53 results on '"Kristin A Waite"'
Search Results
2. The MIF promoter SNP rs755622 is associated with immune activation in glioblastoma
- Author
-
Tyler J. Alban, Matthew M. Grabowski, Balint Otvos, Defne Bayik, Wesley Wang, Ajay Zalavadia, Vlad Makarov, Katie Troike, Mary McGraw, Anja Rabljenovic, Adam Lauko, Chase Neumann, Gustavo Roversi, Kristin A. Waite, Gino Cioffi, Nirav Patil, Thuy T. Tran, Kathleen McCortney, Alicia Steffens, C. Marcela Diaz, J. Mark Brown, Kathleen M. Egan, Craig M. Horbinski, Jill S. Barnholtz-Sloan, Prajwal Rajappa, Michael A. Vogelbaum, Richard Bucala, Timothy A. Chan, Manmeet S. Ahluwalia, and Justin D. Lathia
- Subjects
Immunology ,Oncology ,Medicine - Abstract
Intratumoral heterogeneity is a defining hallmark of glioblastoma, driving drug resistance and ultimately recurrence. Many somatic drivers of microenvironmental change have been shown to affect this heterogeneity and, ultimately, the treatment response. However, little is known about how germline mutations affect the tumoral microenvironment. Here, we find that the single-nucleotide polymorphism (SNP) rs755622 in the promoter of the cytokine macrophage migration inhibitory factor (MIF) is associated with increased leukocyte infiltration in glioblastoma. Furthermore, we identified an association between rs755622 and lactotransferrin expression, which could also be used as a biomarker for immune-infiltrated tumors. These findings demonstrate that a germline SNP in the promoter region of MIF may affect the immune microenvironment and further reveal a link between lactotransferrin and immune activation.
- Published
- 2023
- Full Text
- View/download PDF
3. Mortality trends in primary malignant brain and central nervous system tumors vary by histopathology, age, race, and sex
- Author
-
Marisa Thierheimer, Gino Cioffi, Kristin A. Waite, Carol Kruchko, Quinn T. Ostrom, and Jill S. Barnholtz-Sloan
- Subjects
Cancer Research ,Neurology ,Oncology ,Neurology (clinical) - Abstract
Purpose Primary malignant brain and other central nervous system tumors are rare cancers that have shown rising mortality rates in recent years. To elucidate potential factors involved in this rising death rate, we examined mortality trends for primary malignant BT in the United States stratified by histopathology groupings, age, race, and sex. Methods Mortality rates for demographic factors within primary malignant BT were generated using the National Center for Health Statistics' National Vital Statistics Systems data from 2004 to 2018. Additionally, histopathology-specific incidence-based mortality rates were calculated using the National Cancer Institute’s Surveillance, Epidemiology, and End-Results (SEER) 18 data from 2004 to 2018. Joinpoint modeling was used to estimate mortality trends and annual percent changes with corresponding 95% confidence intervals. Results Overall, there was a very small increase in mortality from 2004 to 2018. Individuals > 65 years saw a small increase in mortality, while changes in individuals of other ages were non-significant. Asian/Pacific Islander or American Indian/Alaskan Native had the largest increase in mortality. Among histopathology groupings, there was a small mortality increase in adults ages > 65 years with glioblastoma, while the mortality rate of other malignant gliomas declined in the same age group. CNS lymphoma mortality rates in patients ages 15–39 and 40–64 years declined significantly while rising significantly in the > 65 age group. In pediatric patients, embryonal tumor mortality had a non-significant increase between 2004 and 2007 but declined significantly between 2007 and 2018. Conclusion Examining age, race, sex, and histopathology-specific mortality trends at the population level can provide important information for clinicians, researchers, and aid in public health planning.
- Published
- 2023
4. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2015–2019
- Author
-
Quinn T Ostrom, Mackenzie Price, Corey Neff, Gino Cioffi, Kristin A Waite, Carol Kruchko, and Jill S Barnholtz-Sloan
- Subjects
Adult ,Male ,Cancer Research ,Adolescent ,Brain Neoplasms ,Incidence ,Infant, Newborn ,Brain ,Infant ,United States ,Central Nervous System Neoplasms ,Young Adult ,Oncology ,Child, Preschool ,Meningeal Neoplasms ,Humans ,Female ,Registries ,Neurology (clinical) ,Child ,Glioblastoma ,Meningioma - Abstract
The Central Brain Tumor Registry of the United States (CBTRUS), in collaboration with the Centers for Disease Control and Prevention and the National Cancer Institute, is the largest population-based registry focused exclusively on primary brain and other central nervous system (CNS) tumors in the United States (US) and represents the entire US population. This report contains the most up-to-date population-based data on primary brain tumors available and supersedes all previous reports in terms of completeness and accuracy. All rates are age-adjusted using the 2000 US standard population and presented per 100,000 population. The average annual age-adjusted incidence rate (AAAIR) of all malignant and non-malignant brain and other CNS tumors was 24.71 per 100,000 population (malignant AAAIR=7.02 and non-malignant AAAIR=17.69). This overall rate was higher in females compared to males (27.62 versus 21.60 per 100,000) and non-Hispanic persons compared to Hispanic persons (25.09 versus 22.95 per 100,000). The most commonly occurring malignant brain and other CNS histopathology was glioblastoma (14.2% of all tumors and 50.1% of all malignant tumors), and the most common non-malignant histopathology was meningioma (39.7% of all tumors and 55.4% of all non-malignant tumors). Glioblastoma was more common in males, and meningiomas were more common in females. In children and adolescents (ages 0-19 years), the incidence rate of all primary brain and other CNS tumors was 6.20 per 100,000 population. An estimated 93,470 new cases of malignant and non-malignant brain and other CNS tumors are expected to be diagnosed in the US population in 2022 (26,670 malignant and 66,806 non-malignant). There were 84,264 deaths attributed to malignant brain and other CNS tumors between 2015 and 2019. This represents an average annual mortality rate of 4.41 per 100,000 population and an average of 16,853 deaths per year. The five-year relative survival rate following diagnosis of a malignant brain and other CNS tumor was 35.7%, while for non-malignant brain and other CNS tumors the five-year relative survival rate was 91.8%.
- Published
- 2022
5. CBTRUS Statistical Report: Pediatric Brain Tumor Foundation Childhood and Adolescent Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2014–2018
- Author
-
Quinn T Ostrom, Mackenzie Price, Katherine Ryan, Jacob Edelson, Corey Neff, Gino Cioffi, Kristin A Waite, Carol Kruchko, and Jill S Barnholtz-Sloan
- Subjects
Adult ,Cancer Research ,Adolescent ,Brain Neoplasms ,Incidence ,Infant, Newborn ,Brain ,Infant ,United States ,Central Nervous System Neoplasms ,Young Adult ,Oncology ,Child, Preschool ,Humans ,Registries ,Neurology (clinical) ,Child - Abstract
The CBTRUS Statistical Report: Pediatric Brain Tumor Foundation Childhood and Adolescent Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2014–2018 comprehensively describes the current population-based incidence of primary malignant and non-malignant brain and other CNS tumors in children and adolescents ages 0–19 years, collected and reported by central cancer registries covering approximately 100% of the United States population. Overall, brain and other CNS tumors are the most common solid tumor, the most common cancer, and the most common cause of cancer death in children and adolescents ages 0–19 years. This report aims to serve as a useful resource for researchers, clinicians, patients, and families.
- Published
- 2022
6. Incidence and survival of choroid plexus tumors in the United States
- Author
-
Kailey Takaoka, Gino Cioffi, Kristin A Waite, Jonathan L Finlay, Daniel Landi, Kaitlyn Greppin, Carol Kruchko, Quinn T Ostrom, and Jill S Barnholtz-Sloan
- Subjects
Medicine (miscellaneous) - Abstract
Background There are limited data available on incidence and survival of patients with choroid plexus tumors (CPT). This study provides the most current epidemiological analysis of choroid plexus tumors from 2004 to 2017 in the United States. Methods Data on 2013 patients with CPT were acquired from the Central Brain Tumor Registry of the United States in collaboration with the Centers for Disease Control and Prevention (CDC) and the National Cancer Institute, from 2004 to 2017. CPT cases were classified by the following pathological subtypes: choroid plexus papilloma (CPP), atypical choroid plexus papilloma (aCPP), and choroid plexus carcinoma (CPC). Frequencies and age-adjusted incidence rates (AAIR) per 100 000 and rate ratios per 100 000 (IRR) were reported for age, sex, race, and ethnicity for each pathological subtype with 95% confidence intervals (95% CI). Using CDC’s National Program of Cancer Registries survival database, survival curves and hazard ratios (HRs) evaluated overall survival from 2001 to 2016. Results CPP had the highest overall incidence (AAIR: 0.034, 95% CI: 0.033–0.036), followed by CPC (AAIR: 0.008, 95% CI: 0.008–0.009) and aCPP (AAIR: 0.005, 95% CI: 0.005–0.006). Incidence was highest among children less than one year old among all subtypes (CPP AAIR: 0.278; aCPP AAIR: 0.140; CPC AAIR: 0.195), reducing as patients aged. Overall survival was worse among patients with CPC, being five times more likely to die compared to patients with CPP (HR: 5.23, 95% CI: 4.05–7.54, P Conclusions This analysis is the most current and comprehensive study in the US on the incidence and survival for CPT. Population based statistics provide critical information in understanding disease characteristics, which impact patient care and prognosis.
- Published
- 2022
7. Complete prevalence of primary malignant and non‐malignant brain tumors in comparison to other cancers in the United States
- Author
-
Corey Neff, Mackenzie Price, Gino Cioffi, Carol Kruchko, Kristin A. Waite, Jill S. Barnholtz‐Sloan, and Quinn T. Ostrom
- Subjects
Cancer Research ,Oncology - Published
- 2023
8. Molecular biomarker-defined brain tumors: Epidemiology, validity, and completeness in the United States
- Author
-
J Bryan Iorgulescu, Chuxuan Sun, Corey Neff, Gino Cioffi, Catherine Gutierrez, Carol Kruchko, Jennifer Ruhl, Kristin A Waite, Serban Negoita, Jim Hofferkamp, Tarik Tihan, Roger McLendon, Daniel J Brat, Quinn T Ostrom, and Jill S Barnholtz-Sloan
- Subjects
Cancer Research ,Adolescent ,Epidemiology ,Brain Neoplasms ,Glioma ,Astrocytoma ,Corrigenda ,United States ,Isocitrate Dehydrogenase ,Young Adult ,Oncology ,Mutation ,Humans ,Neurology (clinical) ,Child ,Glioblastoma ,Biomarkers - Abstract
Background Selected molecular biomarkers were incorporated into the US cancer registry reporting for patients with brain tumors beginning in 2018. We investigated the completeness and validity of these variables and described the epidemiology of molecularly defined brain tumor types. Methods Brain tumor patients with histopathologically confirmed diagnosis in 2018 were identified within the Central Brain Tumor Registry of the United States and NCI’s Surveillance, Epidemiology, and End Results Incidence databases. The brain molecular markers (BMM) site-specific data item was assessed for coding completeness and validity. 1p/19q status, MGMT promoter methylation, WHO grade data items, and new ICD-O-3 codes were additionally evaluated. These data were used to profile the characteristics and age-adjusted incidence rates per 100 000 population of molecularly defined brain tumors with 95% confidence intervals (95% CI). Results BMM completeness across the applicable tumor types was 75%-92% and demonstrated favorable coding validity. IDH-wildtype glioblastomas’ incidence rate was 1.74 (95% CI: 1.69-1.78), as compared to 0.14 for WHO grade 2 (95% CI: 0.12-0.15), 0.15 for grade 3 (95% CI: 0.14-0.16), and 0.07 for grade 4 (95% CI: 0.06-0.08) IDH-mutant astrocytomas. Irrespective of WHO grade, IDH mutation prevalence was highest in adolescent and young adult patients, and IDH-mutant astrocytomas were more frequently MGMT promoter methylated. Among pediatric-type tumors, the incidence rate was 0.06 for H3K27M-mutant diffuse midline gliomas (95% CI: 0.05-0.07), 0.03 for SHH-activated/TP53-wildtype medulloblastomas (95% CI: 0.02-0.03), and Conclusions Our findings illustrate the success of developing a dedicated, integrated diagnosis variable, which provides critical molecular information about brain tumors related to accurate diagnosis.
- Published
- 2023
9. Medicaid expansion is associated with increased 1-year survival for primary malignant brain tumors
- Author
-
Mantas Dmukauskas, Gino Cioffi, Corey Neff, Mackenzie Price, Kristin A Waite, Carol Kruchko, Justin M Barnes, Quinn T Ostrom, and Jill S Barnholtz-Sloan
- Subjects
Oncology ,Surgery ,Neurology (clinical) ,Brief Communication - Published
- 2023
10. Epidemiology of pineoblastoma in the United States, 2000–2017
- Author
-
Kaitlyn Greppin, Gino Cioffi, Kristin A Waite, Quinn T Ostrom, Daniel Landi, Kailey Takaoka, Carol Kruchko, and Jill S Barnholtz-Sloan
- Subjects
Medicine (miscellaneous) ,Original Articles - Abstract
Background Pineoblastoma (PB) is a rare malignant brain tumor originating in the pineal gland. Here, we provide a comprehensive epidemiological analysis of PB in the United States from 2000 to 2017. Methods Data on 1133 patients with PB were acquired from the Central Brain Tumor Registry of the United States, in collaboration with the Centers for Disease Control and Prevention and the National Cancer Institute, from 2000 to 2017. Age-adjusted incidence rates (AAIRs) per 100 000 and incidence rate ratios (IRRs) were reported for age, sex, race, and ethnicity. Using the National Program of Cancer Registries survival database, median survival and hazard ratios (HRs) were evaluated for overall survival from 2001 to 2016. Results Incidence was highest in ages 0–4 years (AAIR: 0.049, 95% CI: 0.042–0.056), decreasing as age increased. Incidence was higher among patients who are Black compared to patients who are White (IRR: 1.71, 95% CI: 1.48–1.98, P < .001), and was impacted by age at diagnosis, with Black-to-White incidence highest in children ages 5–9 years (IRR: 3.43, 95% CI: 2.36–4.94, P < .001). Overall survival was lower for males (HR: 1.39, 95% CI: 1.07–1.79, P = .013). All age groups, excluding those over 40, had improved survival compared to ages 0–4 years. Those who received surgical intervention had better survival compared to those who did not receive surgical treatment. Conclusion PB incidence is highest among children and patients who are Black, and there may be a potential interaction between these factors. Survival is worse among males, young children, and elderly adults, and those who received no surgery. Comprehensive, population-based statistics provide critical information on PB characteristics that could be useful in impacting patient care and prognosis.
- Published
- 2022
11. Primary brain and other central nervous system tumors in the United States (2014-2018): A summary of the CBTRUS statistical report for clinicians
- Author
-
Justin T Low, Quinn T Ostrom, Gino Cioffi, Corey Neff, Kristin A Waite, Carol Kruchko, and Jill S Barnholtz-Sloan
- Subjects
Medicine (miscellaneous) ,Review - Abstract
Background The Central Brain Tumor Registry of the United States (CBTRUS) contains information on all primary brain and other central nervous system (CNS) tumors diagnosed in the United States (US). Here we summarize the 2021 CBTRUS annual statistical report for clinicians. Methods Incidence survival data are obtained from the Centers for Disease Control’s National Program of Cancer Registries (NPCR) and National Cancer Institute’s Surveillance, Epidemiology, and End Results (SEER) program. Survival data are obtained from NPCR. Mortality data are obtained from the National Vital Statistics System. Incidence and mortality rates are age-adjusted using the 2000 US population and presented per 100,000 population. Results An annual average of 86,355 cases of primary malignant and nonmalignant CNS tumors were diagnosed over the period 2014–2018, corresponding to an average annual age-adjusted incidence rate of 24.25. The most commonly occurring malignant tumor was glioblastoma (14.3%), and the most common predominately nonmalignant tumor was meningioma (39%). Over the 2014–2018 period, there were 16,606 annual average deaths due to malignant primary CNS tumors, corresponding to an average annual age-adjusted mortality rate of 4.43. In this report we detail key incidence, survival, and mortality statistics for major primary CNS tumor histologies, highlighting relevant differences by age, sex, and race. Conclusions This summary describes the most up to date population-based incidence of primary malignant and nonmalignant brain and other CNS tumors in the US, and mortality and survival for primary malignant tumors and aims to serve as a useful resource for clinicians.
- Published
- 2023
12. Independently validated sex-specific nomograms for predicting survival in patients with newly diagnosed glioblastoma: NRG Oncology RTOG 0525 and 0825
- Author
-
Deborah T. Blumenthal, Robin Buerki, Mitchell Machtay, Valerie Panet-Raymond, Stephanie L. Pugh, Nirav Patil, Eashwar Somasundaram, James R. Connor, Andrew E. Sloan, H. Ian Robins, Grant K. Hunter, Marta Penas-Prado, Justin D. Lathia, Serah Choi, Kristin A Waite, John C. Flickinger, Lynn S. Ashby, Maria Werner-Wasik, Joshua B. Rubin, Michael E. Berens, Merideth M Wendland, Mark R. Gilbert, Jill S. Barnholtz-Sloan, and Minesh P. Mehta
- Subjects
Oncology ,Male ,Cancer Research ,medicine.medical_specialty ,Survival ,Malignant brain tumor ,Newly diagnosed ,Extent of resection ,Nomogram ,Internal medicine ,Sex differences ,medicine ,Humans ,In patient ,Promoter Regions, Genetic ,Proportional Hazards Models ,business.industry ,Brain Neoplasms ,medicine.disease ,Prognosis ,Sex specific ,Clinical trial ,Nomograms ,Neurology ,Clinical Study ,Female ,Neurology (clinical) ,business ,Glioblastoma - Abstract
Background/purpose Glioblastoma (GBM) is the most common primary malignant brain tumor. Sex has been shown to be an important prognostic factor for GBM. The purpose of this study was to develop and independently validate sex-specific nomograms for estimation of individualized GBM survival probabilities using data from 2 independent NRG Oncology clinical trials. Methods This analysis included information on 752 (NRG/RTOG 0525) and 599 (NRG/RTOG 0825) patients with newly diagnosed GBM. The Cox proportional hazard models by sex were developed using NRG/RTOG 0525 and significant variables were identified using a backward selection procedure. The final selected models by sex were then independently validated using NRG/RTOG 0825. Results Final nomograms were built by sex. Age at diagnosis, KPS, MGMT promoter methylation and location of tumor were common significant predictors of survival for both sexes. For both sexes, tumors in the frontal lobes had significantly better survival than tumors of multiple sites. Extent of resection, and use of corticosteroids were significant predictors of survival for males. Conclusions A sex specific nomogram that assesses individualized survival probabilities (6-, 12- and 24-months) for patients with GBM could be more useful than estimation of overall survival as there are factors that differ between males and females. A user friendly online application can be found here—https://npatilshinyappcalculator.shinyapps.io/SexDifferencesInGBM/.
- Published
- 2021
13. Changes in survival over time for primary brain and other CNS tumors in the United States, 2004-2017
- Author
-
Gino Cioffi, Kristin A. Waite, Jacob L. Edelson, Carol Kruchko, Quinn T. Ostrom, and Jill S. Barnholtz-Sloan
- Subjects
Cancer Research ,Brain Neoplasms ,Incidence ,Brain ,United States ,Survival Rate ,Central Nervous System Neoplasms ,Neurology ,Oncology ,Humans ,Neurology (clinical) ,Registries ,Child ,Aged - Abstract
Purpose Despite advances in cancer diagnosis and clinical care, survival for many primary brain and other central nervous system (CNS) tumors remain poor. This study performs a comprehensive survival analysis on these tumors. Methods Survival differences were determined utilizing the National Program of Cancer Registries Survival Analytic file for primary brain and CNS tumors. Overall survival and survival of the 5 most common histopathologies, within specific age groups, were determined. Overall survival was compared for three time periods: 2004–2007, 2008–2012, and 2013–2017. Survival differences were evaluated using Kaplan–Meier and multivariable Cox proportional hazards models. Models were adjusted for sex, race/ethnicity, and treatment. Malignant and non-malignant brain tumors were assessed separately. Results Among malignant brain and CNS tumor patients overall, there were notable differences in survival by time period among all age groups. Similar differences were noted in non-malignant brain and CNS tumor patients, except for adults (aged 40–64 years), where no survival changes were observed. Survival differences varied within specific histopathologies across age groups. There were improvements in survival in 2008–2012 and 2013–2017, when compared to 2004–2007, in children, AYA, and older adults with malignant tumors, and among older adults with non-malignant tumors. Conclusion Overall survival for malignant brain and other CNS tumors improved slightly in 2013–2017 for all age groups as compared to 2004–2007. Significant changes were observed for non-malignant brain and other CNS tumors among older adults. Information regarding survival over time can be utilized to identify population level effects of diagnostic and treatment improvements.
- Published
- 2022
14. Aligning the Central Brain Tumor Registry of the United States (CBTRUS) histology groupings with current definitions
- Author
-
Kristin A Waite, Gino Cioffi, Carol Kruchko, Nirav Patil, Daniel J Brat, Janet M Bruner, Roger E McLendon, Tarik Tihan, Quinn T Ostrom, and Jill S Barnholtz-Sloan
- Subjects
Medicine (miscellaneous) ,Original Articles - Abstract
Background The Central Brain Tumor Registry of the United States (CBTRUS) uses a histology grouping model based on the World Health Organization (WHO) classifications to group records for clinically relevant statistical reporting. Newly identified genetic markers more accurately stratify patients than histology alone and were incorporated into the 2016 update to the WHO Classification. Methods CBTRUS and consulting neuropathologists reviewed and aligned histology groupings with the 2016 WHO update. “Obsolete” (terms not currently in use) histology nomenclature along with their International Classification of Disease, Oncology 3rd edition (ICD-O-3) codes were identified, some histologies were reclassified to 2016 WHO, and new codes found in 2016 WHO were incorporated. An evaluation of the frequency of histology codes affected in the realignment process, and incidence and survival pre- and post-realignment was conducted. Results After review, 67 codes were noted as obsolete, 51 codes were reclassified, and 12 new codes were incorporated. Histology groups most affected were mesenchymal tumors and neuronal/mixed neuronal-glial tumors. Reorganization resulted in 2588 (0.65%) cases with grouping reassignment or reporting change, indicating that the 2016 WHO Classification revision has impacted the collection and reporting of primary brain and other CNS tumors. Conclusion This work demonstrates the need to be responsive to changes in classification and coding in order to ensure the most up-to-date and accurate statistics for brain and CNS tumors. This will require collaboration from all stakeholders within the brain tumor community, so to have the ability to reconcile clinical practices and surveillance requirements.
- Published
- 2022
15. The MIF SNP rs755622 is a germline determinant of tumor immune activation in Glioblastoma
- Author
-
Tyler J. Alban, Matthew M. Grabowski, Balint Otvos, Defne Bayik, Ajay Zalavadia, Vlad Makarov, Katie Troike, Mary McGraw, Anja Rabljenovic, Adam Lauko, Chase Neumann, Gustavo Roversi, Kristin A. Waite, Gino Cioffi, Nirav Patil, Thuy T. Tran, Kathleen McCortney, Alicia Steffens, C. Marcela Diaz, J. Mark Brown, Kathleen M. Egan, Craig M. Horbinski, Jill S. Barnholtz-Sloan, Michael A. Vogelbaum, Richard Bucala, Timothy A. Chan, Manmeet S. Ahluwalia, and Justin D. Lathia
- Subjects
animal diseases ,chemical and pharmacologic phenomena - Abstract
While immunotherapies have shown durable responses for multiple tumors, their efficacy remains limited in some advanced cancers, including glioblastoma. This may be due to differences in the immune landscape, as the glioblastoma microenvironment strongly favors immunosuppressive myeloid cells, which are linked to an elevation in immune-suppressive cytokines, including macrophage migration inhibitory factor (MIF). We now find that a single-nucleotide polymorphism (SNP) rs755622 in the MIF promoter associates with increased leukocyte infiltration in glioblastoma. Furthermore, we identified lactotransferrin expression as being associated with the rs755622 SNP, which could also be used as a biomarker for immune infiltrated tumors. These findings provide the first example in glioblastoma of a germline SNP that underlies differences in the immune microenvironment and identifies high lactotransferrin as a potential factor promoting immune activation.
- Published
- 2022
16. The Translocator Protein (
- Author
-
Katie M, Troike, Arlet M, Acanda de la Rocha, Tyler J, Alban, Matthew M, Grabowski, Balint, Otvos, Gino, Cioffi, Kristin A, Waite, Jill S, Barnholtz Sloan, Justin D, Lathia, Tomás R, Guilarte, and Diana J, Azzam
- Subjects
single nucleotide polymorphism ,glioblastoma ,biomarker ,survival ,Article ,TSPO - Abstract
Simple Summary The translocator protein 18 kDa (TSPO) gene is highly expressed in glioblastoma (GBM), the most common primary malignant brain tumor, which remains one of the most difficult tumors to treat. TSPO is located in the outer mitochondrial membrane and binds cholesterol through its C-terminal domain. One frequent single-nucleotide polymorphism (SNP) rs6971, which changes the alanine 147 into threonine (Ala147Thr), has been found in the C-terminal domain of the TSPO region and dramatically alters the affinity with which TSPO binds drug ligands. However, the potential association between the TSPO genetic variants and GBM clinical outcomes is not known. Here, we evaluated the effects of the Ala147Thr SNP localized in this TSPO region on biological, sex-specific, overall, and progression-free GBM survival. Our findings suggest an association between the TSPO rs6971 variant and adverse outcomes in male GBM patients but not in females. These findings also suggest that the TSPO rs6971 SNP could be used as a prognostic marker of survival in GBM patients. Abstract Glioblastoma (GBM) is the most common primary brain tumor in adults, with few available therapies and a five-year survival rate of 7.2%. Hence, strategies for improving GBM prognosis are urgently needed. The translocator protein 18kDa (TSPO) plays crucial roles in essential mitochondria-based physiological processes and is a validated biomarker of neuroinflammation, which is implicated in GBM progression. The TSPO gene has a germline single nucleotide polymorphism, rs6971, which is the most common SNP in the Caucasian population. High TSPO gene expression is associated with reduced survival in GBM patients; however, the relation between the most frequent TSPO genetic variant and GBM pathogenesis is not known. The present study retrospectively analyzed the correlation of the TSPO polymorphic variant rs6971 with overall and progression-free survival in GBM patients using three independent cohorts. TSPO rs6971 polymorphism was significantly associated with shorter overall survival and progression-free survival in male GBM patients but not in females in one large cohort of 441 patients. We observed similar trends in two other independent cohorts. These observations suggest that the TSPO rs6971 polymorphism could be a significant predictor of poor prognosis in GBM, with a potential for use as a prognosis biomarker in GBM patients. These results reveal for the first time a biological sex-specific relation between rs6971 TSPO polymorphism and GBM.
- Published
- 2021
17. Functional significance of Sp1, Sp2, and Sp3 transcription factors in regulation of the murine CTP:phosphocholine cytidylyltransferase α promoter
- Author
-
Marica Bakovic, Kristin A. Waite, and Dennis E. Vance
- Subjects
gene regulation ,promoter response elements ,DrosophilaSL2 cells ,C3H10T1/2 cells ,Biochemistry ,QD415-436 - Abstract
The transcription factor Sp1 has been implicated in regulation of the expression of the murine CTP:phosphocholine cytidylyltransferase α (CTα) gene, Ctpct (M. Bakovic, K. Waite, W. Tang, I. Tabas, and D. E. Vance. 1999. Biochim. Biophys. Acta. 1438: 147–165). We have utilized transient transfections, mutation analysis, electromobility gel-shifts, and immunoblot analysis to test the hypothesis that expression of the CTα gene is controlled in part by the binding of three trans-acting nuclear factors, Sp1, Sp2, and Sp3. Sp1 and Sp3 activate CTα gene transcription through sequence specific binding within three promoter domains. In Sp1-mediated transcription, Sp3 acts as an activator in a dose-dependent manner and vice versa. Sp2 represses Sp1- and Sp3-driven transcription in Drosophila SL2 cells, but stimulates transcription in C3H10T1/2 mammalian cells. Our results suggest that the predominant action of Sp proteins is a direct function of local organization of three cis-acting elements in the regions A (−31/−9), B (−88/−50), and C (−148/−128). The ability of distal C (−148/−128) and proximal A (−31/−9) regions to activate or repress transcription depends upon the cellular background. The multiple binding elements at position B (−88/−50) confer a positive regulation independent of the cell context. However, the effectiveness of Sp proteins at this site is strongly governed by neighboring sites A and C. The results suggest that the level of expression of the CTα gene will depend on the cell type, the availability of Sp proteins, and the structure and organization of three cis-acting elements.—Bakovic, M., K. A. Waite, and D. E. Vance. Functional significance of Spl, Sp2, and Sp3 transcription factors in regulation of the murine CTP:phosphocholine cytidylyltransferase α promoter. J. Lipid Res. 41: 583–594.
- Published
- 2000
- Full Text
- View/download PDF
18. PPAR𝛾, PTEN, and the Fight against Cancer
- Author
-
Rosemary E. Teresi and Kristin A. Waite
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Peroxisome proliferator-activated receptor gamma (PPAR𝛾) is a ligand-activated transcription factor, which belongs to the family of nuclear hormone receptors. Recent in vitro studies have shown that PPAR𝛾 can regulate the transcription of phosphatase and tensin homolog on chromosome ten (PTEN), a known tumor suppressor. PTEN is a susceptibility gene for a number of disorders, including breast and thyroid cancer. Activation of PPAR𝛾 through agonists increases functional PTEN protein levels that subsequently induces apoptosis and inhibits cellular growth, which suggests that PPAR𝛾 may be a tumor suppressor. Indeed, several in vivo studies have demonstrated that genetic alterations of PPAR𝛾 can promote tumor progression. These results are supported by observations of the beneficial effects of PPAR𝛾 agonists in the in vivo cancer setting. These studies signify the importance of PPAR𝛾 and PTEN's interaction in cancer prevention.
- Published
- 2008
- Full Text
- View/download PDF
19. Nuclear PTEN levels and G2 progression in melanoma cells
- Author
-
Charis Eng, Kristin A. Waite, Abraham I. Jacob, and Todd Romigh
- Subjects
G2 Phase ,Cytoplasm ,Cancer Research ,Cell ,Down-Regulation ,Dermatology ,Article ,Cell Line, Tumor ,CDC2 Protein Kinase ,medicine ,Humans ,PTEN ,Tensin ,Phosphorylation ,Melanoma ,Cell Nucleus ,Cyclin-dependent kinase 1 ,biology ,Akt/PKB signaling pathway ,Cell Cycle ,PTEN Phosphohydrolase ,G2-M DNA damage checkpoint ,Cell cycle ,Cell Compartmentation ,Cell biology ,Gene Expression Regulation, Neoplastic ,Cell nucleus ,medicine.anatomical_structure ,Oncology ,Protein Biosynthesis ,biology.protein ,Cancer research ,RNA Interference ,Poly(ADP-ribose) Polymerases - Abstract
The phosphatase and tensin homolog (PTEN) exerts its function, in part, by negatively regulating the well-known phosphatidylinositol-3-kinase/AKT signaling pathway. Previous histological work has suggested that alterations in the nuclear/cytoplasmic compartmentalization of PTEN may play a role in the development and progression of melanoma. In this study, we examined the nuclear/cytoplasmic compartmentalization of PTEN in melanoma cell lines and its correlation with the cell cycle. Studies were performed in melanoma cells lines using classic cell biological techniques. In contrast to breast cancer cell lines, we found that increased levels of nuclear PTEN levels correlate with G2 rather than with G1 arrest. In WM164 and SKmel28 cells, overexpression of PTEN protein did not significantly increase the number of cells in the G2 phase. Differential CDC2 phosphorylation levels in cells that overexpressed PTEN compared with those where PTEN was downregulated suggest some involvement of PTEN in G2 checkpoint regulation. The data suggest that although nuclear PTEN levels correlate with the G2 phase, the role of PTEN in modulating G2/M arrest is not limiting. Further, the specific cell cycle phase regulated by nuclear PTEN is cell-type dependent. Taken together, our observations suggest that in melanoma, nuclear PTEN is involved in G2 progression possibly through the modulation of CDC2, opening up a new arena for investigation.
- Published
- 2009
20. Germline and somatic cancer-associated mutations in the ATP-binding motifs of PTEN influence its subcellular localization and tumor suppressive function
- Author
-
Todd Romigh, Kristin A. Waite, Glenn P. Lobo, Charis Eng, Sarah M. Planchon, and Najah T. Nassif
- Subjects
Male ,Tumor suppressor gene ,Somatic cell ,Amino Acid Motifs ,Molecular Sequence Data ,Biology ,medicine.disease_cause ,Germline ,03 medical and health sciences ,Adenosine Triphosphate ,0302 clinical medicine ,Germline mutation ,Cell Line, Tumor ,Genetics ,medicine ,Humans ,PTEN ,Amino Acid Sequence ,Molecular Biology ,Germ-Line Mutation ,Genetics (clinical) ,030304 developmental biology ,Genetics & Heredity ,0303 health sciences ,Mutation ,PTEN Phosphohydrolase ,Articles ,General Medicine ,Cowden syndrome ,medicine.disease ,3. Good health ,Protein Transport ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Female ,Hamartoma Syndrome, Multiple ,Carcinogenesis ,Sequence Alignment ,Protein Binding - Abstract
Germline and somatic PTEN mutations are found in Cowden syndrome (CS) and multiple sporadic malignancies, respectively. PTEN function appears to be modulated by subcellular compartmentalization, and mislocalization may affect function. We have shown that cellular ATP levels affect nuclear PTEN levels. Here, we examined the ATP-binding capabilities of PTEN and functional consequences, relevant to cancer-associated mutations. PTEN mutation analysis of CS patients and sporadic colorectal carcinomas and comparative aminoacid analysis were utilized to identify mutations in ATP-binding motifs. The ability of wild-type (WT) or mutant PTEN to bind ATP was assessed by ATP-agarose-binding assays. Subcellular fractionation, western blotting, confocal microscopy and growth assays were used to determine relative nuclear-cytoplasmic localization and function. Somatic colorectal carcinoma-derived PTEN missense mutations were associated with nuclear mislocalization. These mutations altered cellular proliferation, apoptosis and anchorage-dependent growth. Examination of PTEN's amino acid sequence revealed these mutations resided in previously undescribed ATP-binding motifs (c.60-73; c.122-136). In contrast to WT PTEN, both cancer-associated somatic and germline-derived PTEN missense mutations, which lie within the ATP-binding motifs, result in mutant PTEN that does not bind ATP efficiently. We also show that CS patients with germline ATP-binding motif-mutations had nuclear PTEN mislocalization. Of four unrelated patients with functional germline ATP-binding domain mutations, all three female patients had breast cancers. Germline and somatic mutations within PTEN's ATP-binding domain play important pathogenic roles in both heritable and sporadic carcinogenesis by PTEN nuclear mislocalization resulting in altered signaling and growth. Manipulation of ATP may represent novel therapies in tumors with such PTEN alterations. © 2009 The Author(s).
- Published
- 2009
21. ATP modulates PTEN subcellular localization in multiple cancer cell lines
- Author
-
Glenn P. Lobo, Charis Eng, Kristin A. Waite, Todd Romigh, Janet A. Houghton, and Sarah M. Planchon
- Subjects
Cell type ,Tumor suppressor gene ,Somatic cell ,Breast Neoplasms ,03 medical and health sciences ,0302 clinical medicine ,Adenosine Triphosphate ,Cell Line, Tumor ,Genetics ,PTEN ,Humans ,Thyroid Neoplasms ,Molecular Biology ,Genetics (clinical) ,PI3K/AKT/mTOR pathway ,030304 developmental biology ,Cell Nucleus ,0303 health sciences ,biology ,Kinase ,PTEN Phosphohydrolase ,General Medicine ,Articles ,Subcellular localization ,3. Good health ,Protein Transport ,030220 oncology & carcinogenesis ,Lipid phosphatase activity ,biology.protein ,Cancer research ,Colorectal Neoplasms - Abstract
The tumour suppressor gene PTEN plays an important somatic role in both hereditary and sporadic breast carcinogenesis. While the role of PTEN's lipid phosphatase activity, as a negative regulator of the cytoplasmic phosphatidylinositol-3-kinase/Akt pathway is well known, it is now well established that PTEN exists and functions in the nucleus. Multiple mechanisms of regulating PTEN's subcellular localization have been reported. However none are ubiquitous across multiple cancer cell lines and tissue types. We show here that adenosine triphosphate (ATP) regulates PTEN subcellular localization in a variety of different cancer cell lines, including those derived from breast, colon and thyroid carcinomas. Cells deficient in ATP show an increased level of nuclear PTEN protein. This increase in PTEN is reversed when cells are supplemented with ATP, ADP or AMP. In contrast, the addition of the non-hydrolyzable analogue ATPgammaS, did not reverse nuclear PTEN protein levels in all the cell types tested. To our knowledge, this is the first report that describes a regulation of PTEN subcellular localization that is not specific to one cell line or tissue type, but appears to be common across a variety of cell lineages.
- Published
- 2008
22. Regulation of the PTEN promoter by statins and SREBP
- Author
-
Kristin A. Waite, Charis Eng, Sarah M. Planchon, and Rosemary E. Teresi
- Subjects
Transcriptional Activation ,Simvastatin ,Peroxisome proliferator-activated receptor gamma ,Indoles ,Statin ,Transcription, Genetic ,Tumor suppressor gene ,medicine.drug_class ,Electrophoretic Mobility Shift Assay ,Cell Line ,Fatty Acids, Monounsaturated ,Rosiglitazone ,Downregulation and upregulation ,Genes, Reporter ,Genetics ,medicine ,Humans ,PTEN ,Fluvastatin ,Promoter Regions, Genetic ,Molecular Biology ,Transcription factor ,Genetics (clinical) ,Pravastatin ,Regulation of gene expression ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,PTEN Phosphohydrolase ,nutritional and metabolic diseases ,General Medicine ,Up-Regulation ,Sterol regulatory element-binding protein ,PPAR gamma ,Gene Expression Regulation ,biology.protein ,Cancer research ,Thiazolidinediones ,lipids (amino acids, peptides, and proteins) ,Hydroxymethylglutaryl-CoA Reductase Inhibitors ,Sterol Regulatory Element Binding Protein 1 ,Transcription Factors - Abstract
Germline mutations in the tumor-suppressor gene PTEN predispose to heritable breast cancer. The transcription factor peroxisome proliferator-activated receptor-gamma (PPARgamma) has also been implicated as a tumor suppressor pertinent to a range of neoplasias, including breast cancer. We previously demonstrated that lovastatin may signal through PPARgamma and directly upregulate PTEN expression at the transcriptional level. In our current study, we show that simvastatin, pravastatin and fluvastatin can induce PTEN expression in a dose-dependent manner. This resulted from an increase in PTEN mRNA indicating transcriptional upregulation. In addition, we observed, for the first time, that upregulation of sterol response element-binding protein (SREBP), known to induce PPARgamma expression, can increase PTEN expression. Using reporter assays, we observed that both the statins and SREBP could specifically induce PPARgamma-mediated transcription. However, the statins do not appear to signal through SREBP. Furthermore, our results indicate that SREBP utilizes PPARgamma's transcriptional activity to induce PTEN transcription, whereas the statins signal through PPARgamma's protein activity to upregulate PTEN expression. Overall, our observations suggest that statins signal through another transcription factor, in a PPARgamma-dependent manner, which in turn induces PTEN transcription. We, therefore, studied the full-length PTEN promoter through serial deletion reporter assays and electromobility shift assays and identified a region between -854 and -791 that binds an as-yet-unidentified transcription factor, through which the statins induce PTEN expression. Since PTEN is constitutively active, our data indicate it may be worthwhile to examine statin and SREBP stimulation as mechanisms to increase PTEN expression for therapeutic and preventative strategies in cancer, diabetes mellitus and cardiovascular disease.
- Published
- 2007
23. Cowden Syndrome–Affected Patients with PTEN Promoter Mutations Demonstrate Abnormal Protein Translation
- Author
-
Marcus G. Pezzolesi, Kristin A. Waite, Rosemary E. Teresi, Kevin Zbuk, and Charis Eng
- Subjects
Untranslated region ,Models, Molecular ,Tumor suppressor gene ,Breast Neoplasms ,Biology ,medicine.disease_cause ,Transfection ,Article ,Germline mutation ,Cell Line, Tumor ,medicine ,Genetics ,Tensin ,PTEN ,Humans ,Point Mutation ,Genetics(clinical) ,RNA, Messenger ,Luciferases ,Promoter Regions, Genetic ,Genetics (clinical) ,DNA Primers ,Mutation ,Binding Sites ,Base Sequence ,Point mutation ,PTEN Phosphohydrolase ,Cowden syndrome ,DNA ,Syndrome ,medicine.disease ,Molecular biology ,Recombinant Proteins ,Case-Control Studies ,Protein Biosynthesis ,Cancer research ,biology.protein ,Nucleic Acid Conformation ,Female ,5' Untranslated Regions ,Hamartoma Syndrome, Multiple ,HeLa Cells ,Protein Binding ,Transcription Factors - Abstract
Germline mutations of PTEN (phosphatase and tensin homolog deleted on chromosome 10) are associated with the multihamartomatous disorder Cowden syndrome (CS). Moreover, patients with CS with germline PTEN promoter mutations have aberrant PTEN protein expression and an increased frequency of breast cancer. Here, we examined the downstream effect of five PTEN promoter variants (−861G/T, −853C/G, −834C/T, −798G/C, and −764G/A) that are not within any known cis-acting regulatory elements. Clinically, all five of these patients have been given diagnoses of breast, thyroid, and/or endometrial cancer. We demonstrated that protein binding to the PTEN promoter (−893 to −755) was not altered in the five variants when compared with the wild-type (WT) promoter. However, reporter assays indicated that three of the variants (−861G/T, −853C/G, and −764G/A) demonstrated an ∼50% decrease in luciferase activity compared with the WT construct. PTEN messenger RNA (mRNA) levels were not altered in these variants, whereas secondary structure predictions indicated that different PTEN 5′ untranslated region transcript-folding patterns exist in three variants, suggesting an inhibition of protein translation. This was confirmed by PTEN protein analysis. These data indicate that variants causing large mRNA secondary structure alterations result in an inhibition of protein translation and a decrease in PTEN protein expression. These data emphasize the importance of PTEN promoter nucleotide variations and their ability to lead to CS progression by a novel regulatory mechanism. Importantly, these patients have a high prevalence of breast, thyroid, and endometrial malignancies; thus, understanding of the mechanism of PTEN dysfunction in these patients will lead to more-sensitive molecular diagnostic and predictive testing and, ultimately, to rational targeted therapies to treat or prevent malignancy.
- Published
- 2007
- Full Text
- View/download PDF
24. PTEN regulates phospholipase D and phospholipase C
- Author
-
Charis Eng, Kristin A. Waite, and Christopher Alvarez-Breckenridge
- Subjects
Male ,Tumor suppressor gene ,Gene Expression ,Breast Neoplasms ,Biology ,Cell Line, Tumor ,Phospholipase D ,Genetics ,medicine ,Homeostasis ,Humans ,PTEN ,Molecular Biology ,Phospholipids ,Genetics (clinical) ,PI3K/AKT/mTOR pathway ,Estradiol ,Phospholipase C ,Phospholipase C gamma ,PTEN Phosphohydrolase ,General Medicine ,Lipid signaling ,Cowden syndrome ,medicine.disease ,Type C Phospholipases ,Lipid phosphatase activity ,Mutation ,Cancer research ,biology.protein ,Female ,lipids (amino acids, peptides, and proteins) ,Signal Transduction - Abstract
PTEN is an ubiquitously expressed tumor suppressor which plays a prominent role in the pathogenesis of many types of sporadic solid tumors, including breast cancer, as well as hematologic malignancies. Germline PTEN mutations cause 85% of Cowden syndrome (CS), characterized by a high risk of breast and thyroid cancers, and 65% of Bannayan-Riley-Ruvalcaba syndrome (BRRS), characterized by lipomatosis, hemangiomas and speckled penis. Historically, PTEN's role in tumor suppression has been linked to the down-regulation of the PI3K/AKT pathway by PTEN's lipid phosphatase activity. Beyond the AKT pathway, however, there has been minimal examination of PTEN's responsibility in lipid-derived cellular signaling. As phospholipids have been shown to be critical components in signal transduction and cellular proliferation and PTEN controls cellular phospholipid levels, we hypothesized that PTEN functions as a regulator of lipid signaling and homeostasis. Increased PTEN expression in unstimulated MCF-7 breast cancer cells results in a 51% increase in phosphatidic acid, with a decrease in phosphatidylcholine, suggesting that PTEN may regulate phospholipase D (PLD). PTEN overexpression results in a 30% increase in basal PLD activity. As phospholipase C (PLC) is both involved in PLD activation and is regulated by PIP2/3 levels, we investigated the role of PTEN on PLC activation. Our data suggest that PTEN modulates PLC:PLD activation pathways and indicate that the pathogenesis of CS/BRRS has a more complex biochemical basis beyond simply activating the PI3K pathway. This provides alternative routes for PTEN's tumor suppressor action that may be beneficial in the creation of novel targets for cancer therapy and prevention.
- Published
- 2007
25. Nuclear Localization of PTEN Is Regulated by Ca2+ through a Tyrosil Phosphorylation–Independent Conformational Modification in Major Vault Protein
- Author
-
Kristin A. Waite, Charis Eng, and Takeo Minaguchi
- Subjects
Cancer Research ,Protein Conformation ,Apoptosis ,Breast Neoplasms ,Transfection ,Cell Line, Tumor ,Major vault protein ,Humans ,Tensin ,PTEN ,RNA, Neoplasm ,Phosphorylation ,RNA, Small Interfering ,Phosphotyrosine ,Protein kinase B ,DNA Primers ,Vault Ribonucleoprotein Particles ,Cell Nucleus ,Base Sequence ,biology ,Cell Cycle ,PTEN Phosphohydrolase ,Membrane Proteins ,Cell cycle ,Cell biology ,Protein Transport ,Oncology ,biology.protein ,Cancer research ,Calcium ,Female ,Signal transduction ,Nuclear localization sequence ,Plasmids - Abstract
We have recently shown in MCF-7 cells that nuclear phosphatase and tensin homologue deleted on chromosome 10 (PTEN) down-regulates phosphorylation of p44/42 and cyclin D1 and induces G1 cell cycle arrest, whereas cytoplasmic PTEN down-regulates phosphorylation of Akt, up-regulates p27, and induces apoptosis. In this manner, nucleocytoplasmic partitioning of PTEN seems to differentially regulate the cell cycle and apoptosis. We have also reported that PTEN has nuclear localization signal–like sequences required for major vault protein (MVP)–mediated nuclear translocation. To date, several other proteins are reported to interact with MVP, including extracellular signal-regulated kinases and steroid receptors, suggesting that MVP is likely to be involved in signal transduction through nucleocytoplasmic transport. However, the exact mechanism of MVP-mediated nucleocytoplasmic shuttling remains elusive. PTEN reportedly interacts in vitro with the EF hand–like motif of MVP in a Ca2+-dependent manner. The current study shows that small interfering RNA–mediated MVP silencing decreases the nuclear localization of PTEN and increases phosphorylation of nuclear p44/42. We show in situ that PTEN-MVP interaction is Ca2+ dependent and is abolished by Mg2+. Nuclear localization of PTEN is decreased by increasing Ca2+ levels in culture medium in a dose-dependent manner. Ca2+ ionophore A23187 increases nuclear localization of PTEN and decreases phosphorylation of nuclear p44/42. Finally, we show that Ca2+-dependent PTEN-MVP interaction is not related to MVP's tyrosil phosphorylation but rather due to its conformational modification. Our observations suggest that Ca2+ regulates PTEN's nuclear entry through a tyrosil phosphorylation–independent conformational change in MVP. Collectively, our data present evidence of a novel crosstalk between the Ca2+ signaling–mediated regulation of the cell cycle and MVP-mediated nuclear PTEN localization and function. (Cancer Res 2006; 66(24): 11677-82)
- Published
- 2006
26. The ERK1/2 pathway modulates nuclear PTEN-mediated cell cycle arrest by cyclin D1 transcriptional regulation
- Author
-
Michael C. Ostrowski, Kristin A. Waite, Takeo Minaguchi, Todd Romigh, Ji Hyun Chung, and Charis Eng
- Subjects
Cell cycle checkpoint ,Transcription, Genetic ,Tumor suppressor gene ,Cyclin D ,Nuclear Localization Signals ,Cyclin A ,Down-Regulation ,Transfection ,Suppression, Genetic ,Cyclin D1 ,Cell Line, Tumor ,Genetics ,Humans ,PTEN ,Phosphorylation ,Molecular Biology ,Genetics (clinical) ,Cyclin ,Cell Nucleus ,biology ,Cell Cycle ,PTEN Phosphohydrolase ,General Medicine ,Cell cycle ,Cell biology ,biology.protein ,Cancer research ,Mitogen-Activated Protein Kinases ,Signal Transduction - Abstract
PTEN, a tumor suppressor phosphatase that dephosphorylates both protein and lipid substrates, is mutated in both heritable and sporadic breast cancer. Until recently, PTEN-mediated cell cycle arrest and apoptosis were thought to occur through its well-documented cytoplasmic activities. We have shown that PTEN localizes to the nucleus coincident with the G0-G1 phases of the cell cycle and that compartmentalization may regulate cell cycle progression dependent upon the down-regulation of cyclin D1. However, the mechanism for cyclin D1-dependent growth suppression by nuclear PTEN has remained largely undefined. Utilizing MCF-7 Tet-Off breast cancer cell lines stably expressing two different nuclear localization defective PTEN mutants, as well as wild-type PTEN and empty vector control cells, we demonstrate that nuclear PTEN down-regulates cyclin D1 transcription and this event is mediated by the down-regulation of MAPK specifically by nuclear localized PTEN. These results provide further evidence that nuclear PTEN plays a role through cell cycle suppression functions in regulating carcinogenesis.
- Published
- 2006
27. Increased PTEN expression due to transcriptional activation of PPARγ by Lovastatin and Rosiglitazone
- Author
-
Rosemary E. Teresi, Chung-Wai Shaiu, V. Krishna K. Chatterjee, Charis Eng, Ching-Shih Chen, and Kristin A. Waite
- Subjects
Transcriptional Activation ,Cancer Research ,Tumor suppressor gene ,Antineoplastic Agents ,Breast Neoplasms ,Rosiglitazone ,Downregulation and upregulation ,Cell Line, Tumor ,Transcriptional regulation ,medicine ,Humans ,Hypoglycemic Agents ,Tensin ,PTEN ,Genetic Predisposition to Disease ,Lovastatin ,Transcription factor ,Germ-Line Mutation ,Dose-Response Relationship, Drug ,biology ,G1 Phase ,PTEN Phosphohydrolase ,Up-Regulation ,PPAR gamma ,Oncology ,Cancer research ,biology.protein ,Thiazolidinediones ,medicine.drug - Abstract
Germline mutations in the tumor suppressor gene PTEN (protein phosphatase and tensin homolog located on chromosome ten) predispose to heritable breast cancer. The transcription factor PPARgamma has also been implicated as a tumor suppressor pertinent to a range of neoplasias, including breast cancer. A putative PPARgamma binding site in the PTEN promoter indicates that PPARgamma may regulate PTEN expression. We show here that the PPARgamma agonist Rosiglitazone, along with Lovastatin, induce PTEN in a dose- and time-dependent manner. Lovastatin- or Rosiglitazone-induced PTEN expression was accompanied by a decrease in phosphorylated-AKT and phosphorylated-MAPK and an increase in G1 arrest. We demonstrate that the mechanism of Lovastatin- and Rosiglitazone-associated PTEN expression was a result of an increase in PTEN mRNA, suggesting that this increase was transcriptionally-mediated. Compound-66, an inactive form of Rosiglitazone, which is incapable of activating PPARgamma, was unable to elicit the same response as Rosiglitazone, signifying that the Rosiglitazone response is PPARgamma-mediated. To support this, we show, using reporter assays including dominant-negative constructs of PPARgamma, that both Lovastatin and Rosiglitazone specifically mediate PPARgamma activation. Additionally, we demonstrated that cells lacking PTEN or PPARgamma were unable to induce PTEN mediated cellular events in the presence of Lovastatin or Rosiglitazone. These data are the first to demonstrate that Lovastatin can signal through PPARgamma and directly demonstrate that PPARgamma can upregulate PTEN at the transcriptional level. Since PTEN is constitutively active, our data indicates it may be worthwhile to examine Rosiglitazone and Lovastatin stimulation as mechanisms to increase PTEN expression for therapeutic and preventative strategies including cancer, diabetes mellitus and cardiovascular disease.
- Published
- 2006
28. Silencing of the Maternally Imprinted Tumor Suppressor ARHI Contributes to Follicular Thyroid Carcinogenesis
- Author
-
Andrea Frilling, Charis Eng, Micheala A. Aldred, Christoph E. Broelsch, Carl Morrison, Kristin A. Waite, Christoph Plass, and Frank Weber
- Subjects
Adenoma ,rho GTP-Binding Proteins ,Antimetabolites, Antineoplastic ,medicine.medical_specialty ,Tumor suppressor gene ,Endocrinology, Diabetes and Metabolism ,Clinical Biochemistry ,Loss of Heterozygosity ,Biology ,Decitabine ,medicine.disease_cause ,Biochemistry ,Thyroid carcinoma ,Genomic Imprinting ,Endocrinology ,Internal medicine ,Adenocarcinoma, Follicular ,medicine ,Humans ,Genes, Tumor Suppressor ,Gene Silencing ,Thyroid Neoplasms ,Thyroid cancer ,Thyroid neoplasm ,DNA Primers ,Base Sequence ,Reverse Transcriptase Polymerase Chain Reaction ,Biochemistry (medical) ,Thyroid ,medicine.disease ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,DNA methylation ,Azacitidine ,Female ,Genomic imprinting ,Carcinogenesis - Abstract
The two most common subtypes of thyroid cancer, follicular thyroid carcinoma (FTC) and papillary thyroid carcinoma, have been extensively studied, but our fundamental understanding of the molecular events in thyroid epithelial oncogenesis is still limited. Unreported data from our previous published global gene expression analysis revealed that the tumor suppressor gene aplysia ras homolog I (ARHI) is frequently underexpressed in FTCs. In this study, we elucidated the frequency and mechanism of ARHI silencing in benign and malignant thyroid neoplasia. We demonstrated that underexpression of ARHI occurs principally in FTCs (P = 0.0018), including its oncocytic variant (11 of 13), even at minimally invasive stage but not classic papillary thyroid carcinoma (two of seven) or follicular adenoma (FA) (three of 14). FTCs show strong allelic imbalance with reduction in copy number/loss of heterozygosity (LOH) in 69%, compared with less than 10% for FAs. In combination with our LOH data, bisulfite sequencing in a subset of samples revealed that FA displays a symmetric methylation pattern, likely representing one unmethylated allele and one presumptively imprinted allele, whereas FTC shows a virtually complete methylation pattern, representing LOH of the nonimprinted allele with only the hypermethylated allele remaining. Furthermore, we showed that pharmacologic inhibition of histone deacetylation but not demethylation could reactivate ARHI expression in the FTC133 FTC cell line. Therefore, our data suggest that silencing of the putative maternally imprinted tumor suppressor gene ARHI, primarily by large genomic deletion in conjunction with hypermethylation of the genomically imprinted allele, serves as a key early event in follicular thyroid carcinogenesis.
- Published
- 2005
29. BMP2 exposure results in decreased PTEN protein degradation and increased PTEN levels
- Author
-
Charis Eng and Kristin A. Waite
- Subjects
Mutation ,biology ,Tumor suppressor gene ,Protein turnover ,General Medicine ,Cowden syndrome ,medicine.disease ,medicine.disease_cause ,BMPR1A ,Germline mutation ,Genetics ,biology.protein ,Cancer research ,medicine ,PTEN ,Signal transduction ,Molecular Biology ,Genetics (clinical) - Abstract
The tumour suppressor gene PTEN encodes a dual-specificity phosphatase that recognizes protein and phosphatidylinositiol substrates and modulates cellular functions such as migration and proliferation. Germline mutations of PTEN have been shown to cause Cowden syndrome, Bannayan-Riley-Ruvalcaba syndrome and Proteus syndrome. Recently, germline mutations in BMPR1A, the gene encoding the type 1A receptor of bone morphogenetic proteins (BMP) have been found in rare families with Cowden syndrome, suggesting that there may be a link between BMP signaling and PTEN. We thus sought to determine whether BMP2 stimulation alters PTEN protein levels in the breast cancer line, MCF-7. We found that exposure to BMP2 increased PTEN protein levels in a time- and dose-dependent manner. The increase in PTEN protein was rapid and was not due to an increase in new protein synthesis, as cycloheximide treatment did not inhibit BMP2-induced PTEN accumulation, suggesting that BMP2 stimulation inhibited PTEN protein degradation. Indeed, we found that BMP2 treatment of MCF-7 cells decreased the association of PTEN with two proteins in the degradative pathway, UbCH7 and UbC9. These data indicate that BMP2 exposure can regulate PTEN protein levels by decreasing PTEN's association with the degradative pathway. This opens up a new mode of regulating PTEN activity to be investigated further and may explain why BMPR1A can act as a minor susceptibility gene for PTEN mutation negative Cowden syndrome.
- Published
- 2003
30. Phosphatidic Acid Regulates Tyrosine Phosphorylating Activity in Human Neutrophils
- Author
-
Kristin A. Waite, Javid Heravi, Linda C. McPhail, and Susan Sergeant
- Subjects
Phosphatidylethanolamine ,Phospholipase D ,Kinase ,Cell Biology ,Phosphatidylserine ,Phosphatidic acid ,Biology ,Biochemistry ,Molecular biology ,chemistry.chemical_compound ,chemistry ,Phosphorylation ,Phosphatidylinositol ,Tyrosine ,Molecular Biology - Abstract
In human neutrophils, the activation of phospholipase D and the Tyr phosphorylation of proteins are early signaling events upon cell stimulation. We found that the pretreatment of neutrophils with ethanol (0.8%) or 1-butanol (0.3%), which results in the accumulation of phosphatidylalcohol at the expense of phosphatidic acid (PA), decreased the phorbol myristate acetate-stimulated Tyr phosphorylation of endogenous proteins (42, 115 kDa). When neutrophil cytosol was incubated in the presence or absence of PA, these and other endogenous proteins became Tyr-phosphorylated in a PA-dependent manner. In contrast, phosphatidylalcohols exhibited only 25% (phosphatidylethanol) or 5% (phosphatidylbutanol) of the ability of PA to stimulate Tyr phosphorylation in the cell-free assay. Similarly, other phospholipids (phosphatidylcholine, phosphatidylethanolamine, phosphatidylglycerol, phosphatidylserine, phosphatidylinositol, polyphosphoinositides, and sphingosine 1-phosphate) showed little ability to stimulate Tyr phosphorylation. These data suggest that PA can function as an intracellular regulator of Tyr phosphorylating activity. Gel filtration chromatography of leukocyte cytosol revealed a peak of PA-dependent Tyr phosphorylating activity distinct from a previously described PA-dependent phosphorylating activity (Waite, K. A., Wallin, R., Qualliotine-Mann, D., and McPhail, L. C. (1997) J. Biol. Chem. 272, 15569-15578). Among the protein Tyr kinases expressed in neutrophils, only Fgr eluted exclusively in the peak of PA-dependent Tyr phosphorylating activity. Importantly, Fgr isolated from unstimulated neutrophil lysates showed increased activity in the presence of PA but not phosphatidylbutanol. Moreover, the pretreatment of neutrophils with 1-butanol decreased Fgr activity in cells stimulated with formyl-methionyl-leucyl phenylalanine plus dihydrocytochalasin B. Together, these results suggest a new second messenger role for PA in the regulation of Tyr phosphorylation.
- Published
- 2001
31. Why Expression of PhosphatidylethanolamineN-Methyltransferase Does Not Rescue Chinese Hamster Ovary Cells That Have an Impaired CDP-Choline Pathway
- Author
-
Dennis E. Vance and Kristin A. Waite
- Subjects
Cytidine Diphosphate Choline ,Phosphatidylethanolamine N-Methyltransferase ,CHO Cells ,Biology ,Transfection ,Biochemistry ,Gene Expression Regulation, Enzymologic ,chemistry.chemical_compound ,Methionine ,Cricetinae ,Animals ,Choline-Phosphate Cytidylyltransferase ,CDP-choline pathway ,Enzyme Inhibitors ,Molecular Biology ,Triglycerides ,Phosphocholine ,Cell growth ,Chinese hamster ovary cell ,Temperature ,Methyltransferases ,Cell Biology ,Clone Cells ,Culture Media ,Cell biology ,Kinetics ,chemistry ,Phospholipases ,Cell culture ,Phosphatidylethanolamine N-methyltransferase ,Apoptosis ,Phosphatidylcholines ,Cell Division - Abstract
The mutant Chinese hamster ovary cell line (CHO), MT58, has a temperature-sensitive mutation in CTP:phosphocholine cytidylyltransferase (CT), preventing phosphatidylcholine (PC) synthesis at 40 degrees C which results in apoptosis. Previous studies (Houweling, M., Cui, Z., and Vance, D. E. (1995) J. Biol. Chem. 270, 16277-16282) showed that expression of wild-type CT-alpha rescued the cells at 40 degrees C, whereas expression of phosphatidylethanolamine N-methyltransferase-2 (PEMT2) did not, even though PC levels appeared to be maintained at wild-type levels after 24 h at the restrictive temperature. We report that the failure of PEMT2 to rescue the MT58 cell line is due to inadequate long term PC synthesis. We found that changing the medium every 24 h rescued the PEMT2-expressing MT58 cells grown at 40 degrees C. This was due to the uptake and utilization of lipids in the serum. At 40 degrees C, PC levels in the wild-type CHO cells and CT-expressing MT58 cells increased over time whereas PC levels did not change in both the MT58 and PEMT2-expressing MT58 cell lines. Further investigation found that both the PEMT2-expressing MT58 and MT58 cell lines accumulated triacylglycerol at 40 degrees C. Pulse-chase experiments indicated that lyso-PC accumulated to a higher degree at 40 degrees C in the PEMT2-expressing MT58 cells compared with CT-expressing MT58 cells. Transfection of the PEMT-expressing MT58 cells with additional PEMT2 cDNA partially rescued the growth of these cells at 40 degrees C. Inhibition of PC degradation, by inhibitors of phospholipases, also stimulated PEMT-expressing MT58 cell growth at 40 degrees C. Best results were observed using a calcium-independent phospholipase A(2) inhibitor, methyl arachidonyl fluorophosphonate. This inhibitor also increased PC mass in the PEMT2-expressing MT58 cells. When the cells are shifted to 40 degrees C, PC degradation by enzymes such as phospholipases is greater than PC synthesis in the mutant PEMT2-expressing MT58 cells. Taken together, these results indicate that PEMT2 expression fails to rescue the mutant cell line at 40 degrees C because it does not maintain PC levels required for cellular replication.
- Published
- 2000
32. A Phosphatidic Acid-activated Protein Kinase and Conventional Protein Kinase C Isoforms Phosphorylate p22 , an NADPH Oxidase Component
- Author
-
Reidar Wallin, Kristin A. Waite, Debra S. Regier, and Linda C. McPhail
- Subjects
inorganic chemicals ,Phosphatidic Acids ,macromolecular substances ,Mitogen-activated protein kinase kinase ,environment and public health ,Biochemistry ,MAP2K7 ,ASK1 ,Phosphorylation ,Protein kinase A ,Molecular Biology ,Protein Kinase C ,Protein kinase C ,NADPH oxidase ,Cell-Free System ,biology ,MAP kinase kinase kinase ,Chemistry ,NADPH Dehydrogenase ,Membrane Transport Proteins ,NADPH Oxidases ,Cell Biology ,Phosphoproteins ,Molecular biology ,Enzyme Activation ,Isoenzymes ,enzymes and coenzymes (carbohydrates) ,biology.protein ,bacteria ,Cyclin-dependent kinase 9 - Abstract
Using a phosphorylation-dependent cell-free system to study NADPH oxidase activation (McPhail, L. C., Qualliotine-Mann, D., and Waite, K. A. (1995) Proc. Natl. Acad. Sci. U. S. A. 92, 7931-7935), we previously showed that p47(phox), a cytosolic NADPH oxidase component, is phosphorylated. Now, we show that p22(phox), a subunit of the NADPH oxidase component flavocytochrome b(558), also is phosphorylated. Phosphorylation is selectively activated by phosphatidic acid (PA) versus other lipids and occurs on a threonine residue in p22(phox). We identified two protein kinase families capable of phosphorylating p22(phox): 1) a potentially novel, partially purified PA-activated protein kinase(s) known to phosphorylate p47(phox) and postulated to mediate the phosphorylation-dependent activation of NADPH oxidase by PA and 2) conventional, but not novel or atypical, isoforms of protein kinase C (PKC). In contrast, all classes of PKC isoforms could phosphorylate p47(phox). In a gel retardation assay both the phosphatidic acid-dependent kinase and conventional PKC isoforms phosphorylated all molecules of p22(phox). These findings suggest that phosphorylation of p22(phox) by conventional PKC and/or a novel PA-activated protein kinase regulates the activation/assembly of NADPH oxidase.
- Published
- 1999
33. Transcriptional activation of the murine CTP:phosphocholine cytidylyltransferase gene (Ctpct): combined action of upstream stimulatory and inhibitory cis-acting elements
- Author
-
Marica Bakovic, Dennis E. Vance, Wei Tang, Kristin A. Waite, and Ira Tabas
- Subjects
Transcriptional Activation ,Regulation of gene expression ,Base Sequence ,Activator (genetics) ,Molecular Sequence Data ,Cytidylyltransferase ,Response element ,Nuclear Proteins ,Promoter ,Cell Biology ,Biology ,Transfection ,Molecular biology ,Choline-phosphate cytidylyltransferase ,Mice ,AP-1 transcription factor ,Enhancer Elements, Genetic ,Animals ,Humans ,Choline-Phosphate Cytidylyltransferase ,Promoter Regions, Genetic ,Molecular Biology ,Transcription factor ,Cells, Cultured ,HeLa Cells - Abstract
CTP:phosphocholine cytidylyltransferase plays a key role in regulating the rate of phosphatidylcholine biosynthesis. However, the proximal regulatory elements for the gene (Ctpct) that encode this enzyme and the cognate transcription factors involved have not been characterized. Ctpct promoter activities were deduced from promoter deletion constructs linked to a luciferase reporter and transiently transfected into C3H10T1/2 and McArdle RH7777 cells. Positive regulatory elements were located between -130 and -52 bp from the transcription start site. Basal expression resided downstream between -52 and +38 bp. DNase I protection and electromobility-shift assays indicated that Sp1-related nuclear factors bind to a stimulatory, a possible inhibitory and minimal promoter element. Gel-shift assays confirmed that all three regulatory regions bound Sp1. Sp1 was further implicated when Sp1-deficient Drosophila cells were co-transfected with promoter-reporter constructs and an Sp1 construct. DNase I assays also indicated that the Ap1 binding elements could be occupied in the proximal activator and minimal promoter regions. Gel-shift assays demonstrated that the distal activator region could bind Ap1 and an unknown transcription factor. We conclude that Sp1, Ap1 and an unknown transcription factor have important roles in regulating expression of the Ctpct gene.
- Published
- 1999
34. Phosphatidic Acid-mediated Phosphorylation of the NADPH Oxidase Component p47-phox
- Author
-
Diane Qualliotine-Mann, Reidar Wallin, Linda C. McPhail, and Kristin A. Waite
- Subjects
biology ,MAP kinase kinase kinase ,Cyclin-dependent kinase 2 ,Cell Biology ,Mitogen-activated protein kinase kinase ,Biochemistry ,MAP2K7 ,Cell biology ,biology.protein ,ASK1 ,Cyclin-dependent kinase 9 ,Protein kinase A ,Molecular Biology ,cGMP-dependent protein kinase - Abstract
Phosphatidic acid (PA), generated by phospholipase D activation, has been linked to the activation of the neutrophil respiratory burst enzyme, NADPH oxidase; however, the intracellular enzyme targets for PA remain unclear. We have recently shown (McPhail, L. C., Qualliotine-Mann, D., and Waite, K. A. (1995) Proc. Natl. Acad. Sci. U. S. A. 92, 7931-7935) that a PA-activated protein kinase is involved in the activation of NADPH oxidase in a cell-free system. This protein kinase phosphorylates numerous endogenous proteins, including p47-phox, a component of the NADPH oxidase complex. Phospholipids other than PA were less effective at inducing endogenous protein phosphorylation. Several of these endogenous substrates were also phosphorylated during stimulation of intact cells by opsonized zymosan, an agonist that induces phospholipase D activation. We sought to identify the PA-activated protein kinase that phosphorylates p47-phox. The PA-dependent protein kinase was shown to be cytosolic. cis-Unsaturated fatty acids were poor inducers of protein kinase activity, suggesting that the PA-activated protein kinase is not a fatty acid-regulated protein kinase (e.g. protein kinase N). Chromatographic techniques separated the PA-activated protein kinase from a number of other protein kinases known to be activated by PA or to phosphorylate p47-phox. These included isoforms of protein kinase C, p21 (Cdc42/Rac)-activated protein kinase, and mitogen-activated protein kinase. Gel filtration chromatography indicated that the protein kinase has an apparent molecular size of 125 kDa. Screening of cytosolic fractions from several cell types and rat brain suggested the enzyme has widespread cell and tissue distribution. The partially purified protein kinase was sensitive to the same protein kinase inhibitors that diminished NADPH oxidase activation and was independent of guanosine 5'-3-O-(thio)triphosphate and Ca2+. Phosphoamino acid analysis showed that serine and tyrosine residues were phosphorylated on p47-phox by this kinase(s). These data indicate that one or more potentially novel protein kinases are targets for PA in neutrophils and other cell types. Furthermore, a PA-activated protein kinase is likely to be an important regulator of the neutrophil respiratory burst by phosphorylation of the NADPH oxidase component p47-phox.
- Published
- 1997
35. Major Vault Protein
- Author
-
Takeo Minaguchi and Kristin A. Waite
- Published
- 2011
36. Cost-effective method for growing three-dimensional cell cultures in extracellular matrix extract
- Author
-
Charis Eng, Kristin A. Waite, and Avi Jacob
- Subjects
Matrigel ,Histology ,Chemistry ,Gasket ,Cost-Benefit Analysis ,Cell Culture Techniques ,Nanotechnology ,General Medicine ,Cell Line ,Culture Media ,Extracellular Matrix ,Extracellular matrix ,Medical Laboratory Technology ,Cell culture ,Animals ,Biomedical engineering - Abstract
Three-dimensional (3-D) cell cultures are advantageous for modeling complex cellular behavior. A major issue with 3-D cultures grown in Engelbreth-Holm-Swarm (EHS) extracellular matrix extract, however, is the expense. We describe here a simple method using a rubber gasket laid over a coverslip, which provides sufficient 3-D area filled with a minimal amount of EHS, resulting in cost-effective 3-D cellular cultures that are easy to manipulate.
- Published
- 2008
37. Germline Mutations and Variants in the Succinate Dehydrogenase Genes in Cowden and Cowden-like Syndromes
- Author
-
Petra Platzer, Kristin A. Waite, Kevin Zbuk, Mohammed S. Orloff, Emily A Edelman, Charis Eng, Ying Ni, Attila Patócs, Tammy Sadler, and Glenn P. Lobo
- Subjects
0303 health sciences ,Mutation ,biology ,SDHB ,Thyroid ,Cowden syndrome ,medicine.disease ,medicine.disease_cause ,Germline ,Article ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Germline mutation ,030220 oncology & carcinogenesis ,Genetics ,medicine ,biology.protein ,Cancer research ,PTEN ,Genetics(clinical) ,SDHD ,Genetics (clinical) ,030304 developmental biology - Abstract
Individuals with PTEN mutations have Cowden syndrome (CS), associated with breast, thyroid, and endometrial neoplasias. Many more patients with features of CS, not meeting diagnostic criteria (termed CS-like), are evaluated by clinicians for CS-related cancer risk. Germline mutations in succinate dehydrogenase subunits SDHB-D cause pheochromocytoma-paraganglioma syndrome. One to five percent of SDHB/SDHD mutation carriers have renal cell or papillary thyroid carcinomas, which are also CS-related features. SDHB-D may be candidate susceptibility genes for some PTEN mutation-negative individuals with CS-like cancers. To address this hypothesis, germline SDHB-D mutation analysis in 375 PTEN mutation-negative CS/CS-like individuals was performed, followed by functional analysis of identified SDH mutations/variants. Of 375 PTEN mutation-negative CS/CS-like individuals, 74 (20%) had increased manganese superoxide dismutase (MnSOD) expression, a manifestation of mitochondrial dysfunction. Among these, 10 (13.5%) had germline mutations/variants in SDHB (n = 3) or SDHD (7), not found in 700 controls (p < 0.001). Compared to PTEN mutation-positive CS/CS-like individuals, those with SDH mutations/variants were enriched for carcinomas of the female breast (6/9 SDH versus 30/107 PTEN, p < 0.001), thyroid (5/10 versus 15/106, p < 0.001), and kidney (2/10 versus 4/230, p = 0.026). In the absence of PTEN alteration, CS/CS-like-related SDH mutations/variants show increased phosphorylation of AKT and/or MAPK, downstream manifestations of PTEN dysfunction. Germline SDH mutations/variants occur in a subset of PTEN mutation-negative CS/CS-like individuals and are associated with increased frequencies of breast, thyroid, and renal cancers beyond those conferred by germline PTEN mutations. SDH testing should be considered for germline PTEN mutation-negative CS/CS-like individuals, especially in the setting of breast, thyroid, and/or renal cancers.
- Published
- 2008
38. The nuclear affairs of PTEN
- Author
-
Kristin A. Waite, Charis Eng, and Sarah M. Planchon
- Subjects
DNA Repair ,DNA repair ,Active Transport, Cell Nucleus ,Biology ,medicine.disease_cause ,Models, Biological ,Mice ,Chromosomal Instability ,Neoplasms ,medicine ,PTEN ,Animals ,Humans ,PI3K/AKT/mTOR pathway ,Cell Nucleus ,Mutation ,Cell Cycle ,PTEN Phosphohydrolase ,Cell Biology ,Cell cycle ,Protein Structure, Tertiary ,Cell nucleus ,medicine.anatomical_structure ,Cancer research ,biology.protein ,Signal transduction ,Nuclear localization sequence ,Signal Transduction - Abstract
PTEN encodes a major tumor-suppressor protein that is a dual-specificity phosphatase. Inactivation of PTEN has been shown to be involved in heritable and sporadic cancers. Mutation or deletion of PTEN, historically the most commonly identified mechanisms of inactivation of tumor suppressors, is found only in the minority of sporadic non-cultured primary cancers, which indicates that there might be other, novel mechanisms of inactivation. Despite the absence of a classic nuclear localization signal, PTEN enters the nucleus by several mechanisms, including simple diffusion, active shuttling, cytoplasmic-localization-signal-dependent export and monoubiquitylation-dependent import. Cytoplasmic PTEN has a well-known role as a negative regulator of the PI3K/AKT pathway; however, it is becoming clear that cytosolic PTEN is not the same as nuclear PTEN. Nuclear PTEN plays a role in chromosome stability, DNA repair, cell cycle arrest and cellular stability. The balance between these functions is an important factor in determining whether a cell remains benign or becomes neoplastic.
- Published
- 2008
39. Comparative genomic and functional analyses reveal a novel cis-acting PTEN regulatory element as a highly conserved functional E-box motif deleted in Cowden syndrome
- Author
-
Marcus G. Pezzolesi, Charis Eng, Kristin A. Waite, and Kevin Zbuk
- Subjects
Transcriptional Activation ,Blotting, Western ,DNA Mutational Analysis ,Molecular Sequence Data ,Oligonucleotides ,E-box ,Electrophoretic Mobility Shift Assay ,Biology ,Regulatory Sequences, Nucleic Acid ,Germline ,Conserved sequence ,E-Box Elements ,Proto-Oncogene Proteins c-myc ,Exon ,Germline mutation ,Cell Line, Tumor ,Sequence Homology, Nucleic Acid ,Genetics ,medicine ,PTEN ,Humans ,Promoter Regions, Genetic ,Molecular Biology ,Genetics (clinical) ,Base Sequence ,PTEN Phosphohydrolase ,Promoter ,General Medicine ,Cowden syndrome ,medicine.disease ,Mutation ,biology.protein ,Upstream Stimulatory Factors ,Hamartoma Syndrome, Multiple ,HeLa Cells ,Protein Binding - Abstract
Germline mutations in PTEN, encoding a phosphatase on 10q23, cause Cowden syndrome (CS) and Bannayan-Riley-Ruvalcaba syndrome (BRRS). Approximately, 10% of CS-related PTEN mutations occur in the PTEN promoter and 11% of BRRS-related mutations include large deletions, often favoring the gene's 5' end (exon 1, promoter). In order to better understand the mechanism(s) underlying the deregulation of PTEN in these syndromes, it is important that functional cis-regulatory elements be identified. We employed a comparative genomic approach combined with molecular genetic techniques to identify a highly conserved sequence upstream of the PTEN promoter, sharing 80% sequence identity among Homo sapiens, Mus musculus and Rattus norvegicus. Within this region, we identified a canonical E-box sequence (CACGTG) located at position -2181 to -2176, approximately 800 bp upstream of the PTEN core promoter and more than 1.1 kb upstream of its minimal promoter region (located at -958 to -821). In vitro assays suggest that this motif is recognized by members of the basic region-helix-loop-helix-leucine-zipper (bHLH-LZ) transcription factor family, USF1 and USF2, and reporter assays indicate that this novel E-box is involved in mediating PTEN transcriptional activation. Four of 30 CS/CS-like patients, without previously identified PTEN mutations, were found with germline deletions of the E-box element. Of the four, three had deletions stretching to exon 1, but not 3' of it; importantly, one classic CS patient harbored a germline deletion localizing to this E-box region, further affirming the role of this element in PTEN's regulation and deregulation, and its contribution to the pathogenesis of CS.
- Published
- 2007
40. Phytoestrogen exposure elevates PTEN levels
- Author
-
Charis Eng, Kristin A. Waite, and Michelle R. Sinden
- Subjects
MAPK/ERK pathway ,medicine.medical_specialty ,Tumor suppressor gene ,Blotting, Western ,Genistein ,Phytoestrogens ,chemistry.chemical_compound ,Cyclin D1 ,Internal medicine ,Cell Line, Tumor ,Genetics ,medicine ,PTEN ,Humans ,RNA, Messenger ,Phosphorylation ,Molecular Biology ,Protein kinase B ,Genetics (clinical) ,DNA Primers ,biology ,Base Sequence ,Reverse Transcriptase Polymerase Chain Reaction ,PTEN Phosphohydrolase ,food and beverages ,General Medicine ,Endocrinology ,chemistry ,Lipid phosphatase activity ,biology.protein ,Cancer research ,Mitogen-Activated Protein Kinases - Abstract
Epidemiological data suggest that consumption of phytoestrogens can be protective against the development of breast cancer. It may be logical to postulate that phytoestrogens may regulate proteins that control cellular division, such as the tumor suppressor PTEN. Germline, and more significantly, somatic PTEN mutations have been observed in a broad range of human cancers, especially those of the breast. Active PTEN results in decreased phosphorylation of Akt and MAPK, the up-regulation of p27 and down-regulation of cyclin D1 protein levels resulting in decreased proliferation and an increase in apoptosis. We hypothesized that phytoestrogen exposure regulates PTEN protein expression in the breast cancer cell line, MCF-7. When MCF-7 cells were stimulated with resveratrol, quercetin or genistein, there was an increase in PTEN protein levels. Concomitantly, phytoestrogen stimulation resulted in decreased Akt phosphorylation and an increase in p27 protein levels, indicating active PTEN lipid phosphatase activity. In contrast, we found that MAPK phosphorylation and cyclin D1 levels, which are regulated by PTEN's protein phosphatase activity, were not altered. Using semi-quantitative RT-PCR, we found that mRNA levels were slightly increased in cells stimulated by phytoestrogens, suggesting that the mechanism for increased PTEN protein expression is dependent upon transcription. Concurrently, our data provide evidence that a mechanism for phytoestrogens' protective nature is partially through increased PTEN expression. More importantly, it provides a novel target for the regulation of PTEN expression and suggests that dietary changes may be adjunctive to traditional preventive and therapeutic strategies against breast cancer.
- Published
- 2005
41. From developmental disorder to heritable cancer: it's all in the BMP/TGF-beta family
- Author
-
Charis Eng and Kristin A. Waite
- Subjects
Genetics ,Mutation ,Bone Diseases, Developmental ,biology ,Cancer ,ACVRL1 ,Cowden syndrome ,medicine.disease ,Bone morphogenetic protein ,medicine.disease_cause ,Transforming Growth Factor beta ,Neoplasms ,Bone Morphogenetic Proteins ,medicine ,Cancer research ,biology.protein ,Tensin ,PTEN ,Animals ,Humans ,Molecular Biology ,Genetics (clinical) ,Transforming growth factor - Abstract
Transforming growth factor-beta (TGF-beta) regulates many cellular processes through complex signal-transduction pathways that have crucial roles in normal development. Disruption of these pathways can lead to a range of diseases, including cancer. Mutations in the genes that encode members of the TGF-beta pathway are involved in vascular diseases as well as gastrointestinal neoplasia. More recently, they have been implicated in Cowden syndrome, which is normally associated with mutations in the phosphatase and tensin homologue gene PTEN. Molecular studies of TGF-beta signalling are now showing why mutations in genes that encode components of this pathway result in inherited cancer and developmental diseases.
- Published
- 2003
42. Enhanced expression and activation of CTP:phosphocholine cytidylyltransferase beta2 during neurite outgrowth
- Author
-
Dennis E. Vance, Jean E. Vance, Kristin A. Waite, Robert B. Campenot, and Jodi M. Carter
- Subjects
Neurite ,Cellular differentiation ,Cytidylyltransferase ,MAP Kinase Kinase 2 ,MAP Kinase Kinase 1 ,Gene Expression ,Tretinoin ,Biology ,Mitogen-activated protein kinase kinase ,Protein Serine-Threonine Kinases ,Biochemistry ,PC12 Cells ,Choline-phosphate cytidylyltransferase ,chemistry.chemical_compound ,Mice ,Neuroblastoma ,Cytosol ,Nerve Growth Factor ,Nitriles ,Butadienes ,Neurites ,Tumor Cells, Cultured ,Animals ,Choline-Phosphate Cytidylyltransferase ,RNA, Messenger ,Enzyme Inhibitors ,Molecular Biology ,Phosphocholine ,Mitogen-Activated Protein Kinase Kinases ,Neurons ,Kinase ,Reverse Transcriptase Polymerase Chain Reaction ,Cell Membrane ,Cell Differentiation ,Cell Biology ,Protein-Tyrosine Kinases ,Molecular biology ,Cell biology ,Rats ,Enzyme Activation ,Isoenzymes ,Nerve growth factor ,nervous system ,chemistry ,Phosphatidylcholines - Abstract
During differentiation neurons increase phospholipid biosynthesis to provide new membrane for neurite growth. We studied the regulation of phosphatidylcholine (PC) biosynthesis during differentiation of two neuronal cell lines: PC12 cells and Neuro2a cells. We hypothesized that in PC12 cells nerve growth factor (NGF) would up-regulate the activity and expression of the rate-limiting enzyme in PC biosynthesis, CTP:phosphocholine cytidylyltransferase (CT). During neurite outgrowth, NGF doubled the amount of cellular PC and CT activity. CTbeta2 mRNA increased within 1 day of NGF application, prior to the formation of visible neurites, and continued to increase during neurite growth. When neurites retracted in response to NGF withdrawal, CTbeta2 mRNA, protein, and CT activity decreased. NGF specifically activated CTbeta2 by promoting its translocation from cytosol to membranes. In contrast, NGF did not alter CTalpha expression or translocation. The increase in both CTbeta2 mRNA and CT activity was inhibited by U0126, an inhibitor of mitogen-activated kinase/extracellular signal-regulated kinase kinase 1/2 (MEK1/2). In Neuro2a cells, retinoic acid significantly increased CT activity (by 54%) and increased CTbeta2 protein, coincident with neurite outgrowth but did not change CTalpha expression. Together, these data suggest that the CTbeta2 isoform of CT is specifically up-regulated and activated during neuronal differentiation to increase PC biosynthesis for growing neurites.
- Published
- 2003
43. Dimethylethanolamine does not prevent liver failure in phosphatidylethanolamine N-methyltransferase-deficient mice fed a choline-deficient diet
- Author
-
Dennis E. Vance and Kristin A. Waite
- Subjects
medicine.medical_specialty ,Apolipoprotein B ,Phosphatidylethanolamine N-Methyltransferase ,Choline ,chemistry.chemical_compound ,Mice ,Phosphatidylcholine ,Internal medicine ,Deficient mouse ,medicine ,Animals ,Molecular Biology ,Dimethylethanolamine ,biology ,Liver failure ,Deanol ,Cell Biology ,Methyltransferases ,Diet ,Mice, Inbred C57BL ,Endocrinology ,chemistry ,Biochemistry ,Phosphatidylethanolamine N-methyltransferase ,biology.protein ,Apolipoprotein A1 ,Liver Failure - Abstract
Mice that lack phosphatidylethanolamine-N-methyltransferase (PEMT) and are fed a choline-deficient (CD) diet suffer severe liver damage and do not survive. Since phosphatidyldimethylethanolamine (PDME) has physical properties similar to those of phosphatidylcholine (PC), we hypothesized that dimethylethanolamine (DME) would be converted into PDME that might substitute for PC, and therefore abrogate the liver damage in the Pemt -/- mice fed a CD diet. We fed Pemt -/- mice either a CD diet, a CD diet supplemented with choline, or a CD diet supplemented with DME (CD + DME). Pemt -/- mice fed the CD diet developed severe liver failure by 4 days while CD + DME-fed mice developed severe liver failure by 5 days. The hepatic PC level in choline-supplemented (CS) mice was 67 +/- 4 nmol/mg protein, whereas the PC content was reduced in CD- and CD + DME-fed mice (49 +/- 3 and 30 +/- 3 nmol/mg protein, respectively). Upon supplementation of the CD diet with DME the amount of hepatic PDME was 81 +/- 9 nmol/mg protein so that the hepatic content of PC + PDME combined was 111 nmol/mg protein. Moreover, plasma apolipoprotein B100 and Al levels were markedly lower in mice fed the CD + DME diet compared to mice fed the CS diet, as was the plasma content of PC. Thus, despite replacement of the deficit in hepatic PC with PDME in Pemt -/- mice fed a CD diet, normal liver function was not restored. We conclude that although PC and PDME exhibit similar physical properties, the three methyl groups of choline are required for hepatic function in mice.
- Published
- 2003
44. BMP2 exposure results in decreased PTEN protein degradation and increased PTEN levels
- Author
-
Kristin A, Waite and Charis, Eng
- Subjects
Protein Synthesis Inhibitors ,Time Factors ,Dose-Response Relationship, Drug ,Tumor Suppressor Proteins ,Blotting, Western ,PTEN Phosphohydrolase ,Bone Morphogenetic Protein 2 ,Breast Neoplasms ,Syndrome ,Protein Serine-Threonine Kinases ,Precipitin Tests ,Phosphoric Monoester Hydrolases ,Ligases ,Cell Movement ,Transforming Growth Factor beta ,Bone Morphogenetic Proteins ,Mutation ,Ubiquitin-Conjugating Enzymes ,Tumor Cells, Cultured ,Humans ,Genetic Predisposition to Disease ,Receptors, Growth Factor ,Cycloheximide ,Bone Morphogenetic Protein Receptors, Type I ,Cell Division - Abstract
The tumour suppressor gene PTEN encodes a dual-specificity phosphatase that recognizes protein and phosphatidylinositiol substrates and modulates cellular functions such as migration and proliferation. Germline mutations of PTEN have been shown to cause Cowden syndrome, Bannayan-Riley-Ruvalcaba syndrome and Proteus syndrome. Recently, germline mutations in BMPR1A, the gene encoding the type 1A receptor of bone morphogenetic proteins (BMP) have been found in rare families with Cowden syndrome, suggesting that there may be a link between BMP signaling and PTEN. We thus sought to determine whether BMP2 stimulation alters PTEN protein levels in the breast cancer line, MCF-7. We found that exposure to BMP2 increased PTEN protein levels in a time- and dose-dependent manner. The increase in PTEN protein was rapid and was not due to an increase in new protein synthesis, as cycloheximide treatment did not inhibit BMP2-induced PTEN accumulation, suggesting that BMP2 stimulation inhibited PTEN protein degradation. Indeed, we found that BMP2 treatment of MCF-7 cells decreased the association of PTEN with two proteins in the degradative pathway, UbCH7 and UbC9. These data indicate that BMP2 exposure can regulate PTEN protein levels by decreasing PTEN's association with the degradative pathway. This opens up a new mode of regulating PTEN activity to be investigated further and may explain why BMPR1A can act as a minor susceptibility gene for PTEN mutation negative Cowden syndrome.
- Published
- 2003
45. Oncogenic Ha-Ras transformation modulates the transcription of the CTP:phosphocholine cytidylyltransferase alpha gene via p42/44MAPK and transcription factor Sp3
- Author
-
Marica Bakovic, Dennis E. Vance, and Kristin A. Waite
- Subjects
Gene isoform ,Transcription, Genetic ,Biology ,Oncogene Protein p21(ras) ,Biochemistry ,Transcription Factor Sp3 ,chemistry.chemical_compound ,Mice ,Transformation, Genetic ,Sp3 transcription factor ,Transcription (biology) ,Transcriptional regulation ,Animals ,Protein Isoforms ,Choline-Phosphate Cytidylyltransferase ,Phosphorylation ,Promoter Regions, Genetic ,Molecular Biology ,Cell Line, Transformed ,Mitogen-Activated Protein Kinase 1 ,Mitogen-Activated Protein Kinase 3 ,Tyrosine phosphorylation ,Cell Biology ,Transfection ,3T3 Cells ,Molecular biology ,DNA-Binding Proteins ,Sp3 Transcription Factor ,chemistry ,Gene Expression Regulation ,Signal transduction ,Mitogen-Activated Protein Kinases ,Signal Transduction ,Transcription Factors - Abstract
We have shown previously that expression of the murine CTP:phosphocholine cytidylyltransferase (CT) alpha gene is regulated during cell proliferation (Golfman, L. S., Bakovic, M., and Vance, D. E. (2001) J. Biol. Chem. 276, 43688-43692). We have now characterized the role of Ha-Ras in the transcriptional regulation of the CTalpha gene. The expression of CTalpha and CTbeta2 proteins and mRNAs was stimulated in C3H10T1/2 murine fibroblasts expressing oncogenic Ha-Ras. Incubation of cells with the specific inhibitor (PD98059) of p42/44(MAPK) decreased the expression of both CT isoforms. Transfection of fibroblasts with CTalpha promoter-luciferase constructs resulted in an approximately 2-fold enhanced luciferase expression in Ha-Ras-transformed, compared with nontransformed, fibroblasts. Electromobility shift assays indicated enhanced binding of the Sp3 transcription factor to the CTalpha promoter in Ha-Ras-transformed cells. Expression of several forms of Sp3 was increased in nuclear extracts of Ha-Ras-transformed fibroblasts compared with nontransformed cells. Tyrosine phosphorylation of one Sp3 form was decreased, whereas phosphorylation of two other forms of Sp3 was increased in nuclear extracts of Ha-Ras-transformed cells. When control fibroblasts were transfected with a Sp3-expressing plasmid, an enhanced expression of CTalpha and CTbeta was observed. However, the expression of CTalpha or CTbeta was not increased in Ha-Ras-transformed cells transfected with a Sp3 plasmid presumably because expression was already maximally enhanced. The results suggest that Sp3 is a downstream effector of a Ras/p42/44(MAPK) signaling pathway which increases CTalpha gene transcription.
- Published
- 2003
46. Choline deficiency-induced liver damage is reversible in Pemt(-/-) mice
- Author
-
Dennis E. Vance, Nora R. Cabilio, and Kristin A. Waite
- Subjects
medicine.medical_specialty ,food.ingredient ,Methyltransferase ,Phosphatidylethanolamine N-Methyltransferase ,Medicine (miscellaneous) ,Biology ,Lecithin ,Choline ,chemistry.chemical_compound ,Mice ,food ,Phosphatidylcholine ,Internal medicine ,medicine ,Animals ,Liver damage ,Transaminases ,Nutrition and Dietetics ,Choline Deficiency ,Methylation ,Methyltransferases ,Endocrinology ,chemistry ,Biochemistry ,Liver ,Phosphatidylcholines ,Choline chloride - Abstract
Hepatic tissue has two pathways for phosphatidylcholine (PC) synthesis, i.e., the cytidinediphosphocholine (CDP-choline) pathway and the methylation pathway, which utilizes phosphatidylethanolamine-N-methyltransferase (PEMT). Fatal liver damage occurs in Pemt -/- mice fed a choline-deficient (CD) diet. We investigated whether liver damage can be reversed by the addition of dietary choline. Mice (8 wk old) were fed the CD purified diet for 4 d, a choline-supplemented (CS) diet (CD diet + 0.4% choline chloride) for 4 d, or the CD diet for 3 d and a CS diet for 1 d (CD/CS). Pemt -/- mice fed the CD diet for 3 d exhibited liver damage as assayed by plasma aminotransferase levels. The livers appeared normal after subsequent feeding of the CS diet for 1 d (CD/CS). The activities of plasma aminotransferases of CD/CS fed mice were comparable to Pemt -/- mice fed the CS diet. Hepatic PC and triacylglycerol levels as well as plasma PC levels in the CD/CS-fed Pemt -/- mice were lower than those of mice fed the CD diet and began to approach normal levels. Although the CD diet induces liver damage in Pemt -/- mice, this damage can be rapidly reversed by the addition of dietary choline.
- Published
- 2002
47. A novel protein kinase target for the lipid second messenger phosphatidic acid
- Author
-
Kristin A. Waite, Debra S. Regier, Reidar Wallin, Susan Sergeant, Jennifer B. Nixon, Linda C. McPhail, Diane Qualliotine-Mann, and Wen-Xiao Zhang
- Subjects
Neutrophils ,Phosphatidic Acids ,Mitogen-activated protein kinase kinase ,Biology ,Second Messenger Systems ,MAP2K7 ,Cell Line ,Phospholipase D ,Animals ,Humans ,ASK1 ,Phosphorylation ,Protein kinase A ,Molecular Biology ,Respiratory Burst ,MAP kinase kinase kinase ,Cell-Free System ,Cyclin-dependent kinase 2 ,NADPH Dehydrogenase ,Membrane Transport Proteins ,NADPH Oxidases ,Cell Biology ,Phosphoproteins ,Cell biology ,Enzyme Activation ,Biochemistry ,biology.protein ,cGMP-dependent protein kinase ,Protein Kinases - Abstract
Activation of phospholipase D occurs in response to a wide variety of hormones, growth factors, and other extracellular signals. The initial product of phospholipase D, phosphatidic acid (PA), is thought to serve a signaling function, but the intracellular targets for this lipid second messenger are not clearly identified. The production of PA in human neutrophils is closely correlated with the activation of NADPH oxidase, the enzyme responsible for the respiratory burst. We have developed a cell-free system, in which the activation of NADPH oxidase is induced by the addition of PA. Characterization of this system revealed that a multi-functional cytosolic protein kinase was a target for PA, and that two NADPH oxidase components were substrates for the enzyme. Partial purification of the PA-activated protein kinase separated the enzyme from known protein kinase targets of PA. The partially purified enzyme was selectively activated by PA, compared to other phospholipids, and phosphorylated the oxidase component p47-phox on both serine and tyrosine residues. PA-activated protein kinase activity was present in a variety of hematopoietic cells and cell lines and in rat brain, suggesting it has widespread distribution. We conclude that this protein kinase may be a novel target for the second messenger function of PA.
- Published
- 1999
48. Cell-free activation of neutrophil NADPH oxidase by a phosphatidic acid-regulated protein kinase
- Author
-
Kristin A. Waite, Linda C. McPhail, and Diane Qualliotine-Mann
- Subjects
Indoles ,Neutrophils ,Phosphatidic Acids ,Granulomatous Disease, Chronic ,Substrate Specificity ,Diglycerides ,Maleimides ,Reference Values ,Humans ,Protein phosphorylation ,NADH, NADPH Oxidoreductases ,Phosphorylation ,Protein kinase A ,Protein Kinase Inhibitors ,Protein kinase C ,Protein Kinase C ,Diacylglycerol kinase ,NADPH dehydrogenase ,Multidisciplinary ,NADPH oxidase ,biology ,Cell-Free System ,Phospholipase D ,Kinase ,NADPH Dehydrogenase ,NADPH Oxidases ,Phosphoproteins ,Cell biology ,Enzyme Activation ,Kinetics ,Biochemistry ,biology.protein ,Protein Kinases ,Research Article - Abstract
The phosphorylation-dependent mechanisms regulating activation of the human neutrophil respiratory-burst enzyme, NADPH oxidase, have not been elucidated. We have shown that phosphatidic acid (PA) and diacylglycerol (DG), products of phospholipase activation, synergize to activate NADPH oxidase in a cell-free system. We now report that activation by PA plus DG involves protein kinase activity, unlike other cell-free system activators. NADPH oxidase activation by PA plus DG is reduced approximately 70% by several protein kinase inhibitors [1-(5-isoquinolinesulfonyl)piperazine, staurosporine, GF-109203X]. Similarly, depletion of ATP by dialysis reduces PA plus DG-mediated NADPH oxidase activation by approximately 70%. Addition of ATP, but not a nonhydrolyzable ATP analog, to the dialyzed system restores activation levels to normal. In contrast, these treatments have little effect on NADPH oxidase activation by arachidonic acid or SDS plus DG. PA plus DG induces the phosphorylation of a number of endogenous proteins. Phosphorylation is largely mediated by PA, not DG. A predominant substrate is p47-phox, a phosphoprotein component of NADPH oxidase. Phosphorylation of p47-phox precedes activation of NADPH oxidase and is markedly reduced by the protein kinase inhibitors. In contrast, arachidonic acid alone or SDS plus DG is a poor activator of protein phosphorylation in the cell-free system. Thus, PA induces activation of one or more protein kinases that regulate NADPH oxidase activation in a cell-free system. This cell-free system will be useful for identifying a functionally important PA-activated protein kinase(s) and for dissecting the phosphorylation-dependent mechanisms responsible for NADPH oxidase activation.
- Published
- 1995
49. Germline PTEN Promoter Mutations and Deletions in Cowden/Bannayan-Riley-Ruvalcaba Syndrome Result in Aberrant PTEN Protein and Dysregulation of the Phosphoinositol-3-Kinase/Akt Pathway
- Author
-
Donna Russo, Cindy Bos, Charis Eng, Kristin A. Waite, Mary Ella M Pierpont, Ellen T. Matloff, Robert Pilarski, Lois A. Greenberg, Magali Fernandez, Gerald L. Feldman, Xiao Ping Zhou, Annette R. Patterson, Heather Hampel, Majed Dasouki, Najah T. Nassif, and Jennifer Ivanovich
- Subjects
Male ,medicine.disease_cause ,Polymerase Chain Reaction ,Germline ,Exon ,Phosphatidylinositol 3-Kinases ,0302 clinical medicine ,Bannayan–Riley–Ruvalcaba syndrome ,Genetics(clinical) ,Promoter Regions, Genetic ,Genetics (clinical) ,Sequence Deletion ,Genetics & Heredity ,0303 health sciences ,Mutation ,biology ,Cowden syndrome ,Syndrome ,Exons ,Protein-Serine-Threonine Kinases ,3. Good health ,030220 oncology & carcinogenesis ,Uterine Neoplasms ,Female ,Genotype ,Breast Neoplasms ,Protein Serine-Threonine Kinases ,03 medical and health sciences ,Germline mutation ,Report ,Proto-Oncogene Proteins ,medicine ,Genetics ,PTEN ,Humans ,Thyroid Neoplasms ,PI3K/AKT/mTOR pathway ,Germ-Line Mutation ,030304 developmental biology ,Polymorphism, Genetic ,Base Sequence ,Tumor Suppressor Proteins ,PTEN Phosphohydrolase ,DNA ,medicine.disease ,Molecular biology ,Phosphoric Monoester Hydrolases ,Case-Control Studies ,Cancer research ,biology.protein ,Hamartoma Syndrome, Multiple ,Proto-Oncogene Proteins c-akt - Abstract
Germline intragenic mutations in PTEN are associated with 80% of patients with Cowden syndrome (CS) and 60% of patients with Bannayan-Riley-Ruvalcaba syndrome (BRRS). The underlying genetic causes remain to be determined in a considerable proportion of classic CS and BRRS without a polymerase chain reaction (PCR)-detectable PTEN mutation. We hypothesized that gross gene deletions and mutations in the PTEN promoter might alternatively account for a subset of apparently mutation-negative patients with CS and BRRS. Using real time and multiplex PCR techniques, we identified three germline hemizygous PTEN deletions in 122 apparently mutation-negative patients with classic CS (N=95) or BRRS (N=27). Fine mapping suggested that one deletion encompassed the whole gene and the other two included exon 1 and encompassed exons 1–5 of PTEN, respectively. Two patients with the deletion were diagnosed with BRRS, and one patient with the deletion was diagnosed with BRRS/CS overlap (features of both). Thus 3 (11%) of 27 patients with BRRS or BRRS/CS-overlap had PTEN deletions. Analysis of the PTEN promoter revealed nine cases (7.4%) harboring heterozygous germline mutations. All nine had classic CS, representing almost 10% of all subjects with CS. Eight had breast cancers and/or benign breast tumors but, otherwise, oligo-organ involvement. PTEN protein analysis, from one deletion-positive and five PTEN-promoter-mutation–positive samples, revealed a 50% reduction in protein and multiple bands of immunoreactive protein, respectively. In contrast, control samples showed only the expected band. Further, an elevated level of phosphorylated Akt was detected in the five promoter-mutation–positive samples, compared with controls, indicating an absence of or marked reduction in functional PTEN. These data suggest that patients with BRRS and CS without PCR-detected intragenic PTEN mutations be offered clinical deletion analysis and promoter-mutation analysis, respectively.
- Full Text
- View/download PDF
50. Protean PTEN: Form and Function
- Author
-
Charis Eng and Kristin A. Waite
- Subjects
MAPK/ERK pathway ,Tumor suppressor gene ,Review Article ,Proteus Syndrome ,Mice ,Phosphatidylinositol 3-Kinases ,Structure-Activity Relationship ,03 medical and health sciences ,0302 clinical medicine ,Germline mutation ,Genetics ,medicine ,Animals ,Humans ,PTEN ,Genetics(clinical) ,Protein kinase B ,Germ-Line Mutation ,Genetics (clinical) ,PI3K/AKT/mTOR pathway ,030304 developmental biology ,0303 health sciences ,biology ,Kinase ,Tumor Suppressor Proteins ,PTEN Phosphohydrolase ,Cowden syndrome ,medicine.disease ,Phosphoric Monoester Hydrolases ,Disease Models, Animal ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Hamartoma Syndrome, Multiple ,Signal Transduction - Abstract
Germline mutations distributed across the PTEN tumor-suppressor gene have been found to result in a wide spectrum of phenotypic features. Originally shown to be a major susceptibility gene for both Cowden syndrome (CS), which is characterized by multiple hamartomas and an increased risk of breast, thyroid, and endometrial cancers, and Bannayan-Riley-Ruvalcaba syndrome, which is characterized by lipomatosis, macrocephaly, and speckled penis, the PTEN hamartoma tumor syndrome spectrum has broadened to include Proteus syndrome and Proteus-like syndromes. Exon 5, which encodes the core motif, is a hotspot for mutations likely due to the biology of the protein. PTEN is a major lipid 3-phosphatase, which signals down the PI3 kinase/AKT pro-apoptotic pathway. Furthermore, PTEN is a protein phosphatase, with the ability to dephosphorylate both serine and threonine residues. The protein-phosphatase activity has also been shown to regulate various cell-survival pathways, such as the mitogen-activated kinase (MAPK) pathway. Although it is well established that PTEN’s lipid-phosphatase activity, via the PI3K/AKT pathway, mediates growth suppression, there is accumulating evidence that the protein-phosphatase/MAPK pathway is equally important in the mediation of growth arrest and other crucial cellular functions.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.