Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
2.
J Allergy Clin Immunol ; 154(1): 209-221.e6, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38513838

ABSTRACT

BACKGROUND: Millions of people are exposed to landscape fire smoke (LFS) globally, and inhalation of LFS particulate matter (PM) is associated with poor respiratory and cardiovascular outcomes. However, how LFS affects respiratory and cardiovascular function is less well understood. OBJECTIVE: We aimed to characterize the pathophysiologic effects of representative LFS airway exposure on respiratory and cardiac function and on asthma outcomes. METHODS: LFS was generated using a customized combustion chamber. In 8-week-old female BALB/c mice, low (25 µg/m3, 24-hour equivalent) or moderate (100 µg/m3, 24-hour equivalent) concentrations of LFS PM (10 µm and below [PM10]) were administered daily for 3 (short-term) and 14 (long-term) days in the presence and absence of experimental asthma. Lung inflammation, gene expression, structural changes, and lung function were assessed. In 8-week-old male C57BL/6 mice, low concentrations of LFS PM10 were administered for 3 days. Cardiac function and gene expression were assessed. RESULTS: Short- and long-term LFS PM10 airway exposure increased airway hyperresponsiveness and induced steroid insensitivity in experimental asthma, independent of significant changes in airway inflammation. Long-term LFS PM10 airway exposure also decreased gas diffusion. Short-term LFS PM10 airway exposure decreased cardiac function and expression of gene changes relating to oxidative stress and cardiovascular pathologies. CONCLUSIONS: We characterized significant detrimental effects of physiologically relevant concentrations and durations of LFS PM10 airway exposure on lung and heart function. Our study provides a platform for assessment of mechanisms that underpin LFS PM10 airway exposure on respiratory and cardiovascular disease outcomes.


Subject(s)
Asthma , Mice, Inbred BALB C , Particulate Matter , Smoke , Animals , Female , Smoke/adverse effects , Asthma/physiopathology , Asthma/etiology , Male , Mice , Particulate Matter/adverse effects , Mice, Inbred C57BL , Lung/immunology , Lung/physiopathology , Wildfires , Disease Models, Animal
3.
Am J Physiol Heart Circ Physiol ; 326(5): H1219-H1251, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38363215

ABSTRACT

Sex-based differences in the development of obesity-induced cardiometabolic dysfunction are well documented, however, the specific mechanisms are not completely understood. Obesity has been linked to dysregulation of the epitranscriptome, but the role of N6-methyladenosine (m6A) RNA methylation has not been investigated in relation to the sex differences during obesity-induced cardiac dysfunction. In the current study, male and female C57BL/6J mice were subjected to short- and long-term high-fat/high-sucrose (HFHS) diet to induce obesogenic stress. Cardiac echocardiography showed males developed systolic and diastolic dysfunction after 4 mo of diet, but females maintained normal cardiac function despite both sexes being metabolically dysfunctional. Cardiac m6A machinery gene expression was differentially regulated by duration of HFHS diet in male, but not female mice, and left ventricular ejection fraction correlated with RNA machinery gene levels in a sex- and age-dependent manner. RNA-sequencing of cardiac transcriptome revealed that females, but not males may undergo protective cardiac remodeling early in the course of obesogenic stress. Taken together, our study demonstrates for the first time that cardiac RNA methylation machinery genes are regulated early during obesogenic stress in a sex-dependent manner and may play a role in the sex differences observed in cardiometabolic dysfunction.NEW & NOTEWORTHY Sex differences in obesity-associated cardiomyopathy are well documented but incompletely understood. We show for the first time that RNA methylation machinery genes may be regulated in response to obesogenic diet in a sex- and age-dependent manner and levels may correspond to cardiac systolic function. Our cardiac RNA-seq analysis suggests female, but not male mice may be protected from cardiac dysfunction by a protective cardiac remodeling response early during obesogenic stress.


Subject(s)
Adenosine/analogs & derivatives , Diet, High-Fat , Mice, Inbred C57BL , Obesity , Animals , Female , Male , Sex Factors , Obesity/metabolism , Obesity/genetics , Obesity/physiopathology , Ventricular Function, Left , Mice , Ventricular Remodeling , Adenosine/metabolism , Heart Diseases/metabolism , Heart Diseases/genetics , Heart Diseases/etiology , Heart Diseases/physiopathology , Time Factors , Disease Models, Animal , Myocardium/metabolism , Transcriptome , Ventricular Dysfunction, Left/physiopathology , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/etiology
4.
Heart Lung Circ ; 33(5): 747-752, 2024 May.
Article in English | MEDLINE | ID: mdl-38365500

ABSTRACT

BACKGROUND: Increased cancer survivorship represents a remarkable achievement for modern medicine. Unfortunately, cancer treatments have inadvertently contributed to cardiovascular (CV) damage, significantly threatening the health and quality of life of patients living with, through and beyond cancer. Without understanding the mechanisms, including whether the cardiotoxicity is due to the direct or indirect effects on cardiomyocytes, prevention and management of cardiotoxicity can pose challenges in many patients. To date, the cardiotoxicity profiles of most of the chemotherapy drugs are still poorly understood. AIM: To conduct a pilot study to investigate the direct effects of a range of cancer therapies on cardiomyocyte viability. METHODS: Primary human cardiomyocytes (HCM) were cultured and seeded into 96-well culture plates. A total of 35 different Food and Drug Administration-approved anti-cancer drugs were added to the HCM cells with a concentration of 1uM for 72 hours. The viability of HCMs was determined using CellTitre-Glo. The experiments were repeated at least three times for each drug with HCMs of different passages. RESULTS: We identified 15 anti-cancer agents that significantly reduced HCM viability. These drugs were: (1) anthracyclines (daunorubicin [HCM viability, mean %±standard error, 13.7±3.2%], epirubicin [47.6±5.3%]), (2) antimetabolite (azacitidine [67.1±2.4%]), (3) taxanes (paclitaxel [60.2±3.0%]), (4) protein kinase inhibitors (lapatinib [49.8±7.0%], ponatinib [42.4±9.0%], pemigatinib [68.1±2.3%], sorafenib [52.9±10.6%], nilotinib [64.4±4.5%], dasatinib [38.5±3.6%]), (5) proteasome inhibitors (ixazomib citrate [65.4±7.2%]), (6) non-selective histone-deacetylase inhibitor (panobinostat [19.1±4.1%]), poly adenosine diphosphate-ribose polymerase inhibitor (olaparib [68.2±1.7%]) and (7) vinca alkaloids (vincristine [44.6±7.4%], vinblastine [31.2±3.9%]). CONCLUSIONS: In total, 15 of the 35 commercially available anti-cancer drugs have direct cardiotoxic effects on HCM. Some of those, have not been associated with clinical cardiotoxicity, while others, known to be cardiotoxic do not appear to mediate it via direct effects on cardiomyocytes. More detailed investigations of the effects of cancer therapies on various cardiovascular cells should be performed to comprehensively determine the mechanisms of cardiotoxicity.


Subject(s)
Antineoplastic Agents , Cardiotoxicity , Cell Survival , Myocytes, Cardiac , Humans , Myocytes, Cardiac/drug effects , Antineoplastic Agents/toxicity , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Cardiotoxicity/etiology , Neoplasms/drug therapy , Cells, Cultured , Pilot Projects , Female
5.
J Cardiovasc Dev Dis ; 10(7)2023 Jun 28.
Article in English | MEDLINE | ID: mdl-37504530

ABSTRACT

Secreted frizzled-related protein 5 (SFRP5) is a novel anti-inflammatory adipokine that may play a role in cardiovascular development and disease. However, there is yet to be a comprehensive investigation into whether circulating SFRP5 can be a biomarker for cardiac function. Plasma SFRP5 levels were measured via ELISA in 262 patients admitted to a cardiology unit. Plasma SFRP5 levels were significantly lower in patients with a history of heart failure (HF), coronary artery disease (CAD), and atrial fibrillation (AF; p = 0.001). In univariate analyses, SFRP5 levels were also significantly positively correlated with left ventricular ejection fraction (LVEF) (r = 0.52, p < 0.001) and negatively correlated with E/E' (r = -0.30, p < 0.001). Patients with HF, CAD, low LVEF, low triglycerides, high CRP, and high eGFR were associated with lower SFRP5 levels independent of age, BMI, or diabetes after multivariate analysis (overall model r = 0.729, SE = 0.638). Our results show that low plasma SFRP5 levels are independently associated with the presence of HF, CAD, and, importantly, impaired LV function. These results suggest a potential role of SFRP5 as a biomarker, as well as a mediator of cardiac dysfunction independent of obesity and metabolic regulation.

6.
Antioxidants (Basel) ; 12(5)2023 May 22.
Article in English | MEDLINE | ID: mdl-37238003

ABSTRACT

Obesity is associated with significant metabolic co-morbidities, such as diabetes, hypertension, and dyslipidaemia, as well as a range of cardiovascular diseases, all of which lead to increased hospitalisations, morbidity, and mortality. Adipose tissue dysfunction caused by chronic nutrient stress can result in oxidative stress, mitochondrial dysfunction, inflammation, hypoxia, and insulin resistance. Thus, we hypothesised that reducing adipose tissue oxidative stress via adipose tissue-targeted overexpression of the antioxidant mitochondrial catalase (mCAT) may improve systemic metabolic function. We crossed mCAT (floxed) and Adipoq-Cre mice to generate mice overexpressing catalase with a mitochondrial targeting sequence predominantly in adipose tissue, designated AdipoQ-mCAT. Under normal diet conditions, the AdipoQ-mCAT transgenic mice demonstrated increased weight gain, adipocyte remodelling, and metabolic dysfunction compared to the wild-type mice. Under obesogenic dietary conditions (16 weeks of high fat/high sucrose feeding), the AdipoQ-mCAT mice did not result in incremental impairment of adipose structure and function but in fact, were protected from further metabolic impairment compared to the obese wild-type mice. While AdipoQ-mCAT overexpression was unable to improve systemic metabolic function per se, our results highlight the critical role of physiological H2O2 signalling in metabolism and adipose tissue function.

7.
J Clin Med ; 12(8)2023 Apr 09.
Article in English | MEDLINE | ID: mdl-37109127

ABSTRACT

The role of soluble suppression of tumorigenicity (sST2) as a biomarker in predicting clinical outcomes in patients with cardiovascular diseases (CVD) has not been fully elucidated. In this study, we sought to determine the relationship between sST2 levels and any unplanned hospital readmissions due to a major adverse cardiovascular event (MACE) within 1 year of first admission. Patients (n = 250) admitted to the cardiology unit at John Hunter Hospital were recruited. Occurrences of MACE, defined as the composite of total death, myocardial infarction (MI), stroke, readmissions for heart failure (HF), or coronary revascularization, were recorded after 30, 90, 180, and 365 days of first admission. On univariate analysis, patients with atrial fibrillation (AF) and HF had significantly higher sST2 levels vs. those who did not. Increasing levels of sST2 by quartiles were significantly associated with AF, HF, older age, low hemoglobin, low eGFR, and high CRP levels. On multivariate analysis: high sST2 levels and diabetes remained as risk predictors of any MACE occurrence; an sST2 level in the highest quartile (Q4: >28.4 ng/mL) was independently associated with older age, use of beta-blockers, and number of MACE events within a 1 year period. In this patient cohort, elevated sST2 levels are associated with unplanned hospital admission due to MACE within 1 year, independent of the nature of the index cardiovascular admission.

8.
Curr Heart Fail Rep ; 18(6): 362-377, 2021 12.
Article in English | MEDLINE | ID: mdl-34731413

ABSTRACT

PURPOSE OF REVIEW: Breast cancer survival rate has greatly improved in the last two decades due to the emergence of next-generation anti-cancer agents. However, cardiotoxicity remains a significant adverse effect arising from traditional and emerging chemotherapies as well as targeted therapies for breast cancer patients. In this review, we will discuss cardiotoxicities of both traditional and emerging therapies for breast cancer. We will discuss current practices to detect cardiotoxicity of these therapies with the focus on new and emerging biomarkers. We will then focus on 'omics approaches, especially the use of epigenetics to discover novel biomarkers and therapeutics to mitigate cardiotoxicity. RECENT FINDINGS: Significant cardiotoxicities of conventional chemotherapies remain and new and unpredictable new forms of cardiac and/or vascular toxicity emerge with the surge in novel and targeted therapies. Yet, there is no clear guidance on detection of cardiotoxicity, except for significant left ventricular systolic dysfunction, and even then, there is no uniform definition of what constitutes cardiotoxicity. The gold standard for detection of cardiotoxicity involves a serial echocardiography in conjunction with blood-based biomarkers to detect early subclinical cardiac dysfunction. However, the ability of these tests to detect early disease remains limited and not all forms of toxicity are detectable with these modalities. There is an unprecedented need to discover novel biomarkers that are sensitive and specific for early detection of subclinical cardiotoxicity. In that space, novel echocardiographic techniques, such as strain, are becoming more common-place and new biomarkers, discovered by epigenetic approaches, seem to become promising alternatives or adjuncts to conventional non-specific cardiac biomarkers.


Subject(s)
Breast Neoplasms , Cancer Survivors , Heart Failure , Biomarkers , Breast Neoplasms/drug therapy , Early Detection of Cancer , Female , Heart Failure/diagnosis , Humans
9.
Nurs Manag (Harrow) ; 26(5): 24-27, 2019 Oct 01.
Article in English | MEDLINE | ID: mdl-31468916

ABSTRACT

Advanced clinical practice (ACP) roles evolved to fill gaps in healthcare provision and improve consistency in standards of care. Historically, variance in service specific requirements, combined with inconsistent approaches to role governance and education, resulted in a multitude of ACP roles and remits. Definitions of roles and titles vary, but there is agreement on academic level and role competency assessment. Irrespective of these definitions, staff and services need support to operationalise guidance. In Scotland, development of advanced practice roles is supported through regional NHS Advanced Practice Academies. One Scottish Board has initiated a local board level advanced care academy to fulfil the three key components of advanced practitioner development: master's level theoretical knowledge, effective supervision and robust clinical competency assessment in practice. This development model, known as the 'triangle of capability', has allowed rapid progress with the advanced practice agenda, which is a central component of sustainable healthcare provision.

10.
Mol Carcinog ; 57(2): 284-294, 2018 02.
Article in English | MEDLINE | ID: mdl-29068469

ABSTRACT

Past studies have shown that mutant KRAS colon cancer cells are susceptible to apoptosis induced by the HSP90 inhibitor AUY922. Nevertheless, intrinsic and acquired resistance remains an obstacle for the potential application of the inhibitor in the treatment of the disease. Here we report that Mcl-1 is important for survival of colon cancer cells in the presence of AUY922. Mcl-1 was upregulated in mutant KRAS colon cancer cells selected for resistance to AUY922-induced apoptosis. This was due to its increased stability mediated by Bcl-2-associated athanogene domain 3 (BAG3), which was also increased in resistant colon cancer cells by heat shock factor 1 (HSF1) as a result of chronic endoplasmic reticulum (ER) stress. Functional investigations demonstrated that inhibition of Mcl-1, BAG3, or HSF1 triggered apoptosis in resistant colon cancer cells, and rendered AUY922-naïve colon cancer cells more sensitive to the inhibitor. Together, these results identify that the HSF1-BAG3-Mcl-1 signal axis is critical for protection of mutant KRAS colon cancer cells from AUY922-induced apoptosis, with potential implications for targeting HSF1/BAG3/Mcl-1 to improve the efficacy of AUY922 in the treatment of colon cancer.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Isoxazoles/pharmacology , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Resorcinols/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm , Endoplasmic Reticulum Stress/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Heat Shock Transcription Factors/metabolism , Humans , Proto-Oncogene Proteins c-bcl-2/metabolism
11.
J Invest Dermatol ; 138(4): 922-932, 2018 04.
Article in English | MEDLINE | ID: mdl-29174371

ABSTRACT

Although the expression of programmed death-ligand 1 (PD-L1) is an important mechanism by which cancer cells evade the immune system, PD-L1 expression in cancer cells is commonly associated with patients' responses to treatment with anti-programmed death 1/PD-L1 antibodies. However, how PD-L1 expression is regulated in melanoma cells remains to be fully elucidated. Here we report that the class I histone deacetylase (HDAC) HDAC8 controls transcriptional activation of PD-L1 by a transcription complex consisting of transcription factors homeobox A5 and signal transducer and activator of transcription 3. Inhibition of HDAC8 upregulated PD-L1 in melanoma cells. This was due to an increase in the activity of a fragment of the PD-L1 gene promoter that is enriched with binding sites for both homeobox A5 and signal transducer and activator of transcription 3. Indeed, knockdown of homeobox A5 or signal transducer and activator of transcription 3 abolished upregulation of PD-L1 by HDAC8 inhibition. Moreover, homeobox A5 and signal transducer and activator of transcription 3 were physically associated and appeared interdependent in activating PD-L1 transcription. Functional studies showed that HDAC8-mediated regulation of PD-L1 expression participated in modulating anti-melanoma T-cell responses. Collectively, these results identify HDAC8 as an important epigenetic regulator of PD-L1 expression, with implications for better understanding of the interaction between melanoma cells and the immune system.


Subject(s)
B7-H1 Antigen/genetics , Gene Expression Regulation, Neoplastic , Histone Deacetylases/genetics , Homeodomain Proteins/genetics , Melanoma/genetics , Repressor Proteins/genetics , STAT3 Transcription Factor/genetics , B7-H1 Antigen/biosynthesis , Cell Line, Tumor , DNA, Neoplasm/genetics , Histone Deacetylases/biosynthesis , Homeodomain Proteins/biosynthesis , Humans , Melanoma/metabolism , Melanoma/pathology , Repressor Proteins/biosynthesis , STAT3 Transcription Factor/biosynthesis , Signal Transduction , Transcription Factors , Transcriptional Activation , Up-Regulation
12.
Biochem Biophys Res Commun ; 485(1): 47-53, 2017 03 25.
Article in English | MEDLINE | ID: mdl-28189677

ABSTRACT

The 4-phosphatase Inositol polyphosphate 4-phosphatase II (INPP4B) is a regulator of the PI3K signalling pathway and functions to suppress or promote activation of downstream kinases depending on cell type and context. Here we report the identification of a novel small transcript variant of INPP4B (INPP4B-S) that has a role in promoting proliferation of colon and breast cancer cells. INPP4B-S differed from full length INPP4B (INPP4B-FL) by the insertion of a small exon between exons 15 and 16 and the deletion of exons 20-24. Nevertheless, INPP4B-S retained all the functional domains of INPP4B-FL and was similarly located to the cytoplasm. Overexpression of INPP4B-S increased, whereas selective knockdown of INPP4B-S reduced the rate of proliferation in HCT116 and MCF-7 cells. These results warrant further investigation of the role INPP4B-S in activation of downstream kinases and in regulation of cancer pathogenesis.


Subject(s)
Alternative Splicing , Breast Neoplasms/genetics , Colonic Neoplasms/genetics , Phosphoric Monoester Hydrolases/genetics , Base Sequence , Breast/metabolism , Breast/pathology , Breast Neoplasms/pathology , Cell Proliferation , Colon/metabolism , Colon/pathology , Colonic Neoplasms/pathology , Exons , Female , HCT116 Cells , Humans , MCF-7 Cells , Phosphoric Monoester Hydrolases/analysis , Protein Isoforms/genetics , Transcription, Genetic
13.
J Invest Dermatol ; 136(11): 2277-2286, 2016 11.
Article in English | MEDLINE | ID: mdl-27427486

ABSTRACT

The effect of MTH1 inhibition on cancer cell survival has been elusive. Here we report that although silencing of MTH1 does not affect survival of melanoma cells, TH588, one of the first-in-class MTH1 inhibitors, kills melanoma cells through apoptosis independently of its inhibitory effect on MTH1. Induction of apoptosis by TH588 was not alleviated by MTH1 overexpression or introduction of the bacterial homolog of MTH1 that has 8-oxodGTPase activity but cannot be inhibited by TH588, indicating that MTH1 inhibition is not the cause of TH588-induced killing of melanoma cells. Although knockdown of MTH1 did not impinge on the viability of melanoma cells, it rendered melanoma cells sensitive to apoptosis induced by the oxidative stress inducer elesclomol. Of note, treatment with elesclomol also enhanced TH588-induced apoptosis, whereas a reactive oxygen species scavenger or an antioxidant attenuated the apoptosis triggered by TH588. Indeed, the sensitivity of melanoma cells to TH588 was correlated with endogenous levels of reactive oxygen species. Collectively, these results indicate that the cytotoxicity of TH588 toward melanoma cells is not associated with its inhibitory effect on MTH1, although it is mediated by cellular production of ROS.


Subject(s)
Apoptosis/drug effects , Melanoma/drug therapy , Oxidative Stress , Pyrimidines/pharmacology , Reactive Oxygen Species/metabolism , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Humans , Melanoma/metabolism , Melanoma/pathology
14.
Oncotarget ; 6(37): 39891-907, 2015 Nov 24.
Article in English | MEDLINE | ID: mdl-26573229

ABSTRACT

Inositol polyphosphate 4-phosphatase type II (INPP4B) negatively regulates PI3K/Akt signalling and has a tumour suppressive role in some types of cancers. However, we have found that it is upregulated in a subset of melanomas. Here we report that INPP4B can function as an oncogenic driver through activation of serum- and glucocorticoid-regulated kinase 3 (SGK3) in melanoma. While INPP4B knockdown inhibited melanoma cell proliferation and retarded melanoma xenograft growth, overexpression of INPP4B enhanced melanoma cell and melanocyte proliferation and triggered anchorage-independent growth of melanocytes. Noticeably, INPP4B-mediated melanoma cell proliferation was not related to activation of Akt, but was mediated by SGK3. Upregulation of INPP4B in melanoma cells was associated with loss of miRNA (miR)-494 and/or miR-599 due to gene copy number reduction. Indeed, overexpression of miR-494 or miR-599 downregulated INPP4B, reduced SGK3 activation, and inhibited melanoma cell proliferation, whereas introduction of anti-miR-494 or anti-miR-599 upregulated INPP4B, enhanced SGK3 activation, and promoted melanoma cell proliferation. Collectively, these results identify upregulation of INPP4B as an oncogenic mechanism through activation of SGK3 in a subset of melanomas, with implications for targeting INPP4B and restoring miR-494 and miR-599 as novel approaches in the treatment of melanomas with high INPP4B expression.


Subject(s)
Melanoma/metabolism , Phosphoric Monoester Hydrolases/metabolism , Protein Serine-Threonine Kinases/metabolism , Up-Regulation , Animals , Cell Line , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Humans , Immunoblotting , Immunohistochemistry , Male , Melanoma/genetics , Melanoma/pathology , Mice, Nude , MicroRNAs/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoric Monoester Hydrolases/genetics , Protein Serine-Threonine Kinases/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Transplantation, Heterologous , Tumor Burden/genetics
15.
Rural Remote Health ; 15(4): 3276, 2015.
Article in English | MEDLINE | ID: mdl-26446199

ABSTRACT

INTRODUCTION: The Rural Clinical Training and Support (RCTS) program is an Australian Government initiative to address the shortage of medical practitioners within rural and remote Australia. There is a large amount of published information about the RCTS program and rural medical student cohorts who have undertaken short- and long-term rotations. However, very little is known about the academic and professional staff involved in the program, a knowledge gap that may impact workforce and succession planning. To address this, the Federation of Rural Australian Medical Educators (FRAME) initiated the pilot 2014 RCTS Snapshot survey to obtain data on the current RCTS workforce. METHODS: All professional, academic and clinical academic staff (fixed-term and continuing, regardless of fraction) employed through the RCTS program were invited to complete a short, web-based survey. The survey was conducted from March to June 2014. The quantitative variables in the survey included demographics (age and gender), rural background and exposure, employment history in rural/regional areas and at rural clinical schools (RCS), experience and expertise, reasons for working at RCS, and future employment intentions. The last three questions also were of a qualitative open-ended format to allow respondents to provide additional details regarding their reasons for working at RCSs and their future intentions. RESULTS: The estimated total RCTS workforce was 970. A total of 413 responses were received and 316 (40.9%) complete responses analysed. The majority of respondents were female (71%), the 40-60-year age group was predominant (28%), and professional staff constituted the majority (62%). The below 40-year age group had more professionals than academics (21% vs 12%) and more than 62% of academics were aged above 50 years. Notably, there were no academics aged less than 30 years. The percentage of professional staff with a rural background was higher (62%) than that of academics with a rural background (42%). However, more than 70% of academics had previous exposure to a rural area as an adult and 32% had an exposure as a part of university or the TAFE (technical and further education) system. More than half (62%) of RCTS academics were aged more than 50 years and thus approaching retirement age. The implementation of a FRAME-sponsored leadership and succession program was considered by most staff (84%) as one strategy that could be used to prevent a future shortage of academics. Lifestyle reasons for working at an RCS were common to both academic (54%) and professional (63%) staff. A passion for rural health and building capacity within the rural health workforce were other central themes to emerge from the qualitative data. Uncertainty around contract renewal and future funding were dominant themes to emerge from respondents regarding their future employment intentions within the RCTS program. CONCLUSIONS: This study has provided valuable insights into the professional and academic staff's views and aspirations about the RCTS program. These data on the current RCTS workforce provide a benchmark to which future surveys of the workforce can be compared to monitor trends in turnover or predict future shortages due to cohort ageing.


Subject(s)
Career Choice , Education, Medical, Undergraduate/organization & administration , Faculty, Medical/organization & administration , Rural Health Services , Schools, Medical/organization & administration , Adult , Australia , Education, Medical, Graduate/organization & administration , Female , Health Workforce , Humans , Job Satisfaction , Male , Middle Aged , Pilot Projects , Program Evaluation , Qualitative Research , Students, Medical/statistics & numerical data , Surveys and Questionnaires
16.
Cancer Res ; 75(8): 1736-48, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25724678

ABSTRACT

Although many studies have uncovered an important role for the receptor-binding protein kinase RIP1 in controlling cell death signaling, its possible contributions to cancer pathogenesis have been little explored. Here, we report that RIP1 functions as an oncogenic driver in human melanoma. Although RIP1 was commonly upregulated in melanoma, RIP1 silencing inhibited melanoma cell proliferation in vitro and retarded the growth of melanoma xenografts in vivo. Conversely, while inducing apoptosis in a small proportion of melanoma cells, RIP1 overexpression enhanced proliferation in the remaining cells. Mechanistic investigations revealed that the proliferative effects of RIP1 overexpression were mediated by NF-κB activation. Strikingly, ectopic expression of RIP1 enhanced the proliferation of primary melanocytes, triggering their anchorage-independent cell growth in an NF-κB-dependent manner. We identified DNA copy-number gain and constitutive ubiquitination by a TNFα autocrine loop mechanism as two mechanisms of RIP1 upregulation in human melanomas. Collectively, our findings define RIP1 as an oncogenic driver in melanoma, with potential implications for targeting its NF-κB-dependent activation mechanism as a novel approach to treat this disease.


Subject(s)
Cell Transformation, Neoplastic/genetics , Melanoma/genetics , Oncogenes , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Skin Neoplasms/genetics , Animals , Cells, Cultured , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Melanocytes/metabolism , Melanocytes/pathology , Melanoma/pathology , Mice , Mice, Nude , NF-kappa B/genetics , NF-kappa B/metabolism , RNA Interference , RNA, Small Interfering/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Skin Neoplasms/pathology , Xenograft Model Antitumor Assays
17.
J Invest Dermatol ; 134(2): 488-497, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23921951

ABSTRACT

Cancer cells commonly undergo chronic endoplasmic reticulum (ER) stress, to which the cells have to adapt for survival and proliferation. We report here that in melanoma cells intrinsic activation of the ER stress response/unfolded protein response (UPR) is, at least in part, caused by increased outputs of protein synthesis driven by oncogenic activation of mitogen-activated protein kinase kinase/extracellular signal-regulated kinase (MEK/ERK) and promotes proliferation and protects against apoptosis induced by acute ER stress. Inhibition of oncogenic BRAF(V600E) or MEK-attenuated activation of inositol-requiring enzyme 1 (IRE1) and activating transcription factor 6 (ATF6) signaling of the UPR in melanoma cells. This was associated with decreased phosphorylation of eukaryotic initiation factor 4E (eIF4E) and nascent protein synthesis and was recapitulated by knockdown of eIF4E. In line with this, introduction of BRAF(V600E) into melanocytes led to increases in eIF4E phosphorylation and protein production and triggered activation of the UPR. Similar to knockdown of glucose-regulated protein 78 (GRP78), inhibition of XBP1 decelerated melanoma cell proliferation and enhanced apoptosis induced by the pharmacological ER stress inducers tunicamycin and thapasigargin. Collectively, these results reveal that potentiation of adaptation to chronic ER stress is another mechanism by which oncogenic activation of the MEK/ERK pathway promotes the pathogenesis of melanoma.


Subject(s)
Adaptation, Physiological/physiology , Endoplasmic Reticulum Stress/physiology , MAP Kinase Signaling System/physiology , Melanoma/metabolism , Skin Neoplasms/metabolism , Activating Transcription Factor 6/metabolism , Apoptosis/physiology , Cell Line, Tumor , Cell Survival/physiology , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum Chaperone BiP , Endoribonucleases/metabolism , Heat-Shock Proteins/metabolism , Humans , Indoles/pharmacology , MAP Kinase Signaling System/drug effects , Melanocytes/metabolism , Melanocytes/pathology , Melanoma/pathology , Membrane Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/metabolism , Regulatory Factor X Transcription Factors , Skin Neoplasms/pathology , Sulfonamides/pharmacology , Transcription Factors/metabolism , X-Box Binding Protein 1
18.
Cell Signal ; 26(2): 287-94, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24240056

ABSTRACT

Apoptosis triggered by endoplasmic reticulum (ER) stress is associated with rapid attenuation of the IRE1α and ATF6 pathways but persistent activation of the PERK branch of the unfolded protein response (UPR) in cells. However, melanoma cells are largely resistant to ER stress-induced apoptosis, suggesting that the kinetics and durations of activation of the UPR pathways are deregulated in melanoma cells undergoing ER stress. We show here that the IRE1α and ATF6 pathways are sustained along with the PERK signaling in melanoma cells subjected to pharmacological ER stress, and that this is, at least in part, due to increased activation of the MEK/ERK pathway. In contrast to an initial increase followed by rapid reduction in activation of IRE1α and ATF6 signaling in control cells that were relatively sensitive to ER stress-induced apoptosis, activation of IRE1α and ATF6 by the pharmacological ER stress inducer tunicamycin (TM) or thapsigargin (TG) persisted in melanoma cells. On the other hand, the increase in PERK signaling lasted similarly in both types of cells. Sustained activation of IRE1α and ATF6 signaling played an important role in protecting melanoma cells from ER stress-induced apoptosis, as interruption of IRE1α or ATF6 rendered melanoma cells sensitive to apoptosis induced by TM or TG. Inhibition of MEK partially blocked IRE1α and ATF6 activation, suggesting that MEK/ERK signaling contributed to sustained activation of IRE1α and ATF6. Taken together, these results identify sustained activation of the IRE1α and ATF6 pathways of the UPR driven by the MEK/ERK pathway as an important protective mechanism against ER stress-induced apoptosis in melanoma cells.


Subject(s)
Activating Transcription Factor 6/metabolism , Endoplasmic Reticulum Stress , Endoribonucleases/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Activating Transcription Factor 6/antagonists & inhibitors , Activating Transcription Factor 6/genetics , Cell Line, Tumor , Endoplasmic Reticulum Stress/drug effects , Endoribonucleases/antagonists & inhibitors , Endoribonucleases/genetics , Eukaryotic Initiation Factor-2/antagonists & inhibitors , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , HEK293 Cells , Humans , Melanoma/metabolism , Melanoma/pathology , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Thapsigargin/toxicity , Tunicamycin/toxicity , Unfolded Protein Response , eIF-2 Kinase/metabolism
19.
PLoS One ; 8(12): e84073, 2013.
Article in English | MEDLINE | ID: mdl-24367627

ABSTRACT

Anti-apoptotic Bcl-2 family proteins, in particular, Mcl-1, are known to play a critical role in resistance of human melanoma cells to induction of apoptosis by endoplasmic reticulum stress and other agents. The present study examined whether the BH3 mimetics, Obatoclax and ABT-737, which inhibit multiple anti-apoptotic Bcl-2 family proteins, would overcome resistance to apoptosis. We report that both agents induced a strong unfolded protein response (UPR) and that RNAi knockdown of UPR signalling proteins ATF6, IRE1α and XBP-1 inhibited Mcl-1 upregulation and increased sensitivity to the agents. These results demonstrate that inhibition of anti-apoptotic Bcl-2 proteins by Obatoclax and ABT-737 appears to elicit a protective feedback response in melanoma cells, by upregulation of Mcl-1 via induction of the UPR. We also report that Obatoclax, but not ABT-737, strongly induces autophagy, which appears to play a role in determining melanoma sensitivity to the agents.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Biphenyl Compounds/pharmacology , Endoplasmic Reticulum Stress/drug effects , Melanoma/pathology , Nitrophenols/pharmacology , Pyrroles/pharmacology , Sulfonamides/pharmacology , Autophagy/drug effects , Calcium/metabolism , Cell Line, Tumor , Cytosol/drug effects , Cytosol/metabolism , Drug Resistance, Neoplasm/drug effects , Humans , Indoles , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Piperazines/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , Unfolded Protein Response/drug effects , Up-Regulation/drug effects
20.
Med J Aust ; 199(11): 779-82, 2013 Dec 16.
Article in English | MEDLINE | ID: mdl-24329657

ABSTRACT

OBJECTIVES: To determine whether recruitment of rural students and uptake of extended rural placements are associated with students' expressed intentions to undertake rural internships and students' acceptance of rural internships after finishing medical school, and to compare any associations. DESIGN, SETTING AND PARTICIPANTS: Longitudinal study of three successive cohorts (commencing 2005, 2006, 2007) of medical students in the Sydney Medical Program (SMP), University of Sydney, New South Wales, using responses to self-administered questionnaires upon entry to and exit from the Sydney Medical School and data recorded in rolls. MAIN OUTCOME MEASURES: Students' expressed intentions to undertake rural internships, and their acceptance of rural internships after finishing medical school. RESULTS: Data from 448 students were included. The proportion of students preferring a rural career dropped from 20.7% (79/382) to 12.5% (54/433) between entry into and exit from the SMP. A total of 98 students took extended rural placements. Ultimately, 8.1% (35/434) accepted a rural internship, although 14.5% (60/415) had indicated a first preference for a rural post. Students who had undertaken an extended rural placement were more than three times as likely as those with rural backgrounds to express a first preference for a rural internship (23.9% v 7.7%; χ(2) = 7.04; P = 0.008) and more than twice as likely to accept a rural internship (21.3% v 9.9%; χ(2) = 3.85; P = 0.05). CONCLUSION: For the three cohorts studied, rural clinical training through extended placements in rural clinical schools had a stronger association than rural background with a preference for, and acceptance of, rural internship.


Subject(s)
Career Choice , Education, Medical, Undergraduate , Internship and Residency/statistics & numerical data , Medically Underserved Area , Rural Health Services , Students, Medical/psychology , Education, Medical, Undergraduate/methods , Education, Medical, Undergraduate/statistics & numerical data , Humans , Intention , Longitudinal Studies , New South Wales , Rural Population , School Admission Criteria , Surveys and Questionnaires , Workforce
SELECTION OF CITATIONS
SEARCH DETAIL
...