equitable greenspaces to improve health outcomes and build climate resilient neighborhoods. (3) Research or academic implication: Research partnerships with community-based organizations for a comprehensive study of benefits associated with NBIs should be encouraged to reduce health disparities and ensure intergenerational health equity. There is a need for investigation of the mechanisms by which NBIs impact CVD and exploration of the role of CVD biological markers of inflammation among cancer survivors.


Assuntos
Doenças Cardiovasculares , Neoplasias , Humanos , Qualidade de Vida , Parques Recreativos , Exercício Físico , Pressão Sanguínea , Neoplasias/terapia
7.
Opt Express ; 30(13): 23544-23555, 2022 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-36225031

RESUMO

We report on new THz electromagnetic emission mechanism from deformational coupling of acoustic (AC) phonons with electrons in the propagation medium of non-polar Si. The epicenters of the AC phonon pulses are the surface and interface of a GaP transducer layer whose thickness (d) is varied in nanoscale from 16 to 45 nm. The propagating AC pulses locally modulate the bandgap, which in turn generates a train of electric field pulses, inducing an abrupt drift motion at the depletion edge of Si. The fairly time-delayed THz bursts, centered at different times (t1T H z, t2T H z, and t3T H z), are concurrently emitted only when a series of AC pulses reach the point of the depletion edge of Si, even without any piezoelectricity. The analysis on the observed peak emission amplitudes is consistent with calculations based on the combined effects of mobile charge carrier density and AC-phonon-induced local deformation, which recapitulates the role of deformational potential coupling in THz wave emission in a formulatively distinct manner from piezoelectric counterpart.

8.
Am J Physiol Heart Circ Physiol ; 323(4): H749-H762, 2022 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-36018760

RESUMO

Aging is associated with blunted coronary microvascular vasodilatory function. Previously, systemically administered adipose stromal vascular fraction (SVF) therapy reversed aging-induced attenuation of ß1-adrenergic- and flow-mediated dilation dependent on reducing mitochondrial reactive oxygen species. We hypothesized that SVF-mediated recovery of microvascular dilatory function is dependent on recovery of mitochondrial function, specifically by reducing mitochondrial hyperfission. Female Fischer-344 rats were allocated into young control, old control, and old + SVF therapy groups. Pressure myography, immunofluorescent staining, Western blot analysis, and RNA sequencing were performed to determine coronary microvascular mitochondrial dynamics and function. Gene and protein expression of fission-mediator DRP-1 was enhanced with aging but reversed by SVF therapy. SVF facilitated an increase in fusion-mediator MFN-1 gene and protein expression. Mitochondrial morphology was characterized as rod-like and densely networked in young controls, isolated circular and punctate with aging, and less circularity with partially restored mitochondrial branch density with SVF therapy. Decreased mitochondrial membrane potential and ATP bioavailability in aged animals at baseline and during flow-mediated dilation were reversed by SVF and accompanied with enhanced oxygen consumption. Dilation to norepinephrine and flow in young controls were dependent on uninhibited mitochondrial fusion, whereas inhibiting fission did not restore aged microvessel response to norepinephrine or flow. SVF-mediated recovery of ß-adrenergic function was dependent on uninhibited mitochondrial fusion, whereas recovery of flow-mediated dilation was dependent on maintained mitochondrial fission. Impaired dilation in aging is mitigated by SVF therapy, which recovers mitochondrial function and fission/fusion balance.NEW & NOTEWORTHY We elucidated the consequences of aging on coronary microvascular mitochondrial health as well as SVF's ability to reverse these effects. Aging shifts gene/protein expression and mitochondrial morphology indicating hyperfission, alongside attenuated mitochondrial membrane potential and ATP bioavailability, all reversed using SVF therapy. Mitochondrial membrane potential and ATP levels correlated with vasodilatory efficiency. Mitochondrial dysfunction is a contributing pathological factor in aging that can be targeted by therapeutic SVF to preserve microvascular dilative function.


Assuntos
Tecido Adiposo , Células Estromais , Trifosfato de Adenosina/metabolismo , Tecido Adiposo/metabolismo , Adrenérgicos , Animais , Feminino , Mitocôndrias , Norepinefrina/metabolismo , Ratos , Ratos Endogâmicos F344 , Espécies Reativas de Oxigênio/metabolismo , Células Estromais/metabolismo , Fração Vascular Estromal
9.
Am J Physiol Heart Circ Physiol ; 323(4): H702-H720, 2022 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-35930448

RESUMO

Many anticancer therapies (CTx) have cardiotoxic side effects that limit their therapeutic potential and cause long-term cardiovascular complications in cancer survivors. This has given rise to the field of cardio-oncology, which recognizes the need for basic, translational, and clinical research focused on understanding the complex signaling events that drive CTx-induced cardiovascular toxicity. Several CTx agents cause mitochondrial damage in the form of mitochondrial DNA deletions, mutations, and suppression of respiratory function and ATP production. In this review, we provide a brief overview of the cardiovascular complications of clinically used CTx agents and discuss current knowledge of local and systemic secondary signaling events that arise in response to mitochondrial stress/damage. Mitochondrial oxidative stress has long been recognized as a contributor to CTx-induced cardiotoxicity; thus, we focus on emerging roles for mitochondria in epigenetic regulation, innate immunity, and signaling via noncoding RNAs and mitochondrial hormones. Because data exploring mitochondrial secondary signaling in the context of cardio-oncology are limited, we also draw upon clinical and preclinical studies, which have examined these pathways in other relevant pathologies.


Assuntos
Antineoplásicos , Cardiopatias , Neoplasias , Trifosfato de Adenosina/metabolismo , Antineoplásicos/farmacologia , Cardiotoxicidade/etiologia , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Epigênese Genética , Hormônios/farmacologia , Humanos , Neoplasias/tratamento farmacológico , Estresse Oxidativo
10.
Front Oncol ; 12: 828177, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35311118

RESUMO

The genetic bases and disparate responses to radiotherapy are poorly understood, especially for cardiotoxicity resulting from treatment of thoracic tumors. Preclinical animal models such as the Dahl salt-sensitive (SS) rat can serve as a surrogate model for salt-sensitive low renin hypertension, common to African Americans, where aldosterone contributes to hypertension-related alterations of peripheral vascular and renal vascular function. Brown Norway (BN) rats, in comparison, are a normotensive control group, while consomic SSBN6 with substitution of rat chromosome 6 (homologous to human chromosome 14) on an SS background manifests cardioprotection and mitochondrial preservation to SS rats after injury. In this study, 2 groups from each of the 3 rat strains had their hearts irradiated (8 Gy X 5 fractions). One irradiated group was treated with the ACE-inhibitor lisinopril, and a separate group in each strain served as nonirradiated controls. Radiation reduced cardiac end diastolic volume by 9-11% and increased thickness of the interventricular septum (11-16%) and left ventricular posterior wall (14-15%) in all 3 strains (5-10 rats/group) after 120 days. Lisinopril mitigated the increase in posterior wall thickness. Mitochondrial function was measured by the Seahorse Cell Mitochondrial Stress test in peripheral blood mononuclear cells (PBMC) at 90 days. Radiation did not alter mitochondrial respiration in PBMC from BN or SSBN6. However, maximal mitochondrial respiration and spare capacity were reduced by radiation in PBMC from SS rats (p=0.016 and 0.002 respectively, 9-10 rats/group) and this effect was mitigated by lisinopril (p=0.04 and 0.023 respectively, 9-10 rats/group). Taken together, these results indicate injury to the heart by radiation in all 3 strains of rats, although the SS rats had greater susceptibility for mitochondrial dysfunction. Lisinopril mitigated injury independent of genetic background.

11.
Artigo em Inglês | MEDLINE | ID: mdl-35206610

RESUMO

Cardiovascular disease (CVD) is a leading cause of global morbidity and mortality. Cancer survivors have significantly elevated risk of poor cardiovascular (CV) health outcomes due to close co-morbid linkages and shared risk factors between CVD and cancer, as well as adverse effects of cancer treatment-related cardiotoxicity. CVD and cancer-related outcomes are exacerbated by increased risk of inflammation. Results from different pharmacological interventions aimed at reducing inflammation and risk of major adverse cardiovascular events (MACEs) have been largely mixed to date. Greenspaces have been shown to reduce inflammation and have been associated with CV health benefits, including reduced CVD behavioral risk factors and overall improvement in CV outcomes. Greenspace may, thus, serve to alleviate the CVD burden among cancer survivors. To understand pathways through which greenspace can prevent or reduce adverse CV outcomes among cancer survivors, we review the state of knowledge on associations among inflammation, CVD, cancer, and existing pharmacological interventions. We then discuss greenspace benefits for CV health from ecological to multilevel studies and a few existing experimental studies. Furthermore, we review the relationship between greenspace and inflammation, and we highlight forest bathing in Asian-based studies while presenting existing research gaps in the US literature. Then, we use the socioecological model of health to present an expanded conceptual framework to help fill this US literature gap. Lastly, we present a way forward, including implications for translational science and a brief discussion on necessities for virtual nature and/or exposure to nature images due to the increasing human-nature disconnect; we also offer guidance for greenspace research in cardio-oncology to improve CV health outcomes among cancer survivors.


Assuntos
Sobreviventes de Câncer , Doenças Cardiovasculares , Sistema Cardiovascular , Neoplasias , Doenças Cardiovasculares/etiologia , Humanos , Inflamação/complicações , Neoplasias/complicações , Neoplasias/epidemiologia , Parques Recreativos
12.
Pharmacol Ther ; 237: 108116, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35063569

RESUMO

Recent strides in anti-cancer therapeutics have improved longevity and led to a growing population of cancer survivors, who are increasingly likely to die of other causes. Treatment-induced cardiotoxicity is a complication of several therapeutic agents with acute and long-term consequences for cancer patients. Vascular endothelial dysfunction is a precursor and hallmark of ischemic coronary disease and may play a role in anti-cancer therapy-induced cardiotoxicity. This review summarizes clinical evidence for endothelial dysfunction following anti-cancer therapy and extends the discussion to include the impact of therapeutic agents on conduit arteries and the microcirculation. We highlight the role of innate immune system activation and cross-talk between inflammation and oxidative stress as pathogenic mechanisms underlying anti-cancer therapy-induced vascular toxicity. Understanding the impact of anti-cancer agents on the vascular endothelium will inform therapeutic approaches to prevent or reverse treatment-induced cardiotoxicity and may serve as an important tool to predict, monitor, and prevent adverse cardiovascular outcomes in patients undergoing treatment.


Assuntos
Antineoplásicos , Neoplasias , Antineoplásicos/efeitos adversos , Cardiotoxicidade/etiologia , Endotélio Vascular , Humanos , Microcirculação , Neoplasias/tratamento farmacológico
14.
PLoS Comput Biol ; 17(11): e1009161, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34762640

RESUMO

Network propagation refers to a class of algorithms that integrate information from input data across connected nodes in a given network. These algorithms have wide applications in systems biology, protein function prediction, inferring condition-specifically altered sub-networks, and prioritizing disease genes. Despite the popularity of network propagation, there is a lack of comparative analyses of different algorithms on real data and little guidance on how to select and parameterize the various algorithms. Here, we address this problem by analyzing different combinations of network normalization and propagation methods and by demonstrating schemes for the identification of optimal parameter settings on real proteome and transcriptome data. Our work highlights the risk of a 'topology bias' caused by the incorrect use of network normalization approaches. Capitalizing on the fact that network propagation is a regularization approach, we show that minimizing the bias-variance tradeoff can be utilized for selecting optimal parameters. The application to real multi-omics data demonstrated that optimal parameters could also be obtained by either maximizing the agreement between different omics layers (e.g. proteome and transcriptome) or by maximizing the consistency between biological replicates. Furthermore, we exemplified the utility and robustness of network propagation on multi-omics datasets for identifying ageing-associated genes in brain and liver tissues of rats and for elucidating molecular mechanisms underlying prostate cancer progression. Overall, this work compares different network propagation approaches and it presents strategies for how to use network propagation algorithms to optimally address a specific research question at hand.


Assuntos
Algoritmos , Biologia Computacional/métodos , Envelhecimento/genética , Envelhecimento/metabolismo , Animais , Viés , Encéfalo/metabolismo , Biologia Computacional/estatística & dados numéricos , Interpretação Estatística de Dados , Progressão da Doença , Perfilação da Expressão Gênica/estatística & dados numéricos , Redes Reguladoras de Genes , Genômica/estatística & dados numéricos , Humanos , Fígado/metabolismo , Masculino , Neoplasias da Próstata/etiologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Mapas de Interação de Proteínas , Proteômica/estatística & dados numéricos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Biologia de Sistemas
15.
Am J Physiol Heart Circ Physiol ; 321(5): H985-H1003, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34559580

RESUMO

Ventilation with gases containing enhanced fractions of oxygen is the cornerstone of therapy for patients with hypoxia and acute respiratory distress syndrome. Yet, hyperoxia treatment increases free reactive oxygen species (ROS)-induced lung injury, which is reported to disrupt autophagy/mitophagy. Altered extranuclear activity of the catalytic subunit of telomerase, telomerase reverse transcriptase (TERT), plays a protective role in ROS injury and autophagy in the systemic and coronary endothelium. We investigated interactions between autophagy/mitophagy and TERT that contribute to mitochondrial dysfunction and pulmonary injury in cultured rat lung microvascular endothelial cells (RLMVECs) exposed in vitro, and rat lungs exposed in vivo to hyperoxia for 48 h. Hyperoxia-induced mitochondrial damage in rat lungs [TOMM20, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT)], which was paralleled by increased markers of inflammation [myeloperoxidase (MPO), IL-1ß, TLR9], impaired autophagy signaling (Beclin-1, LC3B-II/1, and p62), and decreased the expression of TERT. Mitochondrial-specific autophagy (mitophagy) was not altered, as hyperoxia increased expression of Pink1 but not Parkin. Hyperoxia-induced mitochondrial damage (TOMM20) was more pronounced in rats that lack the catalytic subunit of TERT and resulted in a reduction in cellular proliferation rather than cell death in RLMVECs. Activation of TERT or autophagy individually offset mitochondrial damage (MTT). Combined activation/inhibition failed to alleviate hyperoxic-induced mitochondrial damage in vitro, whereas activation of autophagy in vivo decreased mitochondrial damage (MTT) in both wild type (WT) and rats lacking TERT. Functionally, activation of either TERT or autophagy preserved transendothelial membrane resistance. Altogether, these observations show that activation of autophagy/mitophagy and/or TERT mitigate loss of mitochondrial function and barrier integrity in hyperoxia.NEW & NOTEWORTHY In cultured pulmonary artery endothelial cells and in lungs exposed in vivo to hyperoxia, autophagy is activated, but clearance of autophagosomes is impaired in a manner that suggests cross talk between TERT and autophagy. Stimulation of autophagy prevents hyperoxia-induced decreases in mitochondrial metabolism and sustains monolayer resistance. Hyperoxia increases mitochondrial outer membrane (TOMM20) protein, decreases mitochondrial function, and reduces cellular proliferation without increasing cell death.


Assuntos
Células Endoteliais/enzimologia , Hiperóxia/complicações , Lesão Pulmonar/enzimologia , Pulmão/irrigação sanguínea , Microvasos/enzimologia , Mitocôndrias/enzimologia , Mitofagia , Telomerase/metabolismo , Animais , Proteínas Relacionadas à Autofagia/metabolismo , Permeabilidade Capilar , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/patologia , Feminino , Técnicas de Inativação de Genes , Mediadores da Inflamação/metabolismo , Lesão Pulmonar/etiologia , Lesão Pulmonar/genética , Lesão Pulmonar/patologia , Masculino , Proteínas de Membrana Transportadoras/metabolismo , Microvasos/patologia , Mitocôndrias/genética , Mitocôndrias/patologia , Proteínas do Complexo de Importação de Proteína Precursora Mitocondrial , Ratos Sprague-Dawley , Ratos Transgênicos , Receptores de Superfície Celular/metabolismo , Telomerase/deficiência , Telomerase/genética , Receptor 4 Toll-Like/metabolismo , Receptor Toll-Like 9/metabolismo
16.
Circ Res ; 129(1): e21-e34, 2021 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-33934611

RESUMO

Although cardiovascular toxicity from traditional chemotherapies has been well recognized for decades, the recent explosion of effective novel targeted cancer therapies with cardiovascular sequelae has driven the emergence of cardio-oncology as a new clinical and research field. Cardiovascular toxicity associated with cancer therapy can manifest as a broad range of potentially life-threatening complications, including heart failure, arrhythmia, myocarditis, and vascular events. Beyond toxicology, the intersection of cancer and heart disease has blossomed to include discovery of genetic and environmental risk factors that predispose to both. There is a pressing need to understand the underlying molecular mechanisms of cardiovascular toxicity to improve outcomes in patients with cancer. Preclinical cardiovascular models, ranging from cellular assays to large animals, serve as the foundation for mechanistic studies, with the ultimate goal of identifying biologically sound biomarkers and cardioprotective therapies that allow the optimal use of cancer treatments while minimizing toxicities. Given that novel cancer therapies target specific pathways integral to normal cardiovascular homeostasis, a better mechanistic understanding of toxicity may provide insights into fundamental pathways that lead to cardiovascular disease when dysregulated. The goal of this scientific statement is to summarize the strengths and weaknesses of preclinical models of cancer therapy-associated cardiovascular toxicity, to highlight overlapping mechanisms driving cancer and cardiovascular disease, and to discuss opportunities to leverage cardio-oncology models to address important mechanistic questions relevant to all patients with cardiovascular disease, including those with and without cancer.


Assuntos
Antineoplásicos/toxicidade , Cardiopatias/induzido quimicamente , Miócitos Cardíacos/efeitos dos fármacos , Testes de Toxicidade , American Heart Association , Animais , Cardiotoxicidade , Células Cultivadas , Modelos Animais de Doenças , Cardiopatias/genética , Cardiopatias/metabolismo , Cardiopatias/patologia , Humanos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Medição de Risco , Estados Unidos
17.
Cell Syst ; 12(5): 401-418.e12, 2021 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-33932331

RESUMO

One goal of precision medicine is to tailor effective treatments to patients' specific molecular markers of disease. Here, we used mass cytometry to characterize the single-cell signaling landscapes of 62 breast cancer cell lines and five lines from healthy tissue. We quantified 34 markers in each cell line upon stimulation by the growth factor EGF in the presence or absence of five kinase inhibitors. These data-on more than 80 million single cells from 4,000 conditions-were used to fit mechanistic signaling network models that provide insight into how cancer cells process information. Our dynamic single-cell-based models accurately predicted drug sensitivity and identified genomic features associated with drug sensitivity, including a missense mutation in DDIT3 predictive of PI3K-inhibition sensitivity. We observed similar trends in genotype-drug sensitivity associations in patient-derived xenograft mouse models. This work provides proof of principle that patient-specific single-cell measurements and modeling could inform effective precision medicine strategies.


Assuntos
Neoplasias da Mama , Preparações Farmacêuticas , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Feminino , Genômica , Humanos , Camundongos , Transdução de Sinais
18.
Genome Biol ; 21(1): 302, 2020 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-33317623

RESUMO

BACKGROUND: Tumor-specific genomic aberrations are routinely determined by high-throughput genomic measurements. It remains unclear how complex genome alterations affect molecular networks through changing protein levels and consequently biochemical states of tumor tissues. RESULTS: Here, we investigate the propagation of genomic effects along the axis of gene expression during prostate cancer progression. We quantify genomic, transcriptomic, and proteomic alterations based on 105 prostate samples, consisting of benign prostatic hyperplasia regions and malignant tumors, from 39 prostate cancer patients. Our analysis reveals the convergent effects of distinct copy number alterations impacting on common downstream proteins, which are important for establishing the tumor phenotype. We devise a network-based approach that integrates perturbations across different molecular layers, which identifies a sub-network consisting of nine genes whose joint activity positively correlates with increasingly aggressive tumor phenotypes and is predictive of recurrence-free survival. Further, our data reveal a wide spectrum of intra-patient network effects, ranging from similar to very distinct alterations on different molecular layers. CONCLUSIONS: This study uncovers molecular networks with considerable convergent alterations across tumor sites and patients. It also exposes a diversity of network effects: we could not identify a single sub-network that is perturbed in all high-grade tumor regions.


Assuntos
Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Neoplasias da Próstata/genética , Biomarcadores Tumorais/genética , Variações do Número de Cópias de DNA , Heterogeneidade Genética , Genômica , Humanos , Masculino , Mutação , Fenótipo , Próstata/patologia , Proteogenômica , Proteoma , Proteômica , RNA Mensageiro , Transcriptoma
19.
ACS Appl Mater Interfaces ; 12(51): 57047-57054, 2020 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-33296166

RESUMO

The thermal stability of cathode active materials (CAMs) is of major importance for the safety of lithium-ion batteries (LIBs). A thorough understanding of how commercially viable layered oxide CAMs behave at the atomic length scale upon heating is indispensable for the further development of LIBs. Here, structural changes of Li(Ni0.85Co0.15Mn0.05)O2 (NCM851005) at elevated temperatures are studied by in situ aberration-corrected scanning transmission electron microscopy (AC-STEM). Heating NCM851005 inside the microscope under vacuum conditions enables us to observe phase transitions and other structural changes at high spatial resolutions. This has been primarily possible by establishing low-dose electron beam conditions in STEM. Specific focus is put on the evolution of inherent nanopore defects found in the primary grains, which are believed to play an important role in LIB degradation. The onset temperature of structural changes is found to be ∼175 °C, resulting in phase transformation from a layered to a rock-salt-like structure, especially at the internal interfaces, and increasing intragrain inhomogeneity. The reducing environment and heat application lead to the formation and subsequent densification of {003}- and {014}-type facets. In the light of these results, postsynthesis electrode drying processes applied under reducing environment and heat, for example, in the preparation of solid-state batteries, should be re-examined carefully.

20.
Microcirculation ; 27(7): e12625, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32395853

RESUMO

OBJECTIVE: Treatment with BCR-ABL tyrosine kinase inhibitors (TKIs) is the standard of care for patients with chronic myeloid leukemia, however evidence indicates these compounds may have cardiovascular side-effects. This study sought to determine if ex vivo exposure of human adipose arterioles to the BCR-ABL TKIs imatinib and nilotinib causes endothelial dysfunction. METHODS: Human adipose arterioles were incubated overnight in cell culture media containing vehicle (PBS), imatinib (10 µmol/L) or nilotinib (100 µmol/L). Arterioles were cannulated onto glass pipettes and flow mediated dilation (FMD) was assessed via video microscopy. To determine the mechanism of vasodilation, FMD was re-assessed in the presence of either the nitric oxide synthase inhibitor L-NAME (100 µmol/L) or the H2 O2 scavenger PEG-Catalase (500 U/mL). RESULTS: Neither imatinib nor nilotinib affected the magnitude of FMD (max dilation = 78±17% vehicle, 80 ± 24% nilotinib, 73 ± 13% imatinib). FMD was decreased by L-NAME in vehicle-treated arterioles (max dilation = 47±29%). Conversely, L-NAME had no effect on FMD in imatinib- or nilotinib-treated vessels (max dilation = 79±14% and 80 ± 24%, respectively), rather FMD was inhibited by PEG-Catalase (max dilation = 29±11% and 29 ± 14%, respectively). CONCLUSION: Incubating human arterioles with imatinib or nilotinib switches the mediator of FMD from vasoprotective nitric oxide to pro-inflammatory H2 O2 .


Assuntos
Proteínas de Fusão bcr-abl/antagonistas & inibidores , Microvasos/efeitos dos fármacos , Microvasos/fisiologia , Inibidores de Proteínas Quinases/efeitos adversos , Vasodilatação/efeitos dos fármacos , Adulto , Idoso , Antineoplásicos/efeitos adversos , Arteríolas/efeitos dos fármacos , Arteríolas/fisiologia , Cardiotônicos/farmacologia , Catalase/farmacologia , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , Peróxido de Hidrogênio/metabolismo , Mesilato de Imatinib/efeitos adversos , Técnicas In Vitro , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Leucemia Mielogênica Crônica BCR-ABL Positiva/fisiopatologia , Masculino , Pessoa de Meia-Idade , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/antagonistas & inibidores , Polietilenoglicóis/farmacologia , Pirimidinas/efeitos adversos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA