97 results on '"Ruth Birbe"'
Search Results
2. PARP‐1 regulates DNA repair factor availability
- Author
-
Matthew J Schiewer, Amy C Mandigo, Nicolas Gordon, Fangjin Huang, Sanchaika Gaur, Renée de Leeuw, Shuang G Zhao, Joseph Evans, Sumin Han, Theodore Parsons, Ruth Birbe, Peter McCue, Christopher McNair, Saswati N Chand, Ylenia Cendon‐Florez, Peter Gallagher, Jennifer J McCann, Neermala Poudel Neupane, Ayesha A Shafi, Emanuela Dylgjeri, Lucas J Brand, Tapio Visakorpi, Ganesh V Raj, Costas D Lallas, Edouard J Trabulsi, Leonard G Gomella, Adam P Dicker, Wm. Kevin Kelly, Benjamin E Leiby, Beatrice Knudsen, Felix Y Feng, and Karen E Knudsen
- Subjects
DNA repair ,E2F1 ,PARP ,transcription ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract PARP‐1 holds major functions on chromatin, DNA damage repair and transcriptional regulation, both of which are relevant in the context of cancer. Here, unbiased transcriptional profiling revealed the downstream transcriptional profile of PARP‐1 enzymatic activity. Further investigation of the PARP‐1‐regulated transcriptome and secondary strategies for assessing PARP‐1 activity in patient tissues revealed that PARP‐1 activity was unexpectedly enriched as a function of disease progression and was associated with poor outcome independent of DNA double‐strand breaks, suggesting that enhanced PARP‐1 activity may promote aggressive phenotypes. Mechanistic investigation revealed that active PARP‐1 served to enhance E2F1 transcription factor activity, and specifically promoted E2F1‐mediated induction of DNA repair factors involved in homologous recombination (HR). Conversely, PARP‐1 inhibition reduced HR factor availability and thus acted to induce or enhance “BRCA‐ness”. These observations bring new understanding of PARP‐1 function in cancer and have significant ramifications on predicting PARP‐1 inhibitor function in the clinical setting.
- Published
- 2018
- Full Text
- View/download PDF
3. siRNA-Encapsulated Hybrid Nanoparticles Target Mutant K-ras and Inhibit Metastatic Tumor Burden in a Mouse Model of Lung Cancer
- Author
-
Maryna Perepelyuk, Olubunmi Shoyele, Ruth Birbe, Chellappagounder Thangavel, Yi Liu, Robert B. Den, Adam E. Snook, Bo Lu, and Sunday A. Shoyele
- Subjects
Therapeutics. Pharmacology ,RM1-950 - Abstract
There is an unmet need in the development of an effective therapy for mutant K-ras-expressing non-small-cell lung cancer (NSCLC). Although various small molecules have been evaluated, an effective therapy remains a dream. siRNAs have the potential to downregulate mutant K-ras both at the protein and mRNA levels. However, a safe and effective delivery of siRNAs to tumors remains a limitation to their translational application in the treatment of this highly debilitating disease. Here we developed a novel hybrid nanoparticle carrier for effective delivery of anti-mutant K-ras to NSCLC (AKSLHN). The ability of this treatment modality to regress lung tumors in mouse models was evaluated as a monotherapy or as a combination treatment with erlotinib. Further, the toxicity of this treatment modality to healthy tissues was evaluated, along with its ability to elicit immune/inflammatory reactions. The results suggest that this treatment modality is a promising prospect for the treatment of mutant K-ras-expressing NSCLC without any accompanying toxicity. However, further understanding of the cellular-level interaction between AHSLHN and erlotinib needs to be attained before this promising treatment modality can be brought to the bedside. Keywords: hybrid nanoparticles, siRNA, mutant K-ras, lung cancer, gene silencing, orthotopic model, human immunoglobulin G
- Published
- 2017
- Full Text
- View/download PDF
4. Potential Impact on Clinical Decision Making via a Genome-Wide Expression Profiling: A Case Report
- Author
-
Hyun Kim, Mohammed Alshalalfa, Jean Hoffman-Censits, Costas D. Lallas, Elai Davicioni, Jianqing Lin, Ruth Birbe, Nicholas Erho, Jonathan Lehrer, Hussam Al-Deen Ashab, Mandeep Takhar, Anders Olson, Lucia L.C. Lam, W. Kevin Kelly, Karen E. Knudsen, Chellappagounder Thangavel, Roland Seiler, Felix Y. Feng, Edward M. Schaeffer, Edouard J. Trabulsi, Leonard G. Gomella, Mark D. Hurwitz, Adam P. Dicker, and Robert B. Den
- Subjects
Prostate ,Neuroendocrine ,Genomics ,Diseases of the genitourinary system. Urology ,RC870-923 - Abstract
Management of men with prostate cancer is fraught with uncertainty as physicians and patients balance efficacy with potential toxicity and diminished quality of life. Utilization of genomics as a prognostic biomarker has improved the informed decision-making process by enabling more rationale treatment choices. Recently investigations have begun to determine whether genomic information from tumor transcriptome data can be used to impact clinical decision-making beyond prognosis. Here we discuss the potential of genomics to alter management of a patient who presented with high-risk prostate adenocarcinoma. We suggest that this information help selecting patients for advanced imaging, chemotherapies, or clinical trial.
- Published
- 2016
- Full Text
- View/download PDF
5. Supplemental Table 2 from Therapeutic Challenge with a CDK 4/6 Inhibitor Induces an RB-Dependent SMAC-Mediated Apoptotic Response in Non–Small Cell Lung Cancer
- Author
-
Robert B. Den, Karen E. Knudsen, Adam P. Dicker, Joseph M. Salvino, Ruth Birbe, Sunday Shoyele, Adam Ertel, Sankar Addya, Anshul Bhardwaj, Maryna Perepelyuk, Alex Haber, Christopher McNair, Yi Liu, Ettickan Boopathi, and Chellappagounder Thangavel
- Abstract
Supplemental Table 2
- Published
- 2023
6. Supplemental Table 1 from RB Loss Promotes Prostate Cancer Metastasis
- Author
-
Robert B. Den, Karen E. Knudsen, Adam P. Dicker, Felix Y. Feng, Kenneth J. Pienta, Eva A. Turley, James B. McCarthy, Ruth Birbe, Shuang G. Zhao, James R. Hernandez, Matt Price, Chris McNair, Adam Snook, Jeffry L. Dean, Paolo Cotzia, Stephen J. Ciment, Noelle Williams, Sankar Addya, Anshul Bhardwaj, Adam Ertel, Alex Haber, Yi Liu, Ettickan Boopathi, and Chellappagounder Thangavel
- Abstract
nucleotide sequence
- Published
- 2023
7. Supplemental Figure 5 from The Retinoblastoma Tumor Suppressor Modulates DNA Repair and Radioresponsiveness
- Author
-
Robert B. Den, Karen E. Knudsen, Adam P. Dicker, Paolo Fortina, Ruth Birbe, Adam Ertel, Sankar Addya, Hestia Mellert, Steve B. McMahon, Ankur Sharma, Raymond O' Neill, Yi Liu, Steve Ciment, Ettickan Boopathi, and Chellappagounder Thangavel
- Abstract
Supplemental Figure 5. RB status is retained in biopsy proven local recurrence.
- Published
- 2023
8. Supplemental Figure Legend from Therapeutic Challenge with a CDK 4/6 Inhibitor Induces an RB-Dependent SMAC-Mediated Apoptotic Response in Non–Small Cell Lung Cancer
- Author
-
Robert B. Den, Karen E. Knudsen, Adam P. Dicker, Joseph M. Salvino, Ruth Birbe, Sunday Shoyele, Adam Ertel, Sankar Addya, Anshul Bhardwaj, Maryna Perepelyuk, Alex Haber, Christopher McNair, Yi Liu, Ettickan Boopathi, and Chellappagounder Thangavel
- Abstract
Supplemental data legends
- Published
- 2023
9. Data from Therapeutic Challenge with a CDK 4/6 Inhibitor Induces an RB-Dependent SMAC-Mediated Apoptotic Response in Non–Small Cell Lung Cancer
- Author
-
Robert B. Den, Karen E. Knudsen, Adam P. Dicker, Joseph M. Salvino, Ruth Birbe, Sunday Shoyele, Adam Ertel, Sankar Addya, Anshul Bhardwaj, Maryna Perepelyuk, Alex Haber, Christopher McNair, Yi Liu, Ettickan Boopathi, and Chellappagounder Thangavel
- Abstract
Purpose: The retinoblastoma tumor suppressor (RB), a key regulator of cell-cycle progression and proliferation, is functionally suppressed in up to 50% of non–small cell lung cancer (NSCLC). RB function is exquisitely controlled by a series of proteins, including the CyclinD–CDK4/6 complex. In this study, we interrogated the capacity of a CDK4/6 inhibitor, palbociclib, to activate RB function.Experimental Design and Results: We employed multiple isogenic RB-proficient and -deficient NSCLC lines to interrogate the cytostatic and cytotoxic capacity of CDK 4/6 inhibition in vitro and in vivo. We demonstrate that while short-term exposure to palbociclib induces cellular senescence, prolonged exposure results in inhibition of tumor growth. Mechanistically, CDK 4/6 inhibition induces a proapoptotic transcriptional program through suppression of IAPs FOXM1 and Survivin, while simultaneously augmenting expression of SMAC and caspase-3 in an RB-dependent manner.Conclusions: This study uncovers a novel function of RB activation to induce cellular apoptosis through therapeutic administration of a palbociclib and provides a rationale for the clinical evaluation of CDK 4/6 inhibitors in the treatment of patients with NSCLC. Clin Cancer Res; 24(6); 1402–14. ©2018 AACR.
- Published
- 2023
10. Supplemental Table 2 from RB Loss Promotes Prostate Cancer Metastasis
- Author
-
Robert B. Den, Karen E. Knudsen, Adam P. Dicker, Felix Y. Feng, Kenneth J. Pienta, Eva A. Turley, James B. McCarthy, Ruth Birbe, Shuang G. Zhao, James R. Hernandez, Matt Price, Chris McNair, Adam Snook, Jeffry L. Dean, Paolo Cotzia, Stephen J. Ciment, Noelle Williams, Sankar Addya, Anshul Bhardwaj, Adam Ertel, Alex Haber, Yi Liu, Ettickan Boopathi, and Chellappagounder Thangavel
- Abstract
in silico docking information
- Published
- 2023
11. Supplementary Figures from Therapeutic Challenge with a CDK 4/6 Inhibitor Induces an RB-Dependent SMAC-Mediated Apoptotic Response in Non–Small Cell Lung Cancer
- Author
-
Robert B. Den, Karen E. Knudsen, Adam P. Dicker, Joseph M. Salvino, Ruth Birbe, Sunday Shoyele, Adam Ertel, Sankar Addya, Anshul Bhardwaj, Maryna Perepelyuk, Alex Haber, Christopher McNair, Yi Liu, Ettickan Boopathi, and Chellappagounder Thangavel
- Abstract
Supplemental Figure 1. RB Regulates RB/E2F Target Gene Transcription and Restricts NSCLC Tumor Burden. Supplemental Figure 2. RB Promotes Cellular Senescence in NSCLC. Supplemental Figure 3. RB Dependent In Vitro Cellular Apoptosis in Response to CDK4/6 Inhibition in NSCLC. Supplemental Figure 4. Mitochondrial Protein SMAC Regulates NSCLC Apoptosis In Vitro. Supplemental Figure 5. Human Transcriptome Array Identifies FOXM1 and Survivin as RB/E2F Signatures. Supplemental Figure 6. RB Mediates Non-small Cell Lung Cancer Apoptosis via RB/IAP/SMAC/Caspase 3 signaling axis.
- Published
- 2023
12. Data from The Retinoblastoma Tumor Suppressor Modulates DNA Repair and Radioresponsiveness
- Author
-
Robert B. Den, Karen E. Knudsen, Adam P. Dicker, Paolo Fortina, Ruth Birbe, Adam Ertel, Sankar Addya, Hestia Mellert, Steve B. McMahon, Ankur Sharma, Raymond O' Neill, Yi Liu, Steve Ciment, Ettickan Boopathi, and Chellappagounder Thangavel
- Abstract
Purpose: Perturbations in the retinoblastoma pathway are over-represented in advanced prostate cancer; retinoblastoma loss promotes bypass of first-line hormone therapy. Conversely, preliminary studies suggested that retinoblastoma-deficient tumors may become sensitized to a subset of DNA-damaging agents. Here, the molecular and in vivo consequence of retinoblastoma status was analyzed in models of clinical relevance.Experimental Design: Experimental work was performed with multiple isogenic prostate cancer cell lines (hormone sensitive: LNCaP and LAPC4 cells and hormone resistant C42, 22Rv1 cells; stable knockdown of retinoblastoma using shRNA). Multiple mechanisms were interrogated including cell cycle, apoptosis, and DNA damage repair. Transcriptome analysis was performed, validated, and mechanisms discerned. Cell survival was measured using clonogenic cell survival assay and in vivo analysis was performed in nude mice with human derived tumor xenografts.Results: Loss of retinoblastoma enhanced the radioresponsiveness of both hormone-sensitive and castrate-resistant prostate cancer. Hypersensitivity to ionizing radiation was not mediated by cell cycle or p53. Retinoblastoma loss led to alteration in DNA damage repair and activation of the NF-κB pathway and subsequent cellular apoptosis through PLK3. In vivo xenografts of retinoblastoma-deficient tumors exhibited diminished tumor mass, lower PSA kinetics, and decreased tumor growth after treatment with ionizing radiation (P < 0.05).Conclusions: Loss of retinoblastoma confers increased radiosensitivity in prostate cancer. This hypersensitization was mediated by alterations in apoptotic signaling. Combined, these not only provide insight into the molecular consequence of retinoblastoma loss, but also credential retinoblastoma status as a putative biomarker for predicting response to radiotherapy. Clin Cancer Res; 20(21); 5468–82. ©2014 AACR.
- Published
- 2023
13. Supplemental Figures 1-5 from RB Loss Promotes Prostate Cancer Metastasis
- Author
-
Robert B. Den, Karen E. Knudsen, Adam P. Dicker, Felix Y. Feng, Kenneth J. Pienta, Eva A. Turley, James B. McCarthy, Ruth Birbe, Shuang G. Zhao, James R. Hernandez, Matt Price, Chris McNair, Adam Snook, Jeffry L. Dean, Paolo Cotzia, Stephen J. Ciment, Noelle Williams, Sankar Addya, Anshul Bhardwaj, Adam Ertel, Alex Haber, Yi Liu, Ettickan Boopathi, and Chellappagounder Thangavel
- Abstract
Supplemental Figure S1. RB loss promotes cell migration and invasion. Supplemental Figure S2. RB regulates RHAMM expression. Supplemental Figure S3. RB/E2F Binds on the RHAMM Promoter. Supplemental Figure S4. RHAMM overexpression drives cancer cell migration, invasion, and EMT. Supplemental Figure S5. RHAMM Inhibition Diminishes Migration and Invasion, but not Cellular Proliferation. Supplemental Figure S3. RB/E2F Binds on the RHAMM Promoter.
- Published
- 2023
14. Supplementary Table 1 from Therapeutic Challenge with a CDK 4/6 Inhibitor Induces an RB-Dependent SMAC-Mediated Apoptotic Response in Non–Small Cell Lung Cancer
- Author
-
Robert B. Den, Karen E. Knudsen, Adam P. Dicker, Joseph M. Salvino, Ruth Birbe, Sunday Shoyele, Adam Ertel, Sankar Addya, Anshul Bhardwaj, Maryna Perepelyuk, Alex Haber, Christopher McNair, Yi Liu, Ettickan Boopathi, and Chellappagounder Thangavel
- Abstract
supplementary Table 1
- Published
- 2023
15. Data from RB Loss Promotes Prostate Cancer Metastasis
- Author
-
Robert B. Den, Karen E. Knudsen, Adam P. Dicker, Felix Y. Feng, Kenneth J. Pienta, Eva A. Turley, James B. McCarthy, Ruth Birbe, Shuang G. Zhao, James R. Hernandez, Matt Price, Chris McNair, Adam Snook, Jeffry L. Dean, Paolo Cotzia, Stephen J. Ciment, Noelle Williams, Sankar Addya, Anshul Bhardwaj, Adam Ertel, Alex Haber, Yi Liu, Ettickan Boopathi, and Chellappagounder Thangavel
- Abstract
RB loss occurs commonly in neoplasia but its contributions to advanced cancer have not been assessed directly. Here we show that RB loss in multiple murine models of cancer produces a prometastatic phenotype. Gene expression analyses showed that regulation of the cell motility receptor RHAMM by the RB/E2F pathway was critical for epithelial–mesenchymal transition, motility, and invasion by cancer cells. Genetic modulation or pharmacologic inhibition of RHAMM activity was sufficient and necessary for metastatic phenotypes induced by RB loss in prostate cancer. Mechanistic studies in this setting established that RHAMM stabilized F-actin polymerization by controlling ROCK signaling. Collectively, our findings show how RB loss drives metastatic capacity and highlight RHAMM as a candidate therapeutic target for treating advanced prostate cancer. Cancer Res; 77(4); 982–95. ©2016 AACR.
- Published
- 2023
16. Supplemental Figure 2 from The Retinoblastoma Tumor Suppressor Modulates DNA Repair and Radioresponsiveness
- Author
-
Robert B. Den, Karen E. Knudsen, Adam P. Dicker, Paolo Fortina, Ruth Birbe, Adam Ertel, Sankar Addya, Hestia Mellert, Steve B. McMahon, Ankur Sharma, Raymond O' Neill, Yi Liu, Steve Ciment, Ettickan Boopathi, and Chellappagounder Thangavel
- Abstract
Supplemental Figure 2. RB deficiency promotes enhanced apoptotic pathway in response to ionizing radiation
- Published
- 2023
17. Supplementary Material and Methods from RB Loss Promotes Prostate Cancer Metastasis
- Author
-
Robert B. Den, Karen E. Knudsen, Adam P. Dicker, Felix Y. Feng, Kenneth J. Pienta, Eva A. Turley, James B. McCarthy, Ruth Birbe, Shuang G. Zhao, James R. Hernandez, Matt Price, Chris McNair, Adam Snook, Jeffry L. Dean, Paolo Cotzia, Stephen J. Ciment, Noelle Williams, Sankar Addya, Anshul Bhardwaj, Adam Ertel, Alex Haber, Yi Liu, Ettickan Boopathi, and Chellappagounder Thangavel
- Abstract
Supplemental Methods and Material
- Published
- 2023
18. Supplemental Figure 1 from The Retinoblastoma Tumor Suppressor Modulates DNA Repair and Radioresponsiveness
- Author
-
Robert B. Den, Karen E. Knudsen, Adam P. Dicker, Paolo Fortina, Ruth Birbe, Adam Ertel, Sankar Addya, Hestia Mellert, Steve B. McMahon, Ankur Sharma, Raymond O' Neill, Yi Liu, Steve Ciment, Ettickan Boopathi, and Chellappagounder Thangavel
- Abstract
Supplemental Figure 1. RB status does not impinge on the alterations of cell cycle after radiation
- Published
- 2023
19. Supplementary Figure Legend from RB Loss Promotes Prostate Cancer Metastasis
- Author
-
Robert B. Den, Karen E. Knudsen, Adam P. Dicker, Felix Y. Feng, Kenneth J. Pienta, Eva A. Turley, James B. McCarthy, Ruth Birbe, Shuang G. Zhao, James R. Hernandez, Matt Price, Chris McNair, Adam Snook, Jeffry L. Dean, Paolo Cotzia, Stephen J. Ciment, Noelle Williams, Sankar Addya, Anshul Bhardwaj, Adam Ertel, Alex Haber, Yi Liu, Ettickan Boopathi, and Chellappagounder Thangavel
- Abstract
Supplementary Figure Legends Supplemental Figure S1. RB loss promotes cell migration and invasion. Supplemental Figure S2. RB regulates RHAMM expression. Supplemental Figure S3. RB/E2F Binds on the RHAMM Promoter. Supplemental Figure S4. RHAMM overexpression drives cancer cell migration, invasion, and EMT. Supplemental Figure S5. RHAMM Inhibition Diminishes Migration and Invasion, but not Cellular Proliferation.
- Published
- 2023
20. Supplemental Figure 4 from The Retinoblastoma Tumor Suppressor Modulates DNA Repair and Radioresponsiveness
- Author
-
Robert B. Den, Karen E. Knudsen, Adam P. Dicker, Paolo Fortina, Ruth Birbe, Adam Ertel, Sankar Addya, Hestia Mellert, Steve B. McMahon, Ankur Sharma, Raymond O' Neill, Yi Liu, Steve Ciment, Ettickan Boopathi, and Chellappagounder Thangavel
- Abstract
Supplemental Figure 4. PLK3 expression promotes apoptotic cell death in the setting of RB loss.
- Published
- 2023
21. Supplemental Figure 3 from The Retinoblastoma Tumor Suppressor Modulates DNA Repair and Radioresponsiveness
- Author
-
Robert B. Den, Karen E. Knudsen, Adam P. Dicker, Paolo Fortina, Ruth Birbe, Adam Ertel, Sankar Addya, Hestia Mellert, Steve B. McMahon, Ankur Sharma, Raymond O' Neill, Yi Liu, Steve Ciment, Ettickan Boopathi, and Chellappagounder Thangavel
- Abstract
Supplemental Figure 3. NFκB is retained in the cytoplasm in the presence of an IκB dominant negative.
- Published
- 2023
22. Supplementary Figures 1 through 6 from Integrin αvβ6 Promotes an Osteolytic Program in Cancer Cells by Upregulating MMP2
- Author
-
Lucia R. Languino, Jane B. Lian, Gary S. Stein, Janet L. Stein, Shelia M. Violette, John Wixted, Ruth Birbe, Zhong Jiang, Thomas J. FitzGerald, Brad J. Zerlanko, Tao Wang, Jitesh Pratap, Jacqueline Akech, Huimin Lu, Jing Li, and Anindita Dutta
- Abstract
PDF - 529K, Expression of alphavbeta6 does not affect PC3-2 integrin profile (S1); alphavbeta6 promotes osteolysis in vivo (S2); alphavbeta6 increases osteolytic lesions and osteoclast recruitment in vivo (S3); Downregulation of alphavbeta6 reduces MMP2 levels in RWPE cells (S4); Exogenous expression of alphavbeta6 and alphavbeta3 in C4-2B transfectants (S5); MMP2 is co-expressed with alphavbeta6 in human prostate cancer osteolytic metastases (S6).
- Published
- 2023
23. Supplementary Methods from Integrin αvβ6 Promotes an Osteolytic Program in Cancer Cells by Upregulating MMP2
- Author
-
Lucia R. Languino, Jane B. Lian, Gary S. Stein, Janet L. Stein, Shelia M. Violette, John Wixted, Ruth Birbe, Zhong Jiang, Thomas J. FitzGerald, Brad J. Zerlanko, Tao Wang, Jitesh Pratap, Jacqueline Akech, Huimin Lu, Jing Li, and Anindita Dutta
- Abstract
PDF - 69K, Description of additional methods and procedures used in the study. Also includes supplementary references.
- Published
- 2023
24. Data from Integrin αvβ6 Promotes an Osteolytic Program in Cancer Cells by Upregulating MMP2
- Author
-
Lucia R. Languino, Jane B. Lian, Gary S. Stein, Janet L. Stein, Shelia M. Violette, John Wixted, Ruth Birbe, Zhong Jiang, Thomas J. FitzGerald, Brad J. Zerlanko, Tao Wang, Jitesh Pratap, Jacqueline Akech, Huimin Lu, Jing Li, and Anindita Dutta
- Abstract
The molecular circuitries controlling osseous prostate metastasis are known to depend on the activity of multiple pathways, including integrin signaling. Here, we demonstrate that the αvβ6 integrin is upregulated in human prostate cancer bone metastasis. In prostate cancer cells, this integrin is a functionally active receptor for fibronectin and latency-associated peptide-TGF-β1; it mediates attachment and migration upon ligand binding and is localized in focal contacts. Given the propensity of prostate cancer cells to form bone metastatic lesions, we investigated whether the αvβ6 integrin promotes this type of metastasis. We show for the first time that αvβ6 selectively induces matrix metalloproteinase 2 (MMP2) in vitro in multiple prostate cancer cells and promotes osteolysis in vivo in an immunodeficient mouse model of bone metastasis through upregulation of MMP2, but not MMP9. The effect of αvβ6 on MMP2 expression and activity is independent of androgen receptor in the analyzed prostate cancer cells. Increased levels of parathyroid hormone–related protein (PTHrP), known to induce osteoclastogenesis, were also observed in αvβ6-expressing cells. However, by using MMP2 short hairpin RNA, we demonstrate that the αvβ6 effect on bone loss is due to upregulation of soluble MMP2 by the cancer cells, not due to changes in tumor growth rate. Another related αv-containing integrin, αvβ5, fails to show similar responses, underscoring the significance of αvβ6 activity. Overall, these mechanistic studies establish that expression of a single integrin, αvβ6, contributes to the cancer cell—mediated program of osteolysis by inducing matrix degradation through MMP2. Our results open new prospects for molecular therapy for metastatic bone disease. Cancer Res; 74(5); 1598–608. ©2014 AACR.
- Published
- 2023
25. Bone Health Management in the Continuum of Prostate Cancer Disease
- Author
-
Ettickan, Boopathi, Ruth, Birbe, Sunday A, Shoyele, Robert B, Den, and Chellappagounder, Thangavel
- Subjects
Cancer Research ,Oncology - Abstract
Prostate cancer (PCa) is the second-leading cause of cancer-related deaths in men. PCa cells require androgen receptor (AR) signaling for their growth and survival. Androgen deprivation therapy (ADT) is the preferred treatment for patients with locally advanced and metastatic PCa disease. Despite their initial response to androgen blockade, most patients eventually will develop metastatic castration-resistant prostate cancer (mCRPC). Bone metastases are common in men with mCRPC, occurring in 30% of patients within 2 years of castration resistance and in >90% of patients over the course of the disease. Patients with mCRPC-induced bone metastasis develop lesions throughout their skeleton; the 5-year survival rate for these patients is 47%. Bone-metastasis-induced early changes in the bone that proceed the osteoblastic response in the bone matrix are monitored and detected via modern magnetic resonance and PET/CT imaging technologies. Various treatment options, such as targeting osteolytic metastasis with bisphosphonates, prednisone, dexamethasone, denosumab, immunotherapy, external beam radiation therapy, radiopharmaceuticals, surgery, and pain medications are employed to treat prostate-cancer-induced bone metastasis and manage bone health. However, these diagnostics and treatment options are not very accurate nor efficient enough to treat bone metastases and manage bone health. In this review, we present the pathogenesis of PCa-induced bone metastasis, its deleterious impacts on vital organs, the impact of metastatic PCa on bone health, treatment interventions for bone metastasis and management of bone- and skeletal-related events, and possible current and future therapeutic options for bone management in the continuum of prostate cancer disease.
- Published
- 2022
26. Increased expression of desmin and vimentin reduces bladder smooth muscle contractility via JNK2
- Author
-
Michael R. Ruggieri, Samuel Chacko, Chellappagounder Thangavel, Deepak A. Deshpande, Jagmohan Singh, Ettickan Boopathi, Joseph A. Hypolite, Stephen A. Zderic, Elham Javed, Ruth Birbe, Nagat Frara, Ipsita Mohanty, Robert B. Den, Alan S. Braverman, Satish Rattan, and Raymond B. Penn
- Subjects
0301 basic medicine ,Myosin light-chain kinase ,MAP Kinase Signaling System ,Urinary Bladder ,Vimentin ,macromolecular substances ,Biochemistry ,Article ,Desmin ,Contractility ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Genetics ,Animals ,Mitogen-Activated Protein Kinase 9 ,Intermediate filament ,Molecular Biology ,Mice, Knockout ,biology ,Chemistry ,Muscle, Smooth ,Smooth muscle contraction ,Cell biology ,030104 developmental biology ,Knockout mouse ,biology.protein ,Signal transduction ,030217 neurology & neurosurgery ,Biotechnology ,Muscle Contraction - Abstract
Bladder dysfunction is associated with the overexpression of the intermediate filament (IF) proteins desmin and vimentin in obstructed bladder smooth muscle (BSM). However, the mechanisms by which these proteins contribute to BSM dysfunction are not known. Previous studies have shown that desmin and vimentin directly participate in signal transduction. In this study, we hypothesized that BSM dysfunction associated with overexpression of desmin or vimentin is mediated via c-Jun N-terminal kinase (JNK). We employed a model of murine BSM tissue in which increased expression of desmin or vimentin was induced by adenoviral transduction to examine the sufficiency of increased IF protein expression to reduce BSM contraction. Murine BSM strips overexpressing desmin or vimentin generated less force in response to KCl and carbachol relative to the levels in control murine BSM strips, an effect associated with increased JNK2 phosphorylation and reduced myosin light chain (MLC20 ) phosphorylation. Furthermore, desmin and vimentin overexpressions did not alter BSM contractility and MLC20 phosphorylation in strips isolated from JNK2 knockout mice. Pharmacological JNK2 inhibition produced results qualitatively similar to those caused by JNK2 knockout. These findings suggest that inhibition of JNK2 may improve diminished BSM contractility associated with obstructive bladder disease.
- Published
- 2019
27. Therapeutic Challenge with a CDK 4/6 Inhibitor Induces an RB-Dependent SMAC-Mediated Apoptotic Response in Non–Small Cell Lung Cancer
- Author
-
Yi Liu, Alex Haber, Maryna Perepelyuk, Robert B. Den, Ettickan Boopathi, Christopher McNair, Karen E. Knudsen, Sunday A. Shoyele, Joseph M. Salvino, Anshul Bhardwaj, Adam Ertel, Chellappagounder Thangavel, Adam P. Dicker, Sankar Addya, and Ruth Birbe
- Subjects
0301 basic medicine ,Cancer Research ,Lung Neoplasms ,Cell Survival ,Apoptosis ,Palbociclib ,Retinoblastoma Protein ,Article ,Mitochondrial Proteins ,Mice ,03 medical and health sciences ,Cyclin-dependent kinase ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Survivin ,medicine ,Animals ,Humans ,Protein Kinase Inhibitors ,Cell Proliferation ,biology ,Cyclin-dependent kinase 4 ,Retinoblastoma ,Chemistry ,Cell Cycle ,Intracellular Signaling Peptides and Proteins ,Retinoblastoma protein ,Cyclin-Dependent Kinase 4 ,Cyclin-Dependent Kinase 6 ,Cell cycle ,medicine.disease ,Xenograft Model Antitumor Assays ,Tumor Burden ,Gene Expression Regulation, Neoplastic ,Disease Models, Animal ,030104 developmental biology ,Oncology ,biology.protein ,Cancer research ,Cyclin-dependent kinase 6 ,Apoptosis Regulatory Proteins ,Reactive Oxygen Species ,Biomarkers - Abstract
Purpose: The retinoblastoma tumor suppressor (RB), a key regulator of cell-cycle progression and proliferation, is functionally suppressed in up to 50% of non–small cell lung cancer (NSCLC). RB function is exquisitely controlled by a series of proteins, including the CyclinD–CDK4/6 complex. In this study, we interrogated the capacity of a CDK4/6 inhibitor, palbociclib, to activate RB function. Experimental Design and Results: We employed multiple isogenic RB-proficient and -deficient NSCLC lines to interrogate the cytostatic and cytotoxic capacity of CDK 4/6 inhibition in vitro and in vivo. We demonstrate that while short-term exposure to palbociclib induces cellular senescence, prolonged exposure results in inhibition of tumor growth. Mechanistically, CDK 4/6 inhibition induces a proapoptotic transcriptional program through suppression of IAPs FOXM1 and Survivin, while simultaneously augmenting expression of SMAC and caspase-3 in an RB-dependent manner. Conclusions: This study uncovers a novel function of RB activation to induce cellular apoptosis through therapeutic administration of a palbociclib and provides a rationale for the clinical evaluation of CDK 4/6 inhibitors in the treatment of patients with NSCLC. Clin Cancer Res; 24(6); 1402–14. ©2018 AACR.
- Published
- 2018
28. RB Loss Promotes Prostate Cancer Metastasis
- Author
-
Jeffry L. Dean, Karen E. Knudsen, Stephen J. Ciment, Robert B. Den, James Hernandez, Alex Haber, Paolo Cotzia, Ettickan Boopathi, Shuang G. Zhao, Sankar Addya, Felix Y. Feng, Matthew Price, Ruth Birbe, Chellappagounder Thangavel, Adam P. Dicker, Noelle L. Williams, Yi Liu, Adam E. Snook, Christopher McNair, James B. McCarthy, Eva A. Turley, Kenneth J. Pienta, Adam Ertel, and Anshul Bhardwaj
- Subjects
Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Motility ,Biology ,Retinoblastoma Protein ,Article ,Metastasis ,Mice ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Cell Line, Tumor ,Internal medicine ,medicine ,Animals ,Humans ,Neoplasm Metastasis ,E2F ,Extracellular Matrix Proteins ,rho-Associated Kinases ,Cyclin-Dependent Kinase 4 ,Prostatic Neoplasms ,Cancer ,Cyclin-Dependent Kinase 6 ,medicine.disease ,Hyaluronan-mediated motility receptor ,Phenotype ,Actins ,E2F Transcription Factors ,Hyaluronan Receptors ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer cell ,Signal Transduction - Abstract
RB loss occurs commonly in neoplasia but its contributions to advanced cancer have not been assessed directly. Here we show that RB loss in multiple murine models of cancer produces a prometastatic phenotype. Gene expression analyses showed that regulation of the cell motility receptor RHAMM by the RB/E2F pathway was critical for epithelial–mesenchymal transition, motility, and invasion by cancer cells. Genetic modulation or pharmacologic inhibition of RHAMM activity was sufficient and necessary for metastatic phenotypes induced by RB loss in prostate cancer. Mechanistic studies in this setting established that RHAMM stabilized F-actin polymerization by controlling ROCK signaling. Collectively, our findings show how RB loss drives metastatic capacity and highlight RHAMM as a candidate therapeutic target for treating advanced prostate cancer. Cancer Res; 77(4); 982–95. ©2016 AACR.
- Published
- 2017
29. NF-κB and GATA-Binding Factor 6 Repress Transcription of Caveolins in Bladder Smooth Muscle Hypertrophy
- Author
-
Stephen A. Zderic, Cristiano Mendes Gomes, Samuel Chacko, Deepak A. Deshpande, Chellappagounder Thangavel, Sankar Addya, Elham Javed, Raymond B. Penn, Ruth Birbe, Sreya Das, Robert B. Den, Ettickan Boopathi, Satish Rattan, and Jagmohan Singh
- Subjects
0301 basic medicine ,Male ,Transcription, Genetic ,Prostatic Hyperplasia ,030204 cardiovascular system & hematology ,Caveolins ,Article ,Pathology and Forensic Medicine ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Transcription (biology) ,GATA6 Transcription Factor ,Gene expression ,Animals ,Humans ,Aged ,Regulation of gene expression ,Gene knockdown ,Chemistry ,Gene Expression Profiling ,NF-kappa B ,Promoter ,Muscle, Smooth ,Smooth muscle contraction ,Hypertrophy ,Middle Aged ,Cell biology ,Mice, Inbred C57BL ,Urinary Bladder Neck Obstruction ,030104 developmental biology ,Gene Expression Regulation ,Smooth muscle hypertrophy ,Chromatin immunoprecipitation ,Biomarkers ,Muscle Contraction - Abstract
Caveolins (CAVs) are structural proteins of caveolae that function as signaling platforms to regulate smooth muscle contraction. Loss of CAV protein expression is associated with impaired contraction in obstruction-induced bladder smooth muscle (BSM) hypertrophy. In this study, microarray analysis of bladder RNA revealed down-regulation of CAV1, CAV2, and CAV3 gene transcription in BSM from models of obstructive bladder disease in mice and humans. We identified and characterized regulatory regions responsible for CAV1, CAV2, and CAV3 gene expression in mice with obstruction-induced BSM hypertrophy, and in men with benign prostatic hyperplasia. DNA affinity chromatography and chromatin immunoprecipitation assays revealed a greater increase in binding of GATA-binding factor 6 (GATA-6) and NF-κB to their cognate binding motifs on CAV1, CAV2, and CAV3 promoters in obstructed BSM relative to that observed in control BSM. Knockout of NF-κB subunits, shRNA-mediated knockdown of GATA-6, or pharmacologic inhibition of GATA-6 and NF-κB in BSM increased CAV1, CAV2, and CAV3 transcription and promoter activity. Conversely, overexpression of GATA-6 decreased CAV2 and CAV3 transcription and promoter activity. Collectively, these data provide new insight into the mechanisms by which CAV gene expression is repressed in hypertrophied BSM in obstructive bladder disease.
- Published
- 2019
30. Potential Impact on Clinical Decision Making via a Genome-Wide Expression Profiling: A Case Report
- Author
-
Edouard J. Trabulsi, Mohammed Alshalalfa, Edward M. Schaeffer, Hyun Kim, Hussam Al-Deen Ashab, Anders Olson, Mark D. Hurwitz, Jonathan Lehrer, Lucia L.C. Lam, Felix Y. Feng, Adam P. Dicker, Elai Davicioni, Chellappagounder Thangavel, Ruth Birbe, Karen E. Knudsen, W. Kevin Kelly, Costas D. Lallas, Roland Seiler, Nicholas Erho, Leonard G. Gomella, Jean H. Hoffman-Censits, Jianqing Lin, Mandeep Takhar, and Robert B. Den
- Subjects
0301 basic medicine ,Urologic Diseases ,Aging ,Urology ,Clinical Trials and Supportive Activities ,Genomics ,Bioinformatics ,lcsh:RC870-923 ,Transcriptome ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Rare Diseases ,Prostate ,Clinical Research ,Behavioral and Social Science ,medicine ,Genetics ,Profiling (information science) ,Genome wide expression ,Cancer ,Potential impact ,screening and diagnosis ,business.industry ,Prostate Cancer ,Human Genome ,Health Services ,medicine.disease ,lcsh:Diseases of the genitourinary system. Urology ,Clinical trial ,Detection ,030104 developmental biology ,medicine.anatomical_structure ,Neuroendocrine ,Oncology ,030220 oncology & carcinogenesis ,Patient Safety ,business ,Biotechnology ,4.2 Evaluation of markers and technologies - Abstract
Management of men with prostate cancer is fraught with uncertainty as physicians and patients balance efficacy with potential toxicity and diminished quality of life. Utilization of genomics as a prognostic biomarker has improved the informed decision-making process by enabling more rationale treatment choices. Recently investigations have begun to determine whether genomic information from tumor transcriptome data can be used to impact clinical decision-making beyond prognosis. Here we discuss the potential of genomics to alter management of a patient who presented with high-risk prostate adenocarcinoma. We suggest that this information help selecting patients for advanced imaging, chemotherapies, or clinical trial.
- Published
- 2016
- Full Text
- View/download PDF
31. Suppression of progranulin expression inhibits bladder cancer growth and sensitizes cancer cells to cisplatin
- Author
-
Andrea Morrione, Igor Moskalev, Ruth Birbe, Alaide Morcavallo, Ryuta Tanimoto, Shi Qiong Xu, Leonard G. Gomella, Marco Genua, Renato V. Iozzo, Antonino Belfiore, Peter C. Black, Simone Buraschi, Stephen C. Peiper, and Manuela Stefanello
- Subjects
0301 basic medicine ,Pathology ,Nude ,Transgenic ,Small hairpin RNA ,Mice ,Progranulins ,Cell Movement ,RNA, Small Interfering ,Tumor ,anchorage-independent growth ,3. Good health ,Gene Expression Regulation, Neoplastic ,Phenotype ,motility ,Oncology ,bladder cancer ,Intercellular Signaling Peptides and Proteins ,Female ,Research Paper ,medicine.drug ,medicine.medical_specialty ,Cell Survival ,MAP Kinase Signaling System ,Mice, Nude ,Motility ,Mice, Transgenic ,Antineoplastic Agents ,Small Interfering ,Cell Line ,03 medical and health sciences ,Downregulation and upregulation ,Cell Line, Tumor ,tumor formation in vivo ,mental disorders ,Biomarkers, Tumor ,progranulin ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,Urothelium ,Cell Proliferation ,Cisplatin ,Neoplastic ,Bladder cancer ,Cell growth ,business.industry ,medicine.disease ,Actins ,030104 developmental biology ,Urinary Bladder Neoplasms ,Gene Expression Regulation ,Cancer cell ,Cancer research ,RNA ,Neoplasm Transplantation ,business ,Biomarkers - Abstract
We have recently demonstrated a critical role for progranulin in bladder cancer. Progranulin contributes, as an autocrine growth factor, to the transformed phenotype by modulating Akt-and MAPK-driven motility, invasion and anchorage-independent growth. Progranulin also induces F-actin remodeling by interacting with the F-actin binding protein drebrin. In addition, progranulin is overexpressed in invasive bladder cancer compared to normal tissue controls, suggesting that progranulin might play a key role in driving the transition to the invasive phenotype of urothelial cancer. However, it is not established whether targeting progranulin could have therapeutic effects on bladder cancer. In this study, we stably depleted urothelial cancer cells of endogenous progranulin by shRNA approaches and determined that progranulin depletion severely inhibited the ability of tumorigenic urothelial cancer cells to migrate, invade and grow in anchorage-independency. We further demonstrate that progranulin expression is critical for tumor growth in vivo, in both xenograft and orthotopic tumor models. Notably, progranulin levels correlated with response to cisplatin treatment and were upregulated in bladder tumors. Our data indicate that progranulin may constitute a novel target for therapeutic intervention in bladder tumors. In addition, progranulin may serve as a novel biomarker for bladder cancer.
- Published
- 2016
32. VPAC1 Targeted 64Cu-TP3805 Positron Emission Tomography Imaging of Prostate Cancer: Preliminary Evaluation in Man
- Author
-
Mathew L. Thakur, Ruth Birbe, Sung M. Kim, Edouard J. Trabulsi, Pardeep Kumar, Ashish Gandhe, Sushil K. Tripathi, Leonard G. Gomella, Charles M. Intenzo, and Peter McCue
- Subjects
Pathology ,medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,Prostatectomy ,Urology ,medicine.medical_treatment ,Cancer ,Hyperplasia ,medicine.disease ,030218 nuclear medicine & medical imaging ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,medicine.anatomical_structure ,Positron emission tomography ,Prostate ,030220 oncology & carcinogenesis ,Biopsy ,medicine ,Nuclear medicine ,business ,Lymph node - Abstract
Objective To evaluate 64 Cu-TP3805 as a novel biomolecule, to positron emission tomography (PET) image prostate cancer (PC), at the onset of which VPAC1, the superfamily of G protein-coupled receptors, is expressed in high density on PC cells, but not on normal cells. Materials and Methods Twenty-five patients undergoing radical prostatectomy were PET/X-ray computerized tomography imaged preoperatively with 64 Cu-TP3805. Standardized maximum uptake (SUVmax) values were determined and malignant lesions (standardized uptake value > 1.0) counted, and compared with histologic findings. Whole-mount pathology slides from 6 VPAC1 PET imaged patients, 3 benign prostatic hyperplasia patients, 1 malignant and 1 benign lymph node underwent digital autoradiography (DAR) after 64 Cu-TP3805 incubation and were compared to hematoxylin- and eosin-stained slides. Results In 25 patients who underwent PET imaging, 212 prostate gland lesions had SUVmax > 1.0 vs 127 lesions identified by histology of biopsy tissues. The status of the additional 85 PET identified prostate lesions remains to be determined. In 68 histologic slides from 6 PET imaged patients, DAR identified 105 of 107 PC foci, 19 of 19 high-grade prostatic intraepithelial neoplasias, and ejaculatory ducts and verumontanum involved with cancer. Additionally, DAR found 9 PC lesions not previously identified histologically. The positive and negative lymph nodes were correctly identified, and in 3 of 3 benign prostatic hyperplasia patients and 5 of 5 cysts, DAR was negative. Conclusion This feasibility study demonstrated that 64 Cu-TP3805 delineates PC in vivo and ex vivo, provided normal images for benign masses, and is worthy of further studies.
- Published
- 2016
33. Multicompartment metabolism in papillary thyroid cancer
- Author
-
Tingting Zhan, William M. Keane, Adam Luginbuhl, Edmund A. Pribitkin, John Sprandio, Ruth Birbe, Madalina Tuluc, Ubaldo E. Martinez-Outschoorn, Patrick Tassone, Marina Domingo-Vidal, Joseph Curry, David Cognetti, Mehri Mollaee, and Paolo Cotzia
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Goiter ,endocrine system diseases ,Adenoma ,business.industry ,Thyroid ,Cancer ,medicine.disease ,Papillary thyroid cancer ,Metastasis ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Otorhinolaryngology ,030220 oncology & carcinogenesis ,medicine ,Carcinoma ,Immunohistochemistry ,business - Abstract
Objectives/Hypothesis In many cancers, varying regions within the tumor are often phenotypically heterogeneous, including their metabolic phenotype. Further, tumor regions can be metabolically compartmentalized, with metabolites transferred between compartments. When present, this metabolic coupling can promote aggressive behavior. Tumor metabolism in papillary thyroid cancer (PTC) is poorly characterized. Study Design Immunohistochemical staining of tissue samples. Methods Papillary thyroid cancer specimens from 46 patients with (n = 19) and without advanced disease (n = 27) were compared to noncancerous thyroid tissue (NCT) and benign thyroid specimens (n = 6 follicular adenoma [FA] and n = 5 nodular goiter [NG]). Advanced disease was defined as the presence of lateral neck lymphadenopathy. Immunohistochemistry was performed for translocase of outer mitochondrial membrane 20 (TOMM20), a marker of oxidative phosphorylation, and monocarboxylate transporter 4 (MCT4), a marker of glycolysis. Results Papillary thyroid cancer and FA thyrocytes had high staining for TOMM20 compared to NCT and nodular goiter (NG) (P < 0.01). High MCT4 staining in fibroblasts was more common in PTC with advanced disease than in any other tissue type studied (P < 0.01). High MCT4 staining was found in all 19 cases of PTC with advanced disease, in 11 of 19 samples with low-stage disease, in one of five samples of FA, in one of 34 NCT, and in 0 of six NG samples. Low fibroblast MCT4 staining in PTC correlated with the absence of clinical adenopathy (P = 0.028); the absence of extrathyroidal extension (P = 0.004); low American Thyroid Association risk (P = 0.001); low AGES (age, grade, extent, size) score (P = 0.004); and low age, metastasis, extent of disease, size risk (P = 0.002). Conclusion This study suggests that multiple metabolic compartments exist in PTC, and low fibroblast MCT4 may be a biomarker of indolent disease. Level of Evidence N/A. Laryngoscope, 2015
- Published
- 2015
34. PARP‐1 regulates DNA repair factor availability
- Author
-
Ylenia Cendon-Florez, Edouard J. Trabulsi, Neermala Poudel Neupane, Benjamin E. Leiby, Felix Y. Feng, Saswati N. Chand, Sumin Han, Matthew J. Schiewer, Sanchaika Gaur, Christopher McNair, Lucas J. Brand, Shuang G. Zhao, Amy C. Mandigo, Emanuela Dylgjeri, Peter A. McCue, Ayesha A. Shafi, Ganesh V. Raj, Renée de Leeuw, Beatrice S. Knudsen, Karen E. Knudsen, Costas D. Lallas, Tapio Visakorpi, Nicolas Gordon, Theodore Parsons, Leonard G. Gomella, Peter Gallagher, Ruth Birbe, Wm. Kevin Kelly, Jennifer J. McCann, Fangjin Huang, Joseph R. Evans, and Adam P. Dicker
- Subjects
Male ,0301 basic medicine ,Medicine (General) ,DNA repair ,Poly ADP ribose polymerase ,Poly (ADP-Ribose) Polymerase-1 ,QH426-470 ,Biology ,Cell Line ,PARP ,Transcriptome ,03 medical and health sciences ,R5-920 ,0302 clinical medicine ,Genetics ,Transcriptional regulation ,Animals ,Humans ,E2F1 ,Homologous Recombination ,Research Articles ,Cancer ,Mice, Inbred BALB C ,Gene Expression Profiling ,Prostatic Neoplasms ,E2F1 Transcription Factor ,Immunohistochemistry ,3. Good health ,Chromatin ,Cell biology ,030104 developmental biology ,Tissue Array Analysis ,030220 oncology & carcinogenesis ,Disease Progression ,Molecular Medicine ,transcription ,Homologous recombination ,Research Article - Abstract
PARP‐1 holds major functions on chromatin, DNA damage repair and transcriptional regulation, both of which are relevant in the context of cancer. Here, unbiased transcriptional profiling revealed the downstream transcriptional profile of PARP‐1 enzymatic activity. Further investigation of the PARP‐1‐regulated transcriptome and secondary strategies for assessing PARP‐1 activity in patient tissues revealed that PARP‐1 activity was unexpectedly enriched as a function of disease progression and was associated with poor outcome independent of DNA double‐strand breaks, suggesting that enhanced PARP‐1 activity may promote aggressive phenotypes. Mechanistic investigation revealed that active PARP‐1 served to enhance E2F1 transcription factor activity, and specifically promoted E2F1‐mediated induction of DNA repair factors involved in homologous recombination (HR). Conversely, PARP‐1 inhibition reduced HR factor availability and thus acted to induce or enhance “BRCA‐ness”. These observations bring new understanding of PARP‐1 function in cancer and have significant ramifications on predicting PARP‐1 inhibitor function in the clinical setting.
- Published
- 2018
35. Vasodilator-Stimulated Phosphoprotein Biomarkers Are Associated with Invasion and Metastasis in Colorectal Cancer
- Author
-
Giovanni Pitari, Frank A. Sinicrope, Alessandro Bombonati, Juan P. Palazzo, Ruth Birbe, David S. Zuzga, Anthony P Shuber, Mehboob Ali, Charalambos C. Solomides, Paolo Cotzia, and Wendy Rizzo
- Subjects
0301 basic medicine ,Microbiology (medical) ,actin cytoskeleton ,Adjuvant chemotherapy ,Colorectal cancer ,Immunology ,macromolecular substances ,lcsh:RC254-282 ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,Immunology and Allergy ,Medicine ,metastasis ,vasodilator-stimulated phosphoprotein ,business.industry ,Vasodilator-stimulated phosphoprotein ,Stage II Colorectal Cancer ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Actin cytoskeleton ,invasion ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,business ,Rapid Communication - Abstract
Background and Aims: The benefit of adjuvant chemotherapy for stage II colorectal cancer (CRC) patients remains unclear, emphasizing the need for improved prognostic biomarkers to identify patients at risk of metastatic recurrence. To address this unmet clinical need, we examined the expression and phosphorylation status of the vasodilator-stimulated phosphoprotein (VASP) in CRC tumor progression. VASP, a processive actin polymerase, promotes the formation of invasive membrane structures leading to extracellular matrix remodeling and tumor invasion. Phosphorylation of VASP serine (Ser) residues 157 and 239 regulate VASP function, directing subcellular localization and inhibiting actin polymerization, respectively. Methods: The expression levels of VASP protein, pSer157-VASP, and pSer239-VASP were determined by immunohistochemistry in tumors and matched normal adjacent tissue from 141 CRC patients, divided into 2 cohorts, and the association of VASP biomarker expression with clinicopathologic features and disease recurrence was examined. Results: We report that changes in VASP expression and phosphorylation were significantly associated with tumor invasion and disease recurrence. Furthermore, we disclose a novel 2-tiered methodology to maximize VASP positive and negative predictive value performance for prognostication. Conclusion: VASP biomarkers may serve as prognostic biomarkers in CRC and should be evaluated in a larger clinical study.
- Published
- 2018
36. Pilot study demonstrating metabolic and anti-proliferative effects of in vivo anti-oxidant supplementation with N-Acetylcysteine in Breast Cancer
- Author
-
Andrew B. Newberg, Ubaldo E. Martinez-Outschoorn, Federica Sotgia, Adam C. Berger, Zhao Lin, Ruth Birbe, Melissa Lazar, Madalina Tuluc, Rossitza Draganova-Tacheva, Daniela Monti, Michael P. Lisanti, Paolo Cotzia, Marina Domingo-Vidal, and Diana Whitaker-Menezes
- Subjects
0301 basic medicine ,Caveolin 1 ,Muscle Proteins ,Apoptosis ,Pilot Projects ,medicine.disease_cause ,0302 clinical medicine ,Tumor Microenvironment ,Medicine ,Mastectomy ,Carcinoma, Ductal, Breast ,Hematology ,Free Radical Scavengers ,Middle Aged ,Immunohistochemistry ,Neoadjuvant Therapy ,Treatment Outcome ,Oncology ,030220 oncology & carcinogenesis ,Female ,Adult ,Monocarboxylic Acid Transporters ,medicine.medical_specialty ,Stromal cell ,Breast Neoplasms ,Article ,03 medical and health sciences ,Breast cancer ,Internal medicine ,Carcinoma ,In Situ Nick-End Labeling ,Humans ,Cell Proliferation ,Neoplasm Staging ,Tumor microenvironment ,business.industry ,Cancer ,medicine.disease ,Carcinoma, Papillary ,Acetylcysteine ,030104 developmental biology ,Endocrinology ,Carcinoma, Intraductal, Noninfiltrating ,Ki-67 Antigen ,Cancer cell ,Cancer research ,Stromal Cells ,business ,Oxidative stress - Abstract
Background High oxidative stress as defined by hydroxyl and peroxyl activity is often found in the stroma of human breast cancers. Oxidative stress induces stromal catabolism, which promotes cancer aggressiveness. Stromal cells exposed to oxidative stress release catabolites such as lactate, which are up-taken by cancer cells to support mitochondrial oxidative phosphorylation. The transfer of catabolites between stromal and cancer cells leads to metabolic heterogeneity between these cells and increased cancer cell proliferation and reduced apoptosis in preclinical models. N-Acetylcysteine (NAC) is an antioxidant that reduces oxidative stress and reverses stromal catabolism and stromal-carcinoma cell metabolic heterogeneity, resulting in reduced proliferation and increased apoptosis of cancer cells in experimental models of breast cancer. The purpose of this clinical trial was to determine if NAC could reduce markers of stromal-cancer metabolic heterogeneity and markers of cancer cell aggressiveness in human breast cancer. Methods Subjects with newly diagnosed stage 0 and I breast cancer who were not going to receive neoadjuvant therapy prior to surgical resection were treated with NAC before definitive surgery to assess intra-tumoral metabolic markers. NAC was administered once a week intravenously at a dose of 150 mg/kg and 600 mg twice daily orally on the days not receiving intravenous NAC. Histochemistry for the stromal metabolic markers monocarboxylate transporter 4 (MCT4) and caveolin-1 (CAV1) and the Ki67 proliferation assay and TUNEL apoptosis assay in carcinoma cells were performed in pre- and post-NAC specimens. Results The range of days on NAC was 14–27 and the mean was 19 days. Post-treatment biopsies showed significant decrease in stromal MCT4 and reduced Ki67 in carcinoma cells. NAC did not significantly change stromal CAV1 and carcinoma TUNEL staining. NAC was well tolerated. Conclusions NAC as a single agent reduces MCT4 stromal expression, which is a marker of glycolysis in breast cancer with reduced carcinoma cell proliferation. This study suggests that modulating metabolism in the tumor microenvironment has the potential to impact breast cancer proliferation.
- Published
- 2017
37. MCT1 in Invasive Ductal Carcinoma: Monocarboxylate Metabolism and Aggressive Breast Cancer
- Author
-
Marina Domingo-Vidal, Zhao Lin, Jason Chen, Lekha Mikkilineni, Ruth Birbe, Daniele Colombo, Mehri Mollaee, Jennifer Johnson, Roberto Fratamico, Madalina Tuluc, Paolo Cotzia, Diana Whitaker-Menezes, Ubaldo E. Martinez-Outschoorn, Juan P. Palazzo, and Tingting Zhan
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,oxidative phosphorylation ,Estrogen receptor ,Biology ,03 medical and health sciences ,Cell and Developmental Biology ,0302 clinical medicine ,Breast cancer ,medicine ,tumor microenvironment ,Progression-free survival ,Triple-negative breast cancer ,Original Research ,Tumor microenvironment ,lactic acid ,Cell Biology ,glycolysis ,medicine.disease ,3. Good health ,030104 developmental biology ,Monocarboxylate transporter 1 ,030220 oncology & carcinogenesis ,triple negative breast cancer ,Cancer cell ,biology.protein ,Cancer research ,Immunohistochemistry ,Developmental Biology - Abstract
Introduction: Monocarboxylate transporter 1 (MCT1) is an importer of monocarboxylates such as lactate and pyruvate and a marker of mitochondrial metabolism. MCT1 is highly expressed in a subgroup of cancer cells to allow for catabolite uptake from the tumor microenvironment to support mitochondrial metabolism. We studied the protein expression of MCT1 in a broad group of breast invasive ductal carcinoma specimens to determine its association with breast cancer subtypes and outcomes. Methods: MCT1 expression was evaluated by immunohistochemistry on tissue micro-arrays (TMA) obtained through our tumor bank. Two hundred and fifty-seven cases were analyzed: 180 cases were estrogen receptor and/or progesterone receptor positive (ER+ and/or PR+), 62 cases were human epidermal growth factor receptor 2 positive (HER2+), and 56 cases were triple negative breast cancers (TNBC). MCT1 expression was quantified by digital pathology with Aperio software. The intensity of the staining was measured on a continuous scale (0-black to 255-bright white) using a co-localization algorithm. Statistical analysis was performed using a linear mixed model. Results: High MCT1 expression was more commonly found in TNBC compared to ER+ and/or PR+ and compared to HER-2+ (p < 0.001). Tumors with an in-situ component were less likely to stain strongly for MCT1 (p < 0.05). High nuclear grade was associated with higher MCT1 staining (p < 0.01). Higher T stage tumors were noted to have a higher expression of MCT1 (p < 0.05). High MCT1 staining in cancer cells was associated with shorter progression free survival, increased risk of recurrence, and larger size independent of TNBC status (p < 0.05). Conclusion: MCT1 expression, which is a marker of high catabolite uptake and mitochondrial metabolism, is associated with recurrence in breast invasive ductal carcinoma. MCT1 expression as quantified with digital image analysis may be useful as a prognostic biomarker and to design clinical trials using MCT1 inhibitors.
- Published
- 2017
- Full Text
- View/download PDF
38. Integrin αvβ6 Promotes an Osteolytic Program in Cancer Cells by Upregulating MMP2
- Author
-
Tao Wang, Jing Li, Jacqueline Akech, Lucia R. Languino, Thomas J. Fitzgerald, Jitesh Pratap, Ruth Birbe, Huimin Lu, Janet L. Stein, Anindita Dutta, Jane B. Lian, Zhong Jiang, Brad J. Zerlanko, Gary S. Stein, Shelia M. Violette, and John J. Wixted
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,MMP2 ,Osteolysis ,biology ,Chemistry ,Integrin ,Bone metastasis ,Cancer ,medicine.disease ,Metastasis ,Prostate cancer ,Oncology ,Cancer cell ,medicine ,biology.protein ,Cancer research - Abstract
The molecular circuitries controlling osseous prostate metastasis are known to depend on the activity of multiple pathways, including integrin signaling. Here, we demonstrate that the αvβ6 integrin is upregulated in human prostate cancer bone metastasis. In prostate cancer cells, this integrin is a functionally active receptor for fibronectin and latency-associated peptide-TGF-β1; it mediates attachment and migration upon ligand binding and is localized in focal contacts. Given the propensity of prostate cancer cells to form bone metastatic lesions, we investigated whether the αvβ6 integrin promotes this type of metastasis. We show for the first time that αvβ6 selectively induces matrix metalloproteinase 2 (MMP2) in vitro in multiple prostate cancer cells and promotes osteolysis in vivo in an immunodeficient mouse model of bone metastasis through upregulation of MMP2, but not MMP9. The effect of αvβ6 on MMP2 expression and activity is independent of androgen receptor in the analyzed prostate cancer cells. Increased levels of parathyroid hormone–related protein (PTHrP), known to induce osteoclastogenesis, were also observed in αvβ6-expressing cells. However, by using MMP2 short hairpin RNA, we demonstrate that the αvβ6 effect on bone loss is due to upregulation of soluble MMP2 by the cancer cells, not due to changes in tumor growth rate. Another related αv-containing integrin, αvβ5, fails to show similar responses, underscoring the significance of αvβ6 activity. Overall, these mechanistic studies establish that expression of a single integrin, αvβ6, contributes to the cancer cell—mediated program of osteolysis by inducing matrix degradation through MMP2. Our results open new prospects for molecular therapy for metastatic bone disease. Cancer Res; 74(5); 1598–608. ©2014 AACR.
- Published
- 2014
39. USP22 Regulates Oncogenic Signaling Pathways to Drive Lethal Cancer Progression
- Author
-
Jeffry L. Dean, Rossitza Draganova-Tacheva, Angelo M. DeMarzo, Ruth Birbe, Jonathan F. Goodwin, Steven B. McMahon, Matthew J. Schiewer, Karen E. Knudsen, Robyn T. Sussman, Timothy J. Stanek, Jessica L. Hicks, Randy S. Schrecengost, Tapio Visakorpi, and Mark W. Urban
- Subjects
Male ,Proteasome Endopeptidase Complex ,Cancer Research ,Cell Culture Techniques ,Gene Expression ,Mice, SCID ,Adenocarcinoma ,Biology ,Article ,Mice ,Cell Line, Tumor ,Androgen Receptor Antagonists ,Biomarkers, Tumor ,medicine ,Animals ,Humans ,Regulation of gene expression ,Effector ,Cancer ,medicine.disease ,Phenotype ,Gene Expression Regulation, Neoplastic ,Androgen receptor ,Disease Models, Animal ,Prostatic Neoplasms, Castration-Resistant ,Oncology ,Nuclear receptor ,Receptors, Androgen ,Tumor progression ,Disease Progression ,Cancer research ,Heterografts ,Thiolester Hydrolases ,Ubiquitin Thiolesterase - Abstract
Increasing evidence links deregulation of the ubiquitin-specific proteases 22 (USP22) deubitiquitylase to cancer development and progression in a select group of tumor types, but its specificity and underlying mechanisms of action are not well defined. Here we show that USP22 is a critical promoter of lethal tumor phenotypes that acts by modulating nuclear receptor and oncogenic signaling. In multiple xenograft models of human cancer, modeling of tumor-associated USP22 deregulation demonstrated that USP22 controls androgen receptor accumulation and signaling, and that it enhances expression of critical target genes coregulated by androgen receptor and MYC. USP22 not only reprogrammed androgen receptor function, but was sufficient to induce the transition to therapeutic resistance. Notably, in vivo depletion experiments revealed that USP22 is critical to maintain phenotypes associated with end-stage disease. This was a significant finding given clinical evidence that USP22 is highly deregulated in tumors, which have achieved therapeutic resistance. Taken together, our findings define USP22 as a critical effector of tumor progression, which drives lethal phenotypes, rationalizing this enzyme as an appealing therapeutic target to treat advanced disease. Cancer Res; 74(1); 272–86. ©2013 AACR.
- Published
- 2014
40. The International Society of Urological Pathology (ISUP) Vancouver Classification of Renal Neoplasia
- Author
-
Stephen Rohan, Neriman Gokden, Victor E. Reuter, Hemamali Samaratunga, Jonathan I. Epstein, George J. Netto, James G. Kench, Adeboye O. Osunkoya, David J. Griffiths, Holger Moch, Cynthia Cohen, Barbara Loftus, Kathrine Lie, Funda Vakar-Lopez, Henry Crist, Rajal B. Shah, Rodolfo Montironi, Isabel Trias, Omar Hameed, Mathilde Sibony, Jesse K. McKenney, Ferran Algaba, Ahmed Shabaik, Rose Miller, Anne Y. Warren, Robert W. Allan, Ai Ying Chuang, Marie O'Donnell, Cheng Wang, Masoumeh Ghayouri, Ming Zhou, Edward C. Jones, Michelle S. Hirsch, Giovanna A. Giannico, Sara M. Falzarano, Jonathan H Shanks, Liang Cheng, John R. Srigley, Cristina Magi-Galluzzi, Eddie Fridman, Maria Merino, Adebowale J. Adeniran, Ondrej Hes, Kiril Trpkov, Ruben Ronchetti, Nilesh S. Gupta, Peter Bethwaite, Anil V. Parwani, Fadi Brimo, Athanase Billis, Bungo Furusato, Asli Yilmaz, John C. Cheville, Chin Chen Pan, Puay Hoon Tan, Samson W. Fine, Yong Mee Cho, Hikmat Al Ahmadie, Mahul B. Amin, Hiroyuki Takahashi, Hiroshi Miyamoto, David J. Grignon, Satish K. Tickoo, Martin Susani, Constantina Petraki, Josep Lloreta, Guido Martignoni, Aysim Ozagari, David G. Bostwick, Peter A. Humphrey, Sundus Hussein, Brett Delahunt, Pedram Argani, Warick Delprado, Teresa McHale, Jiaoti Huang, Zhaoli Lane, Toyonori Tsuzuki, Ying-Bei Chen, Nathalie Rioux-Leclercq, Stewart Fleming, Laura Irene Jufe, Larry True, Masatoshi Kida, Steven S. Shen, Fiona Maclean, Debra L. Zynger, Brian D. Robinson, Jin Zhao, Jorge L. Yao, Oluwole Fadare, Dilek Ertoy Baydar, Joan Sweet, Katayoon Rezaei, Khalid Ahmed, Maria M. Picken, Masoud Ganji, Laurie Russell, Peter W. Nichols, Claudio Lewin, Antonio Lopez-Beltran, Wei Huang, Louis R. Bégin, Ruth Birbe, Bhuvana Srinivasan, Tipu Nazeer, Hedwig Murphy, Kenneth A. Iczkowski, Maria Pyda-Karwicka, Stephen M. Bonsib, John N. Nacey, Lars Egevad, Pheroze Tamboli, Joanna Perry-Keene, Andrew Evans, Rafael E. Jimenez, Helen P. Cathro, Glen Kristiansen, Ulrika Axcrona, Christina Hulsbergen Van De Kaa, Lakshmi P. Kunju, Daniel M. Berney, Sueli Suzigan, Fang Ming Deng, Marc Barry, Gabriella Nesi, Anila Abraham, John N. Eble, Semra Olgac, Marina Scarpelli, Roberto Orozco, Daniel A. Fajardo, Maria Shevchuk, Mathieu Latour, and Theo H. van der Kwast
- Subjects
Tubulocystic renal cell carcinoma ,Pathology ,medicine.medical_specialty ,business.industry ,Cystic nephroma ,Multilocular Cystic Renal Cell Carcinoma ,urologic and male genital diseases ,Clear cell papillary renal cell carcinoma ,medicine.disease ,Kidney Neoplasms ,female genital diseases and pregnancy complications ,Pathology and Forensic Medicine ,Mucinous tubular and spindle cell carcinoma ,Collecting duct carcinoma ,Renal cell carcinoma ,medicine ,Humans ,Surgery ,Hereditary leiomyomatosis and renal cell carcinoma ,Anatomy ,business ,Societies, Medical - Abstract
The classification working group of the International Society of Urological Pathology consensus conference on renal neoplasia was in charge of making recommendations regarding additions and changes to the current World Health Organization Classification of Renal Tumors (2004). Members of the group performed an exhaustive literature review, assessed the results of the preconference survey and participated in the consensus conference discussion and polling activities. On the basis of the above inputs, there was consensus that 5 entities should be recognized as new distinct epithelial tumors within the classification system: tubulocystic renal cell carcinoma (RCC), acquired cystic disease-associated RCC, clear cell (tubulo) papillary RCC, the MiT family translocation RCCs (in particular t(6;11) RCC), and hereditary leiomyomatosis RCC syndrome-associated RCC. In addition, there are 3 rare carcinomas that were considered as emerging or provisional new entities: thyroid-like follicular RCC; succinate dehydrogenase B deficiency-associated RCC; and ALK translocation RCC. Further reports of these entities are required to better understand the nature and behavior of these highly unusual tumors. There were a number of new concepts and suggested modifications to the existing World Health Organization 2004 categories. Within the clear cell RCC group, it was agreed upon that multicystic clear cell RCC is best considered as a neoplasm of low malignant potential. There was agreement that subtyping of papillary RCC is of value and that the oncocytic variant of papillary RCC should not be considered as a distinct entity. The hybrid oncocytic chromophobe tumor, which is an indolent tumor that occurs in 3 settings, namely Birt-Hogg-Dubé Syndrome, renal oncocytosis, and as a sporadic neoplasm, was placed, for the time being, within the chromophobe RCC category. Recent advances related to collecting duct carcinoma, renal medullary carcinoma, and mucinous spindle cell and tubular RCC were elucidated. Outside of the epithelial category, advances in our understanding of angiomyolipoma, including the epithelioid and epithelial cystic variants, were considered. In addition, the apparent relationship between cystic nephroma and mixed epithelial and stromal tumor was discussed, with the consensus that these tumors form a spectrum of neoplasia. Finally, it was thought that the synovial sarcoma should be removed from the mixed epithelial and mesenchymal category and placed within the sarcoma group. The new classification is to be referred to as the International Society of Urological Pathology Vancouver Classification of Renal Neoplasia.
- Published
- 2013
41. Handling and Staging of Renal Cell Carcinoma
- Author
-
John R. Srigley, Cristina Magi-Galluzzi, Asli Yilmaz, Antonio Lopez-Beltran, Omar Hameed, Anne Y. Warren, Jesse K. McKenney, Pedram Argani, Wei Huang, Ruth Birbe, Roberto Orozco, Toyonori Tsuzuki, Edward C. Jones, Gabriella Nesi, Robert W. Allan, Stewart Fleming, Dilek Ertoy Baydar, Anila Abraham, Aysim Ozagari, Victor E. Reuter, Barbara Loftus, Josep Lloreta, Puay Hoon Tan, Jonathan H Shanks, Rodolfo Montironi, Teresa McHale, David G. Bostwick, Laura Irene Jufe, Mathieu Latour, Helen P. Cathro, Peter Bethwaite, Isabel Trias, Yong Mee Cho, Hemamali Samaratunga, Michelle S. Hirsch, Ai Ying Chuang, Masoud Ganji, Maria Merino, Chin Chen Pan, Giovanna A. Giannico, Ondrej Hes, Ming Zhou, Hiroyuki Takahashi, Theo H. van der Kwast, Glen Kristiansen, Semra Olgac, Hiroshi Miyamoto, Masatoshi Kida, Fiona Maclean, Mathilde Sibony, Fadi Brimo, Peter W. Nichols, Katayoon Rezaei, Ulrika Axcrona, Christina Hulsbergen Van De Kaa, Kiril Trpkov, Holger Moch, Lakshmi P. Kunju, Marina Scarpelli, Jonathan I. Epstein, Ruben Ronchetti, Jorge L. Yao, Nilesh S. Gupta, John C. Cheville, Debra L. Zynger, Khalid Ahmed, Louis R. Bégin, James G. Kench, Satish K. Tickoo, Warick Delprado, Rose Miller, Rajal B. Shah, Adeboye O. Osunkoya, Cheng Wang, Claudio Lewin, Daniel M. Berney, Sueli Suzigan, Maria Shevchuk, Sara M. Falzarano, Liang Cheng, Eddie Fridman, Masoumeh Ghayouri, Neriman Gokden, Maria M. Picken, Laurie Russell, Ferran Algaba, Funda Vakar-Lopez, Adebowale J. Adeniran, George J. Netto, Brett Delahunt, Nathalie Rioux-Leclercq, Larry True, Anil V. Parwani, David J. Griffiths, Mahul B. Amin, Martin Susani, David J. Grignon, Ahmed Shabaik, Kathrine Lie, Cynthia Cohen, Constantina Petraki, Fang Ming Deng, Marc Barry, Hedwig Murphy, Kenneth A. Iczkowski, Peter A. Humphrey, Steven S. Shen, Jiaoti Huang, Brian D. Robinson, Zhaoli Lane, Jin Zhao, Marie O'Donnell, Daniel A. Fajardo, Oluwole Fadare, Samson W. Fine, Bhuvana Srinivasan, Andrew Evans, Rafael E. Jimenez, Hikmat Al Ahmadie, Sundus Hussein, Ying-Bei Chen, Athanase Billis, Bungo Furusato, Guido Martignoni, Maria Pyda-Karwicka, Stephen M. Bonsib, Pheroze Tamboli, Joanna Perry-Keene, Stephen Rohan, Henry Crist, Joan Sweet, Tipu Nazeer, John N. Nacey, and Lars Egevad
- Subjects
medicine.medical_specialty ,Kidney ,Pathology ,business.industry ,medicine.medical_treatment ,medicine.disease ,Kidney Neoplasms ,Nephrectomy ,Specimen Handling ,Pathology and Forensic Medicine ,Surgery ,Adipose capsule of kidney ,Dissection ,medicine.anatomical_structure ,Renal cell carcinoma ,medicine ,Humans ,Anatomy ,Renal vein ,business ,Renal sinus ,Carcinoma, Renal Cell ,Societies, Medical ,Sinus (anatomy) - Abstract
The International Society of Urologic Pathology 2012 Consensus Conference on renal cancer, through working group 3, focused on the issues of staging and specimen handling of renal tumors. The conference was preceded by an online survey of the International Society of Urologic Pathology members, and the results of this were used to inform the focus of conference discussion. On formal voting a Z65% majority was considered a consensus agreement. For specimen handling it was agreed that with radical nephrectomy specimens the initial cut should be made along the long axis and that both radical and partial nephrectomy specimens should be inked. It was recommended that sampling of renal tumors should follow a general guideline of sampling 1 block/cm with a minimum of 3 blocks (subject to modification as needed in individual cases). When measuring a renal tumor, the length of a renal vein/caval thrombus should not be part of the measurement of the main tumor mass. In cases with multiple tumors, sampling should include at a minimum the 5 largest tumors. There was a consensus that perinephric fat invasion should be determined by examining multiple perpendicular sections of the tumor/perinephric fat interface and by sampling areas suspicious for invasion. Perinephric fat invasion was defined as either the tumor touching the fat or extending as irregular tongues into the perinephric tissue, with or without desmoplasia. It was agreed upon that renal sinus invasion is present when the tumor is in direct contact with the sinus fat or the loose connective tissue of the sinus, clearly beyond the renal parenchyma, or if there is involvement of any endothelium-lined spaces within the renal sinus, regardless of the size. When invasion of the renal sinus is uncertain, it was recommended that at least 3 blocks of the tumor-renal sinus interface should be submitted. If invasion is grossly evident, or obviously not present (small peripheral tumor), it was agreed that only 1 block was needed to confirm the gross impression. Other recommendations were that the renal vein margin be considered positive only when there is adherent tumor visible microscopically at the actual margin. When a specimen is submitted separately as "caval thrombus, "the recommended sampling strategy is to take 2 or more sections to look for the adherent caval wall tissue. It was also recommended that uninvolved renal parenchyma be sampled by including normal parenchyma with tumor and normal parenchyma distant from the tumor. There was consensus that radical nephrectomy specimens should be examined for the purpose of identifying lymph nodes by dissection/palpation of the fat in the hilar area only; however, it was acknowledged that lymph nodes are found in
- Published
- 2013
42. The International Society of Urological Pathology (ISUP) Grading System for Renal Cell Carcinoma and Other Prognostic Parameters
- Author
-
Asli Yilmaz, Gabriella Nesi, Josep Lloreta, Oluwole Fadare, Athanase Billis, Jesse K. McKenney, Omar Hameed, Isabel Trias, Neriman Gokden, Bungo Furusato, Ai Ying Chuang, Anne Y. Warren, Peter W. Nichols, Puay Hoon Tan, Mahul B. Amin, Guido Martignoni, Rodolfo Montironi, Holger Moch, Ming Zhou, Daniel A. Fajardo, Cynthia Cohen, David G. Bostwick, Bhuvana Srinivasan, Semra Olgac, Stephen Rohan, Laura Irene Jufe, Katayoon Rezaei, Marina Scarpelli, Khalid Ahmed, Ruben Ronchetti, Nilesh S. Gupta, Barbara Loftus, Martin Susani, Constantina Petraki, Hikmat Al Ahmadie, Michelle S. Hirsch, Sundus Hussein, Joanna Perry-Keene, Pedram Argani, Ondrej Hes, John R. Srigley, Rajal B. Shah, Toyonori Tsuzuki, Rose Miller, Tipu Nazeer, John C. Cheville, Andrew Evans, Ying-Bei Chen, Steven S. Shen, Ferran Algaba, Marie O'Donnell, Cheng Wang, John N. Nacey, Lars Egevad, James G. Kench, Rafael E. Jimenez, Larry True, Brian D. Robinson, Liang Cheng, Maria Shevchuk, Masoumeh Ghayouri, Robert W. Allan, Stewart Fleming, Cristina Magi-Galluzzi, Antonio Lopez-Beltran, Ahmed Shabaik, Anil V. Parwani, Debra L. Zynger, Funda Vakar-Lopez, Dilek Ertoy Baydar, Louis R. Bégin, Mathilde Sibony, Giovanna A. Giannico, Joan Sweet, Wei Huang, Samson W. Fine, Claudio Lewin, Ruth Birbe, Henry Crist, Jonathan H Shanks, Peter A. Humphrey, George J. Netto, Edward C. Jones, Fadi Brimo, Jiaoti Huang, Zhaoli Lane, Peter Bethwaite, Roberto Orozco, Satish K. Tickoo, Eddie Fridman, Maria Merino, Masoud Ganji, Chin Chen Pan, Hiroyuki Takahashi, Aysim Ozagari, Brett Delahunt, Nathalie Rioux-Leclercq, Adebowale J. Adeniran, Teresa McHale, Kathrine Lie, Maria Pyda-Karwicka, David J. Grignon, Warick Delprado, Stephen M. Bonsib, Victor E. Reuter, Yong Mee Cho, Mathieu Latour, Hiroshi Miyamoto, Theo H. van der Kwast, Jin Zhao, Sara M. Falzarano, Masatoshi Kida, Fiona Maclean, Jorge L. Yao, Maria M. Picken, Laurie Russell, Hedwig Murphy, Kenneth A. Iczkowski, Daniel M. Berney, Sueli Suzigan, Helen P. Cathro, Glen Kristiansen, Jonathan I. Epstein, Ulrika Axcrona, Christina Hulsbergen Van De Kaa, Lakshmi P. Kunju, Adeboye O. Osunkoya, David J. Griffiths, Anila Abraham, Fang Ming Deng, and Marc Barry
- Subjects
kidney ,renal cell carcinoma ,Pathology ,medicine.medical_specialty ,microvascular invasion ,Chromophobe cell ,rhabdoid differentiation ,urologic and male genital diseases ,necrosis ,Pathology and Forensic Medicine ,Renal cell carcinoma ,medicine ,Carcinoma ,Humans ,tumor morphotype ,Sarcomatoid carcinoma ,Carcinoma, Renal Cell ,Societies, Medical ,grade ,Neoplasm Grading ,Kidney ,International Society of Urological Pathology ,business.industry ,Prognosis ,medicine.disease ,pathology ,sarcomatoid differentiation ,Kidney Neoplasms ,medicine.anatomical_structure ,Immunohistochemistry ,Surgery ,Anatomy ,business ,Clear cell - Abstract
The International Society of Urological Pathology 2012 Consensus Conference made recommendations regarding classification, prognostic factors, staging, and immunohistochemical and molecular assessment of adult renal tumors. Issues relating to prognostic factors were coordinated by a workgroup who identified tumor morphotype, sarcomatoid/rhabdoid differentiation, tumor necrosis, grading, and microvascular invasion as potential prognostic parameters. There was consensus that the main morphotypes of renal cell carcinoma (RCC) were of prognostic significance, that subtyping of papillary RCC (types 1 and 2) provided additional prognostic information, and that clear cell tubulopapillary RCC was associated with a more favorable outcome. For tumors showing sarcomatoid or rhabdoid differentiation, there was consensus that a minimum proportion of tumor was not required for diagnostic purposes. It was also agreed upon that the underlying subtype of carcinoma should be reported. For sarcomatoid carcinoma, it was further agreed upon that if the underlying carcinoma subtype was absent the tumor should be classified as a grade 4 unclassified carcinoma with a sarcomatoid component. Tumor necrosis was considered to have prognostic significance, with assessment based on macroscopic and microscopic examination of the tumor. It was recommended that for clear cell RCC the amount of necrosis should be quantified. There was consensus that nucleolar prominence defined grades 1 to 3 of clear cell and papillary RCCs, whereas extreme nuclear pleomorphism or sarcomatoid and/or rhabdoid differentiation defined grade 4 tumors. It was agreed upon that chromophobe RCC should not be graded. There was consensus that microvascular invasion should not be included as a staging criterion for RCC.
- Published
- 2013
43. Oncogenes and inflammation rewire host energy metabolism in the tumor microenvironment
- Author
-
Richard G. Pestell, Ruth Birbe, Ubaldo E. Martinez-Outschoorn, Anthony Howell, Alessandro Bombonati, Edmund A. Pribitkin, Madalina Tuluc, Federica Sotgia, Ying-Hui Ko, David Cognetti, Zhao Lin, Michael P. Lisanti, and Joseph Curry
- Subjects
Tumor microenvironment ,Stromal cell ,Oncogene ,Inflammation ,Cell Biology ,Biology ,Cell biology ,Mitochondrial biogenesis ,Downregulation and upregulation ,Cancer cell ,medicine ,Reverse Warburg effect ,medicine.symptom ,Molecular Biology ,Developmental Biology - Abstract
Here, we developed a model system to evaluate the metabolic effects of oncogene(s) on the host microenvironment. A matched set of "normal" and oncogenically transformed epithelial cell lines were co-cultured with human fibroblasts, to determine the "bystander" effects of oncogenes on stromal cells. ROS production and glucose uptake were measured by FACS analysis. In addition, expression of a panel of metabolic protein biomarkers (Caveolin-1, MCT1, and MCT4) was analyzed in parallel. Interestingly, oncogene activation in cancer cells was sufficient to induce the metabolic reprogramming of cancer-associated fibroblasts toward glycolysis, via oxidative stress. Evidence for "metabolic symbiosis" between oxidative cancer cells and glycolytic fibroblasts was provided by MCT1/4 immunostaining. As such, oncogenes drive the establishment of a stromal-epithelial "lactate-shuttle", to fuel the anabolic growth of cancer cells. Similar results were obtained with two divergent oncogenes (RAS and NFκB), indicating that ROS production and inflammation metabolically converge on the tumor stroma, driving glycolysis and upregulation of MCT4. These findings make stromal MCT4 an attractive target for new drug discovery, as MCT4 is a shared endpoint for the metabolic effects of many oncogenic stimuli. Thus, diverse oncogenes stimulate a common metabolic response in the tumor stroma. Conversely, we also show that fibroblasts protect cancer cells against oncogenic stress and senescence by reducing ROS production in tumor cells. Ras-transformed cells were also able to metabolically reprogram normal adjacent epithelia, indicating that cancer cells can use either fibroblasts or epithelial cells as "partners" for metabolic symbiosis. The antioxidant N-acetyl-cysteine (NAC) selectively halted mitochondrial biogenesis in Ras-transformed cells, but not in normal epithelia. NAC also blocked stromal induction of MCT4, indicating that NAC effectively functions as an "MCT4 inhibitor". Taken together, our data provide new strategies for achieving more effective anticancer therapy. We conclude that oncogenes enable cancer cells to behave as selfish "metabolic parasites", like foreign organisms (bacteria, fungi, viruses). Thus, we should consider treating cancer like an infectious disease, with new classes of metabolically targeted "antibiotics" to selectively starve cancer cells. Our results provide new support for the "seed and soil" hypothesis, which was first proposed in 1889 by the English surgeon, Stephen Paget.
- Published
- 2013
44. Molecular Diagnosis of Prostate Cancer: Are We Up to Age?
- Author
-
Peter McCue, Ruth Birbe, and Tapan Bhavsar
- Subjects
Male ,Proteomics ,PCA3 ,business.industry ,Prostatic Neoplasms ,Hematology ,Disease ,Bioinformatics ,medicine.disease ,Epigenesis, Genetic ,Prostate cancer ,Circulating tumor cell ,Oncology ,Prostatic acid phosphatase ,Mutation ,Biomarkers, Tumor ,Humans ,Medicine ,Biomarker discovery ,Stage (cooking) ,business ,Early Detection of Cancer - Abstract
Prostate cancer (PCa), a highly heterogeneous disease, is the one of the leading cause of morbidity and mortality in the developed countries. Historically used biomarkers such as prostatic acid phosphatase (PAP), serum prostate-specific antigen (PSA), and its precursor have not stood the challenge of sensitivity and specificity. At present, there is need to re-evaluate the approach to diagnose and monitor PCa. To this end, molecular markers that can accurately identify men with PCa at an early stage, and those who would benefit from early therapeutic intervention, are the need of the hour. There has been unprecedented progress in the development of new PCa biomarkers through advancements in proteomics, tissue DNA and protein/RNA microarray, identification of microRNA, isolation of circulating tumor cells, and tumor immunohistochemistry. This review will examine the current status of prostate cancer biomarkers with emphasis on emerging biomarkers by evaluating their diagnostic and prognostic potentials.
- Published
- 2013
45. Convergence of oncogenic and hormone receptor pathways promotes metastatic phenotypes
- Author
-
Alessandro Fatatis, Kari Wilder-Romans, Jeffry L. Dean, Karen E. Knudsen, Christopher McNair, Danielle Jernigan, Tapio Visakorpi, Donald P. McDonnell, Mark Shilkrut, Daniel E. Frigo, Michael A. Augello, Craig J. Burd, Ruth Birbe, Felix Y. Feng, Adam Ertel, Michael S. Magee, and Sumin Han
- Subjects
Male ,Transcriptional Activation ,Slug ,Cyclin D ,Transplantation, Heterologous ,Cyclin A ,Mice ,Cyclin D1 ,Animals ,Humans ,Neoplasm Invasiveness ,Neoplasm Metastasis ,Cyclin ,biology ,Prostatic Neoplasms ,General Medicine ,Cell cycle ,biology.organism_classification ,Gene Expression Regulation, Neoplastic ,Androgen receptor ,Alternative Splicing ,Receptors, Androgen ,biology.protein ,Cancer research ,Snail Family Transcription Factors ,Neoplasm Transplantation ,Cyclin A2 ,Research Article ,Signal Transduction ,Transcription Factors - Abstract
Cyclin D1b is a splice variant of the cell cycle regulator cyclin D1 and is known to harbor divergent and highly oncogenic functions in human cancer. While cyclin D1b is induced during disease progression in many cancer types, the mechanisms underlying cyclin D1b function remain poorly understood. Herein, cell and human tumor xenograft models of prostate cancer were utilized to resolve the downstream pathways that are required for the protumorigenic functions of cyclin D1b. Specifically, cyclin D1b was found to modulate the expression of a large transcriptional network that cooperates with androgen receptor (AR) signaling to enhance tumor cell growth and invasive potential. Notably, cyclin D1b promoted AR-dependent activation of genes associated with metastatic phenotypes. Further exploration determined that transcriptional induction of SNAI2 (Slug) was essential for cyclin D1b-mediated proliferative and invasive properties, implicating Slug as a critical driver of disease progression. Importantly, cyclin D1b expression highly correlated with that of Slug in clinical samples of advanced disease. In vivo analyses provided strong evidence that Slug enhances both tumor growth and metastatic phenotypes. Collectively, these findings reveal the underpinning mechanisms behind the protumorigenic functions of cyclin D1b and demonstrate that the convergence of the cyclin D1b/AR and Slug pathways results in the activation of processes critical for the promotion of lethal tumor phenotypes.
- Published
- 2012
46. BRCA1 mutations drive oxidative stress and glycolysis in the tumor microenvironment
- Author
-
Michael P. Lisanti, Federica Sotgia, Sharon Sneddon, Alessandro Bombonati, Ruth Birbe, Diana Whitaker-Menezes, Anthony Howell, Stephanos Pavlides, Ubaldo E. Martinez-Outschoorn, Zhao Lin, Rebecca Lamb, and Renee M. Balliet
- Subjects
Monocarboxylic Acid Transporters ,Stromal cell ,Tumor suppressor gene ,Caveolin 1 ,Muscle Proteins ,Apoptosis ,Breast Neoplasms ,Biology ,Antioxidants ,Cell Line ,Cyclic N-Oxides ,Breast cancer ,Report ,Tumor Microenvironment ,medicine ,Humans ,skin and connective tissue diseases ,Molecular Biology ,Triple-negative breast cancer ,Tumor microenvironment ,BRCA1 Protein ,Cancer ,Hydrogen Peroxide ,Cell Biology ,medicine.disease ,Coculture Techniques ,Acetylcysteine ,Up-Regulation ,Oxidative Stress ,Cell culture ,Mutation ,Cancer cell ,Cancer research ,Female ,Spin Labels ,Glycolysis ,Developmental Biology - Abstract
Mutations in the BRCA1 tumor suppressor gene are commonly found in hereditary breast cancer. Similarly, downregulation of BRCA1 protein expression is observed in the majority of basal-like breast cancers. Here, we set out to study the effects of BRCA1 mutations on oxidative stress in the tumor microenvironment. To mimic the breast tumor microenvironment, we utilized an in vitro co-culture model of human BRCA1-mutated HCC1937 breast cancer cells and hTERT-immortalized human fibroblasts. Notably, HCC1937 cells induce the generation of hydrogen peroxide in the fibroblast compartment during co-culture, which can be inhibited by genetic complementation with the wild-type BRCA1 gene. Importantly, treatment with powerful antioxidants, such as NAC and Tempol, induces apoptosis in HCC1937 cells, suggesting that microenvironmental oxidative stress supports cancer cell survival. In addition, Tempol treatment increases the apoptotic rates of MDA-MB-231 cells, which have wild-type BRCA1, but share a basal-like breast cancer phenotype with HCC1937 cells. MCT4 is the main exporter of L-lactate out of cells and is a marker for oxidative stress and glycolytic metabolism. Co-culture with HCC1937 cells dramatically induces MCT4 protein expression in fibroblasts, and this can be prevented by either BRCA1 overexpression or by pharmacological treatment with NAC. We next evaluated caveolin-1 (Cav-1) expression in stromal fibroblasts. Loss of Cav-1 is a marker of the cancer-associated fibroblast (CAF) phenotype, which is linked to high stromal glycolysis, and is associated with a poor prognosis in numerous types of human cancers, including breast cancers. Remarkably, HCC1937 cells induce a loss of Cav-1 in adjacent stromal cells during co-culture. Conversely, Cav-1 expression in fibroblasts can be rescued by administration of NAC or by overexpression of BRCA1 in HCC1937 cells. Notably, BRCA1-deficient human breast cancer samples (9 out of 10) also showed a glycolytic stromal phenotype, with intense mitochondrial staining specifically in BRCA1-deficient breast cancer cells. In summary, loss of BRCA1 function leads to hydrogen peroxide generation in both epithelial breast cancer cells and neighboring stromal fibroblasts, and promotes the onset of a reactive glycolytic stroma, with increased MCT4 and decreased Cav-1 expression. Importantly, these metabolic changes can be reversed by antioxidants, which potently induce cancer cell death. Thus, antioxidant therapy appears to be synthetically lethal with a BRCA1-deficiency in breast cancer cells and should be considered for future cancer prevention trials. In this regard, immunostaining with Cav-1 and MCT4 could be used as cost-effective biomarkers to monitor the response to antioxidant therapy.
- Published
- 2012
47. Metformin effects on head and neck squamous carcinoma microenvironment: Window of opportunity trial
- Author
-
David Cognetti, Tingting Zhan, Diana Whitaker Menezes, Michelle L. Reyzer, Patrick Tassone, Marina Domingo Vidal, Madalina Tuluc, Kurren S. Gill, Zhao Lin, Paolo Cotzia, Benjamin E. Leiby, Elizabeth Duddy, Adam Luginbuhl, Ubaldo E. Martinez-Outschoorn, Joseph Curry, John Sprandio, Jennifer Johnson, Ruth Birbe, and Mehri Mollaee
- Subjects
0301 basic medicine ,Oncology ,Adult ,Male ,medicine.medical_specialty ,Article ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Biopsy ,medicine ,Carcinoma ,Tumor Microenvironment ,Humans ,Aged ,Aged, 80 and over ,Tumor microenvironment ,TUNEL assay ,medicine.diagnostic_test ,business.industry ,Squamous Cell Carcinoma of Head and Neck ,Middle Aged ,medicine.disease ,Head and neck squamous-cell carcinoma ,Metformin ,3. Good health ,Squamous carcinoma ,030104 developmental biology ,Otorhinolaryngology ,Terminal deoxynucleotidyl transferase ,Head and Neck Neoplasms ,030220 oncology & carcinogenesis ,Carcinoma, Squamous Cell ,Female ,business ,medicine.drug - Abstract
Objective The tumor microenvironment frequently displays abnormal cellular metabolism, which contributes to aggressive behavior. Metformin inhibits mitochondrial oxidative phosphorylation, altering metabolism. Though the mechanism is unclear, epidemiologic studies show an association between metformin use and improved outcomes in head and neck squamous cell carcinoma (HNSCC). We sought to determine if metformin alters metabolism and apoptosis in HNSCC tumors. Study Design Window of opportunity trial of metformin between diagnostic biopsy and resection. Participants were patients with newly diagnosed HNSCC. Fifty patients were enrolled, and 39 completed a full-treatment course. Metformin was titrated to standard diabetic dose (2,000 mg/day) for a course of 9 or more days prior to surgery. Methods Immunohistochemistry (IHC) for the metabolic markers caveolin-1 (CAV1), B-galactosidase (GALB), and monocarboxylate transporter 4 (MCT4), as well as the Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) apoptosis assay and Ki-67 IHC, were performed in pre- and postmetformin specimens. Exploratory mass spectroscopy imaging (MSI) to assess lactate levels also was performed in three subjects. Results Metformin was well tolerated. The average treatment course was 13.6 days. Posttreatment specimens showed a significant increase in stromal CAV1 (P < 0.001) and GALB (P < 0.005), as well as tumor cell apoptosis by TUNEL assay (P < 0.001). There was no significant change in stromal MCT4 expression or proliferation measured by Ki67. Lactate levels in carcinoma cells were increased 2.4-fold postmetformin (P < 0.05), as measured by MSI. Conclusion Metformin increases markers of reduced catabolism and increases senescence in stromal cells as well as carcinoma cell apoptosis. This study demonstrates that metformin modulates metabolism in the HNSCC microenvironment. Level of Evidence 4. Laryngoscope, 127:1808–1815, 2017
- Published
- 2016
48. siRNA-Encapsulated Hybrid Nanoparticles Target Mutant K-ras and Inhibit Metastatic Tumor Burden in a Mouse Model of Lung Cancer
- Author
-
Yi Liu, Bo Lu, Sunday A. Shoyele, Adam E. Snook, Ruth Birbe, Robert B. Den, Chellappagounder Thangavel, Maryna Perepelyuk, and Olubunmi Shoyele
- Subjects
0301 basic medicine ,Small interfering RNA ,Mutant ,hybrid nanoparticles ,03 medical and health sciences ,gene silencing ,Immune system ,Downregulation and upregulation ,Drug Discovery ,medicine ,Gene silencing ,Lung cancer ,orthotopic model ,business.industry ,lcsh:RM1-950 ,medicine.disease ,respiratory tract diseases ,mutant K-ras ,lung cancer ,lcsh:Therapeutics. Pharmacology ,030104 developmental biology ,siRNA ,Toxicity ,Immunology ,Cancer research ,Molecular Medicine ,Original Article ,Erlotinib ,human immunoglobulin G ,business ,medicine.drug - Abstract
There is an unmet need in the development of an effective therapy for mutant K-ras-expressing non-small-cell lung cancer (NSCLC). Although various small molecules have been evaluated, an effective therapy remains a dream. siRNAs have the potential to downregulate mutant K-ras both at the protein and mRNA levels. However, a safe and effective delivery of siRNAs to tumors remains a limitation to their translational application in the treatment of this highly debilitating disease. Here we developed a novel hybrid nanoparticle carrier for effective delivery of anti-mutant K-ras to NSCLC (AKSLHN). The ability of this treatment modality to regress lung tumors in mouse models was evaluated as a monotherapy or as a combination treatment with erlotinib. Further, the toxicity of this treatment modality to healthy tissues was evaluated, along with its ability to elicit immune/inflammatory reactions. The results suggest that this treatment modality is a promising prospect for the treatment of mutant K-ras-expressing NSCLC without any accompanying toxicity. However, further understanding of the cellular-level interaction between AHSLHN and erlotinib needs to be attained before this promising treatment modality can be brought to the bedside. Keywords: hybrid nanoparticles, siRNA, mutant K-ras, lung cancer, gene silencing, orthotopic model, human immunoglobulin G
- Published
- 2016
49. TP53-inducible Glycolysis and Apoptosis Regulator (TIGAR) Metabolically Reprograms Carcinoma and Stromal Cells in Breast Cancer
- Author
-
Àurea Navarro-Sabaté, Erin L. Seifert, Marina Domingo-Vidal, Megan Roche, Patrick Tassone, Diana Whitaker-Menezes, Jaime Caro, Ubaldo E. Martinez-Outschoorn, Claudia Capparelli, Ruth Birbe, Zhao Lin, Ramon Bartrons, Joseph Curry, Madalina Tuluc, Anna Manzano, and Ying-Hui Ko
- Subjects
0301 basic medicine ,Stromal cell ,Phosphofructokinase-2 ,Àcid glutàmic ,Glutamic Acid ,Apoptosis ,Breast Neoplasms ,Biology ,Bioenergetics ,Biochemistry ,Càncer de mama ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Carcinoma ,medicine ,Humans ,Glycolysis ,Lactic Acid ,Molecular Biology ,Tumor microenvironment ,L-Lactate Dehydrogenase ,Catabolism ,Intracellular Signaling Peptides and Proteins ,Cell Biology ,Fibroblasts ,medicine.disease ,TP53-inducible glycolysis and apoptosis regulator ,Molecular biology ,Metabolisme ,Coculture Techniques ,Phosphoric Monoester Hydrolases ,Glutamine ,Isoenzymes ,030104 developmental biology ,Metabolism ,030220 oncology & carcinogenesis ,Cancer research ,MCF-7 Cells ,Female ,Glutamic acid ,Lactate Dehydrogenase 5 ,Tumor Suppressor Protein p53 ,Breast carcinoma ,Apoptosis Regulatory Proteins - Abstract
A subgroup of breast cancers has several metabolic compartments. The mechanisms by which metabolic compartmentalization develop in tumors are poorly characterized. TP53 inducible glycolysis and apoptosis regulator (TIGAR) is a bisphosphatase that reduces glycolysis and is highly expressed in carcinoma cells in the majority of human breast cancers. Hence we set out to determine the effects of TIGAR expression on breast carcinoma and fibroblast glycolytic phenotype and tumor growth. The overexpression of this bisphosphatase in carcinoma cells induces expression of enzymes and transporters involved in the catabolism of lactate and glutamine. Carcinoma cells overexpressing TIGAR have higher oxygen consumption rates and ATP levels when exposed to glutamine, lactate, or the combination of glutamine and lactate. Coculture of TIGAR overexpressing carcinoma cells and fibroblasts compared with control cocultures induce more pronounced glycolytic differences between carcinoma and fibroblast cells. Carcinoma cells overexpressing TIGAR have reduced glucose uptake and lactate production. Conversely, fibroblasts in coculture with TIGAR overexpressing carcinoma cells induce HIF (hypoxia-inducible factor) activation with increased glucose uptake, increased 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3 (PFKFB3), and lactate dehydrogenase-A expression. We also studied the effect of this enzyme on tumor growth. TIGAR overexpression in carcinoma cells increases tumor growth in vivo with increased proliferation rates. However, a catalytically inactive variant of TIGAR did not induce tumor growth. Therefore, TIGAR expression in breast carcinoma cells promotes metabolic compartmentalization and tumor growth with a mitochondrial metabolic phenotype with lactate and glutamine catabolism. Targeting TIGAR warrants consideration as a potential therapy for breast cancer.
- Published
- 2016
50. S&T-39 CONCORDANCE OF URETEROSCOPIC BIOPSY GRADE TO RADICAL NEPHROURETERECTOMY GRADE AND STAGE BASED ON THE 1973 AND 2004 WHO CLASSIFICATION SYSTEMS FOR UPPER TRACT UROTHELIAL CARCINOMA
- Author
-
Ryuta Tanimoto, Demetrius H. Bagley, Kelly A. Healy, Ruth Birbe, Marluce Bibbo, and Scott G. Hubosky
- Subjects
medicine.medical_specialty ,Upper tract ,medicine.diagnostic_test ,business.industry ,Urology ,Concordance ,Biopsy ,medicine ,Stage (cooking) ,Who classification ,business ,Urothelial carcinoma - Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.