39 results on '"Ian Pass"'
Search Results
2. Improving drug discovery using image-based multiparametric analysis of the epigenetic landscape
- Author
-
Chen Farhy, Santosh Hariharan, Jarkko Ylanko, Luis Orozco, Fu-Yue Zeng, Ian Pass, Fernando Ugarte, E Camilla Forsberg, Chun-Teng Huang, David W Andrews, and Alexey V Terskikh
- Subjects
high content screening ,glioblastoma ,epigenetics ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
High-content phenotypic screening has become the approach of choice for drug discovery due to its ability to extract drug-specific multi-layered data. In the field of epigenetics, such screening methods have suffered from a lack of tools sensitive to selective epigenetic perturbations. Here we describe a novel approach, Microscopic Imaging of Epigenetic Landscapes (MIEL), which captures the nuclear staining patterns of epigenetic marks and employs machine learning to accurately distinguish between such patterns. We validated the MIEL platform across multiple cells lines and using dose-response curves, to insure the fidelity and robustness of this approach for high content high throughput drug discovery. Focusing on noncytotoxic glioblastoma treatments, we demonstrated that MIEL can identify and classify epigenetically active drugs. Furthermore, we show MIEL was able to accurately rank candidate drugs by their ability to produce desired epigenetic alterations consistent with increased sensitivity to chemotherapeutic agents or with induction of glioblastoma differentiation.
- Published
- 2019
- Full Text
- View/download PDF
3. Data from Serine-Threonine Kinase TAO3-Mediated Trafficking of Endosomes Containing the Invadopodia Scaffold TKS5α Promotes Cancer Invasion and Tumor Growth
- Author
-
Sara A. Courtneidge, Anthony B. Pinkerton, Christian A. Hassig, Shannon K. McWeeney, George V. Thomas, Ian Pass, Fu-Yue Zeng, Eduard Sergienko, Chen-Ting Ma, Matthew M. Abelman, Robert J. Ardecky, Kyle P. Gribbin, Jose C. Navarro, Manuela Quintavalle, and Shinji Iizuka
- Abstract
Invadopodia are actin-based proteolytic membrane protrusions required for invasive behavior and tumor growth. In this study, we used our high-content screening assay to identify kinases whose activity affects invadopodia formation. Among the top hits selected for further analysis was TAO3, an STE20-like kinase of the GCK subfamily. TAO3 was overexpressed in many human cancers and regulated invadopodia formation in melanoma, breast, and bladder cancers. Furthermore, TAO3 catalytic activity facilitated melanoma growth in three-dimensional matrices and in vivo. A novel, potent catalytic inhibitor of TAO3 was developed that inhibited invadopodia formation and function as well as tumor cell extravasation and growth. Treatment with this inhibitor demonstrated that TAO3 activity is required for endosomal trafficking of TKS5α, an obligate invadopodia scaffold protein. A phosphoproteomics screen for TAO3 substrates revealed the dynein subunit protein LIC2 as a relevant substrate. Knockdown of LIC2 or expression of a phosphomimetic form promoted invadopodia formation. Thus, TAO3 is a new therapeutic target with a distinct mechanism of action.Significance:An unbiased screening approach identifies TAO3 as a regulator of invadopodia formation and function, supporting clinical development of this class of target.
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary movie 2 from Serine-Threonine Kinase TAO3-Mediated Trafficking of Endosomes Containing the Invadopodia Scaffold TKS5α Promotes Cancer Invasion and Tumor Growth
- Author
-
Sara A. Courtneidge, Anthony B. Pinkerton, Christian A. Hassig, Shannon K. McWeeney, George V. Thomas, Ian Pass, Fu-Yue Zeng, Eduard Sergienko, Chen-Ting Ma, Matthew M. Abelman, Robert J. Ardecky, Kyle P. Gribbin, Jose C. Navarro, Manuela Quintavalle, and Shinji Iizuka
- Abstract
Effect of SBI-581 on TKS5alpha vesicle trafficking
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary movie 3 from Serine-Threonine Kinase TAO3-Mediated Trafficking of Endosomes Containing the Invadopodia Scaffold TKS5α Promotes Cancer Invasion and Tumor Growth
- Author
-
Sara A. Courtneidge, Anthony B. Pinkerton, Christian A. Hassig, Shannon K. McWeeney, George V. Thomas, Ian Pass, Fu-Yue Zeng, Eduard Sergienko, Chen-Ting Ma, Matthew M. Abelman, Robert J. Ardecky, Kyle P. Gribbin, Jose C. Navarro, Manuela Quintavalle, and Shinji Iizuka
- Abstract
Effect of SBI-581 on Rab11 vesicle trafficking
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Data from Serine-Threonine Kinase TAO3-Mediated Trafficking of Endosomes Containing the Invadopodia Scaffold TKS5α Promotes Cancer Invasion and Tumor Growth
- Author
-
Sara A. Courtneidge, Anthony B. Pinkerton, Christian A. Hassig, Shannon K. McWeeney, George V. Thomas, Ian Pass, Fu-Yue Zeng, Eduard Sergienko, Chen-Ting Ma, Matthew M. Abelman, Robert J. Ardecky, Kyle P. Gribbin, Jose C. Navarro, Manuela Quintavalle, and Shinji Iizuka
- Abstract
Includes Sup. Figures 1-7, Tables 1-6 and Methods.
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary movie 4 from Serine-Threonine Kinase TAO3-Mediated Trafficking of Endosomes Containing the Invadopodia Scaffold TKS5α Promotes Cancer Invasion and Tumor Growth
- Author
-
Sara A. Courtneidge, Anthony B. Pinkerton, Christian A. Hassig, Shannon K. McWeeney, George V. Thomas, Ian Pass, Fu-Yue Zeng, Eduard Sergienko, Chen-Ting Ma, Matthew M. Abelman, Robert J. Ardecky, Kyle P. Gribbin, Jose C. Navarro, Manuela Quintavalle, and Shinji Iizuka
- Abstract
Effect of SU11333 on TKS5alpha vesicle trafficking
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary movie 1 from Serine-Threonine Kinase TAO3-Mediated Trafficking of Endosomes Containing the Invadopodia Scaffold TKS5α Promotes Cancer Invasion and Tumor Growth
- Author
-
Sara A. Courtneidge, Anthony B. Pinkerton, Christian A. Hassig, Shannon K. McWeeney, George V. Thomas, Ian Pass, Fu-Yue Zeng, Eduard Sergienko, Chen-Ting Ma, Matthew M. Abelman, Robert J. Ardecky, Kyle P. Gribbin, Jose C. Navarro, Manuela Quintavalle, and Shinji Iizuka
- Abstract
Trafficking of TKS5alpha vesicles
- Published
- 2023
- Full Text
- View/download PDF
9. Identification and characterization of small molecule inhibitors of the ubiquitin ligases Siah1/2 in melanoma and prostate cancer cells
- Author
-
Gregory W. Cadwell, Martin E. Gleave, Robert C. Liddington, Fan Zhang, David B. Terry, Fuqiang Ban, Chen-Ting Ma, Anthony B. Pinkerton, Ian Pass, Fu-Yue Zeng, E. Hampton Sessions, Eduard Sergienko, Artem Cherkasov, Michael R. Jackson, Thomas D.Y. Chung, Laurie A. Bankston, Veronica Placencio-Hickok, Ze'ev Ronai, Yongmei Feng, and Neil A. Bhowmick
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Cell Survival ,Ubiquitin-Protein Ligases ,In silico ,Down-Regulation ,SIAH2 ,SIAH1 ,Article ,Mass Spectrometry ,Small Molecule Libraries ,Mice ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Ubiquitin ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Computer Simulation ,Melanoma ,Cell Proliferation ,biology ,Chemistry ,Nuclear Proteins ,Prostatic Neoplasms ,medicine.disease ,Xenograft Model Antitumor Assays ,Small molecule ,Ubiquitin ligase ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Drug Screening Assays, Antitumor - Abstract
Inhibition of ubiquitin ligases with small molecule remains a very challenging task, given the lack of catalytic activity of the target and the requirement of disruption of its interactions with other proteins. Siah1/2, which are E3 ubiquitin ligases, are implicated in melanoma and prostate cancer and represent high-value drug targets. We utilized three independent screening approaches in our efforts to identify small-molecule Siah1/2 inhibitors: Affinity Selection-Mass Spectrometry, a protein thermal shift-based assay and an in silico based screen. Inhibitors were assessed for their effect on viability of melanoma and prostate cancer cultures, colony formation, prolyl-hydroxylase-HIF1α signaling, expression of selected Siah2-related transcripts, and Siah2 ubiquitin ligase activity. Several analogs were further characterized, demonstrating improved efficacy. Combination of the top hits identified in the different assays demonstrated an additive effect, pointing to complementing mechanisms that underlie each of these Siah1/2 inhibitors.
- Published
- 2019
- Full Text
- View/download PDF
10. Development of a Robust High-Throughput Screening Platform for Inhibitors of the Striatal-Enriched Tyrosine Phosphatase (STEP)
- Author
-
Sumeet Salaniwal, Nicholas D. P. Cosford, Maria Celeridad, Andrey A. Bobkov, Paul J. Lombroso, Palak Gosalia, Fu Yue Zeng, Stefan Grotegut, Ian Pass, Laurent J.S. De Backer, Lutz Tautz, Thomas D.Y. Chung, and Lester J. Lambert
- Subjects
QH301-705.5 ,High-throughput screening ,PTPN5 ,Computational biology ,Disease ,Protein tyrosine phosphatase ,protein tyrosine phosphatase ,Article ,Catalysis ,Inorganic Chemistry ,Drug Discovery ,Protein-Tyrosine Kinases ,Humans ,Biology (General) ,Enzyme Inhibitors ,Physical and Theoretical Chemistry ,QD1-999 ,Molecular Biology ,Spectroscopy ,Molecular Structure ,Chemistry ,Drug discovery ,Organic Chemistry ,General Medicine ,Protein Tyrosine Phosphatases, Non-Receptor ,High-Throughput Screening Assays ,Computer Science Applications ,Synaptic function ,neurodegenerative disorders ,small-molecule screening ,Tyrosine kinase ,Alzheimer’s disease - Abstract
Many human diseases are the result of abnormal expression or activation of protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPs). Not surprisingly, more than 30 tyrosine kinase inhibitors (TKIs) are currently in clinical use and provide unique treatment options for many patients. PTPs on the other hand have long been regarded as “undruggable” and only recently have gained increased attention in drug discovery. Striatal-enriched tyrosine phosphatase (STEP) is a neuron-specific PTP that is overactive in Alzheimer’s disease (AD) and other neurodegenerative and neuropsychiatric disorders, including Parkinson’s disease, schizophrenia, and fragile X syndrome. An emergent model suggests that the increase in STEP activity interferes with synaptic function and contributes to the characteristic cognitive and behavioral deficits present in these diseases. Prior efforts to generate STEP inhibitors with properties that warrant clinical development have largely failed. To identify novel STEP inhibitor scaffolds, we developed a biophysical, label-free high-throughput screening (HTS) platform based on the protein thermal shift (PTS) technology. In contrast to conventional HTS using STEP enzymatic assays, we found the PTS platform highly robust and capable of identifying true hits with confirmed STEP inhibitory activity and selectivity. This new platform promises to greatly advance STEP drug discovery and should be applicable to other PTP targets.
- Published
- 2021
- Full Text
- View/download PDF
11. Serine-threonine kinase TAO3-mediated trafficking of endosomes containing the invadopodia scaffold TKS5α promotes cancer invasion and tumor growth
- Author
-
Manuela Quintavalle, Fu Yue Zeng, Christian A. Hassig, Shinji Iizuka, Shannon K. McWeeney, Robert Ardecky, Sara A. Courtneidge, Ian Pass, Matthew Mark Abelman, Anthony B. Pinkerton, Jose Navarro, Eduard Sergienko, Chen Ting Ma, George Thomas, and Kyle P. Gribbin
- Subjects
0301 basic medicine ,Scaffold protein ,Cytoplasmic Dyneins ,Male ,Cancer Research ,Skin Neoplasms ,Endosome ,Protein subunit ,Datasets as Topic ,Antineoplastic Agents ,Endosomes ,Protein Serine-Threonine Kinases ,Time-Lapse Imaging ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cell Line, Tumor ,Animals ,Humans ,Neoplasm Invasiveness ,Melanoma ,Serine/threonine-specific protein kinase ,Gene knockdown ,Chemistry ,Kinase ,Gene Expression Profiling ,Phosphoproteomics ,Xenograft Model Antitumor Assays ,Cell biology ,Extracellular Matrix ,High-Throughput Screening Assays ,Adaptor Proteins, Vesicular Transport ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Gene Knockdown Techniques ,Invadopodia ,Podosomes ,Female - Abstract
Invadopodia are actin-based proteolytic membrane protrusions required for invasive behavior and tumor growth. In this study, we used our high-content screening assay to identify kinases whose activity affects invadopodia formation. Among the top hits selected for further analysis was TAO3, an STE20-like kinase of the GCK subfamily. TAO3 was overexpressed in many human cancers and regulated invadopodia formation in melanoma, breast, and bladder cancers. Furthermore, TAO3 catalytic activity facilitated melanoma growth in three-dimensional matrices and in vivo. A novel, potent catalytic inhibitor of TAO3 was developed that inhibited invadopodia formation and function as well as tumor cell extravasation and growth. Treatment with this inhibitor demonstrated that TAO3 activity is required for endosomal trafficking of TKS5α, an obligate invadopodia scaffold protein. A phosphoproteomics screen for TAO3 substrates revealed the dynein subunit protein LIC2 as a relevant substrate. Knockdown of LIC2 or expression of a phosphomimetic form promoted invadopodia formation. Thus, TAO3 is a new therapeutic target with a distinct mechanism of action. Significance: An unbiased screening approach identifies TAO3 as a regulator of invadopodia formation and function, supporting clinical development of this class of target.
- Published
- 2021
12. The ubiquitin ligase RNF5 determines acute myeloid leukemia growth and susceptibility to histone deacetylase inhibitors
- Author
-
Ronit Almog, Eytan Ruppin, Yishai Ofran, Kevin K. Brown, Joo Sang Lee, Ali Khateb, Darren Finlay, Yongmei Feng, Anagha Deshpande, Bertrand Fabre, Carol Burian, Ikrame Lazar, Ido Livneh, Ian Pass, Nurit Horesh, Michael R. Jackson, Irmela Jeremias, Yu Fujita, Yan Li, Jun Yin, Ze'ev Ronai, Kristiina Vuori, Aniruddha J. Deshpande, Tongwu Zhang, and James R. Mason
- Subjects
Science ,Ubiquitin-Protein Ligases ,General Physics and Astronomy ,HL-60 Cells ,Mice, SCID ,Biology ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Ubiquitin ,Mice, Inbred NOD ,Cell Line, Tumor ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Genetic Predisposition to Disease ,Epigenetics ,neoplasms ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,Gene knockdown ,Multidisciplinary ,Cell growth ,Membrane Proteins ,Myeloid leukemia ,U937 Cells ,General Chemistry ,Xenograft Model Antitumor Assays ,3. Good health ,Ubiquitin ligase ,DNA-Binding Proteins ,Histone Deacetylase Inhibitors ,Mice, Inbred C57BL ,HEK293 Cells ,Leukemia, Myeloid ,030220 oncology & carcinogenesis ,Acute Disease ,biology.protein ,Cancer research ,Female ,Histone deacetylase ,K562 Cells ,Carcinogenesis - Abstract
Acute myeloid leukemia (AML) remains incurable, largely due to its resistance to conventional treatments. Here, we find that increased abundance of the ubiquitin ligase RNF5 contributes to AML development and survival. High RNF5 expression in AML patient specimens correlates with poor prognosis. RNF5 inhibition decreases AML cell growth in culture, in patient-derived xenograft (PDX) samples and in vivo, and delays development of MLL-AF9–driven leukemogenesis in mice, prolonging their survival. RNF5 inhibition causes transcriptional changes that overlap with those seen upon histone deacetylase (HDAC)1 inhibition. RNF5 induces the formation of K29 ubiquitin chains on the histone-binding protein RBBP4, promoting its recruitment to and subsequent epigenetic regulation of genes involved in AML maintenance. Correspondingly, RNF5 or RBBP4 knockdown enhances AML cell sensitivity to HDAC inhibitors. Notably, low expression of both RNF5 and HDAC coincides with a favorable prognosis. Our studies identify an ERAD-independent role for RNF5, demonstrating that its control of RBBP4 constitutes an epigenetic pathway that drives AML, and highlight RNF5/RBBP4 as markers useful to stratify patients for treatment with HDAC inhibitors. Epigenetic changes are implicated in Acute myeloid leukemia (AML) tumorigenesis. Here, the authors show that the ubiquitin ligase RNF5 and its substrate RBBP4 contribute to AML development by regulating epigenetic-controlled transcription which determines AML sensitivity to HDAC inhibitors.
- Published
- 2021
- Full Text
- View/download PDF
13. RNF5 Regulation of RBBP4 Defines Acute Myeloid Leukemia Growth and Susceptibility to Histone Deacetylase Inhibitors
- Author
-
Tongwu Zhang, Anagha Deshpande, Ikrame Lazar, Yan Li, Kristiina Vuori, Ali Khateb, Ze'ev Ronai, Aniruddha J. Deshpande, Yu Fujita, Michael R. Jackson, Joo Sang Lee, Ronit Almog, Eytan Ruppin, Kevin K. Brown, James R. Mason, Ian Pass, Nurit Horesh, Yongmei Feng, Yishai Ofran, Carol Burian, Darren Finlay, Ido Livneh, Jun Yin, and Bertrand Fabre
- Subjects
Gene knockdown ,Ubiquitin ,biology ,Cell growth ,hemic and lymphatic diseases ,biology.protein ,Cancer research ,Myeloid leukemia ,Epigenetics ,Histone deacetylase ,HDAC1 ,Ubiquitin ligase - Abstract
Acute myeloid leukemia (AML) remains incurable, largely due to its resistance to conventional treatments. Here, we found that increased expression and abundance of the ubiquitin ligase RNF5 contributes to AML development and survival. High RNF5 expression in AML patients correlated with poor prognosis. RNF5 inhibition decreased AML cell growth in culture and in vivo, and blocked development of MLL-AF9–driven leukemogenesis in mice, prolonging their survival. RNF5 inhibition led to transcriptional changes that overlapped with those seen upon HDAC1 inhibition. RNF5 induced the formation of K29 ubiquitin chains on the histone-binding protein RBBP4, promoting its recruitment and subsequent epigenetic regulation of genes involved in AML development and maintenance. Correspondingly, RNF5 or RBBP4 knockdown enhanced the sensitivity of AML cells to histone deacetylase (HDAC) inhibitors. Notably, low expression of RNF5 and HDAC coincided with a favorable prognosis. Our studies identified ERAD-independent role for RNF5, demonstrating that its control of RBBP4 constitutes an epigenetic pathway that drives AML while highlighting RNF5/RBBP4 as markers to stratify patients for treatment with HDAC inhibitors.
- Published
- 2020
- Full Text
- View/download PDF
14. The serine-threonine kinase TAO3 promotes cancer invasion and tumor growth by facilitating trafficking of endosomes containing the invadopodia scaffold TKS5α
- Author
-
Fu-Yue Zeng, Kyle P. Gribbin, J. Ceja Navarro, M. Quintavalle, Shinji Iizuka, Chen-Ting Ma, Christian A. Hassig, George Thomas, Anthony B. Pinkerton, Shannon K. McWeeney, Eduard Sergienko, Matthew Mark Abelman, Ian Pass, Sara A. Courtneidge, and Robert Ardecky
- Subjects
Scaffold protein ,Serine/threonine-specific protein kinase ,0303 health sciences ,Gene knockdown ,Chemistry ,Endosome ,Kinase ,Protein subunit ,Phosphoproteomics ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Invadopodia ,030304 developmental biology - Abstract
Invadopodia are actin-based proteolytic membrane protrusions required for invasive behavior and tumor growth. We used our high-content screening assay to identify kinases impacting invadopodia formation. Among the top hits we selected TAO3, a STE20-like kinase of the GCK subfamily, for further analysis. TAO3 was over-expressed in many human cancers, and regulated invadopodia formation in melanoma, breast and bladder cancers. Furthermore, TAO3 catalytic activity facilitated melanoma growth in 3-dimensional matrices andin vivo. We developed potent catalytic inhibitors of TAO3 that inhibited invadopodia formation and function, and tumor cell extravasation and growth. Using these inhibitors, we determined that TAO3 activity was required for endosomal trafficking of TKS5α, an obligate invadopodia scaffold protein. A phosphoproteomics screen for TAO3 substrates revealed the dynein subunit protein LIC2 as a relevant substrate. Knockdown of LIC2 or expression of a phosphomimetic form promoted invadopodia formation. Thus, TAO3 is a new therapeutic target with a distinct mechanism of action.SIGNIFICANCETargeting tumor invasive behavior represents an understudied opportunity. We used an unbiased screening approach to identify kinases required for invadopodia formation and function. We validated TAO3, both genetically and with a novel inhibitor, and determined TAO3 function. Our data support clinical development of this class of target.
- Published
- 2020
- Full Text
- View/download PDF
15. RNF5 Defines Acute Myeloid Leukemia Growth and Susceptibility to Histone Deacetylase Inhibitors
- Author
-
Yongmei Feng, Carol Burian, Ian Pass, James R. Mason, Yan Li, Tongwu Zhang, Yishai Ofran, Ronit Almog, Eytan Ruppin, Kristiina Vuori, Darren Finlay, Joo Sang Lee, Ali Khateb, Ido Livneh, Yu Fujita, Bertrand Fabre, Aniruddha J. Deshpande, Nurit Horesh, Kevin K. Brown, Anagha Deshpande, Michael R. Jackson, Ikrame Lazar, and Ze'ev Ronai
- Subjects
business.industry ,hemic and lymphatic diseases ,Immunology ,Cancer research ,Myeloid leukemia ,Medicine ,Cell Biology ,Hematology ,Histone deacetylase ,business ,neoplasms ,Biochemistry - Abstract
Acute myeloid leukemia (AML) remains an incurable blood cancer largely due to rapid emergence of resistance to conventional treatments. Thus, new therapeutic modalities are greatly needed to halt AML development. Here, using genetic and xenograft mouse models, we reveal that inhibition of the ubiquitin ligase RNF5 in human AML cell lines and in MLL-AF9-driven AML severely decreased the leukemogenic potential of those cells and prolonged survival of model leukemic mice. These findings suggest the possibility that targeting a single gene, namely RNF5, could effectively inhibit different AML subtypes. We initially focused on RNF5 as its expression is upregulated in AML patient cohorts as well as in AML-derived cell lines compared with normal hematopoietic cells. Furthermore, high RNF5 expression in AML patient specimens correlated with poor prognosis, relapse and short overall patient survival. By contrast, specimens from AML patients who responded to therapy exhibited low RNF5 levels. In vitro, RNF5 loss impaired the clonogenic potential of MLL-AF9-transduced bone marrow cells and markedly attenuated growth and survival of AML but not CML or T-ALL cell lines, in which RNF5 is also highly expressed. High-throughput screen and bioinformatics analysis identified RNF5 and ER-associated degradation (ERAD) components, as augmenting AML cell sensitivity to histone deacetylase (HDAC) inhibition. Indeed, inhibition of RNF5 sensitized AML cells to HDAC inhibitors. Correspondingly, a favorable prognosis was observed in AML patients exhibiting low expression of RNF5 and HDAC. Collectivity, our studies identify a potential new therapeutic modality based on targeting RNF5 to inhibit AML and suggest that RNF5 expression could serve as a prognostic marker and means to stratify patients for treatment with HDAC inhibitors. Disclosures Ofran: AbbVie: Membership on an entity's Board of Directors or advisory committees. Vuori:Bionano Genomics: Membership on an entity's Board of Directors or advisory committees.
- Published
- 2020
- Full Text
- View/download PDF
16. Improving drug discovery using image-based multiparametric analysis of the epigenetic landscape
- Author
-
Jarkko Ylanko, David W. Andrews, Alexey V. Terskikh, Fu-Yue Zeng, E. Camilla Forsberg, Fernando Ugarte, Santosh Hariharan, Luis Orozco, Ian Pass, Chen Farhy, and Chun-Teng Huang
- Subjects
0301 basic medicine ,Computer science ,Genome ,Epigenesis, Genetic ,Histones ,Machine Learning ,chemistry.chemical_compound ,0302 clinical medicine ,Drug Discovery ,Image Processing, Computer-Assisted ,Biology (General) ,Cancer Biology ,biology ,Brain Neoplasms ,Drug discovery ,General Neuroscience ,General Medicine ,Neoplasm Proteins ,Histone ,Drug development ,030220 oncology & carcinogenesis ,High-content screening ,Medicine ,Research Article ,Human ,QH301-705.5 ,Science ,Antineoplastic Agents ,Computational biology ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Cell Line, Tumor ,Biomarkers, Tumor ,Humans ,Epigenetics ,Human Biology and Medicine ,Gene ,Cell Nucleus ,Dose-Response Relationship, Drug ,high content screening ,epigenetics ,General Immunology and Microbiology ,glioblastoma ,High-Throughput Screening Assays ,030104 developmental biology ,Microscopy, Fluorescence ,chemistry ,biology.protein ,DNA - Abstract
High-content phenotypic screening has become the approach of choice for drug discovery due to its ability to extract drug-specific multi-layered data. In the field of epigenetics, such screening methods have suffered from a lack of tools sensitive to selective epigenetic perturbations. Here we describe a novel approach, Microscopic Imaging of Epigenetic Landscapes (MIEL), which captures the nuclear staining patterns of epigenetic marks and employs machine learning to accurately distinguish between such patterns. We validated the MIEL platform across multiple cells lines and using dose-response curves, to insure the fidelity and robustness of this approach for high content high throughput drug discovery. Focusing on noncytotoxic glioblastoma treatments, we demonstrated that MIEL can identify and classify epigenetically active drugs. Furthermore, we show MIEL was able to accurately rank candidate drugs by their ability to produce desired epigenetic alterations consistent with increased sensitivity to chemotherapeutic agents or with induction of glioblastoma differentiation., eLife digest Each cell contains a complete copy of a person’s genes coded in their DNA. However, for a cell to perform its specific role, it only needs a small fraction of this genetic information. The mechanisms that control which genes a cell is using fall under the umbrella of ‘epigenetics’ (meaning beyond genetics). These mechanisms involve changes in the way that DNA is organized inside the cell nucleus and changes in how accessible different parts of the genome are to various cellular components. DNA is long and fragile so, to maintain its integrity, it is wrapped around protein complexes called histones. Adding chemical modifications to histones is one of the main epigenetic mechanisms that cells use to regulate which genes are turned on and off. Several methods allow researchers to read patterns of histone modification and use this information to derive what state a cell is in and how it might behave. Improving these methods is of particular interest in drug development, where this information could reveal the effects, and side-effects, of new treatments. Unfortunately, existing techniques are costly in both time and money, and they are not well suited to analyzing epigenetic changes caused by the large numbers of compounds tested during drug development. To overcome this barrier, Farhy et al. have developed a new system called ‘Microscopic Imaging of the Epigenetic Landscape’ (MIEL for short). The system allows them to quickly analyze the epigenetic changes caused by each of a large number of different chemical compounds when they are used on cells. MIEL tags different histone modifications by staining each with a different color, and then uses automated microscopy to produce images. It then extracts information from these images using advanced image analysis tools. The changes induced by different drugs can then be compared and categorized using machine learning algorithms. To test the MIEL system, Farhy et al. grew brain cancer cells (derived from human tumors) in the lab, and treated them with compounds that target proteins involved in histone modifications. Using their newly created pipeline, Farhy et al. were able to identify the unique epigenetic changes caused by these compounds, and train the system to correctly predict which type of drug the cells had been treated with. In a different set of experiments Farhy et al. demonstrate the utility of their new pipeline in identifying drugs that induce a set of epigenetic changes associated with a reduced ability to regrow tumors. This new system could help screen thousands of compounds for their epigenetic effects, which could aid the design of new treatments for many diseases.
- Published
- 2019
- Full Text
- View/download PDF
17. Author response: Improving drug discovery using image-based multiparametric analysis of the epigenetic landscape
- Author
-
Fernando Ugarte, Ian Pass, Fu-Yue Zeng, David W. Andrews, Alexey V. Terskikh, Chun-Teng Huang, Luis Orozco, Santosh Hariharan, E. Camilla Forsberg, Jarkko Ylanko, and Chen Farhy
- Subjects
Computer science ,Drug discovery ,Multiparametric Analysis ,Computational biology ,Epigenetics ,Image based - Published
- 2019
- Full Text
- View/download PDF
18. Improving drug discovery using image-based multiparametric analysis of epigenetic landscape
- Author
-
Fu-Yue Zeng, Ian Pass, Fernando Ugarte, Jarkko Ylanko, David W. Andrews, Alexey V. Terskikh, Santosh Hariharan, Luis Orozco, E. Camilla Forsberg, Chun-Teng Huang, and Chen Farhy
- Subjects
0303 health sciences ,Mechanism (biology) ,Drug discovery ,Phenotypic screening ,Robustness (evolution) ,Computational biology ,Biology ,3. Good health ,Chemical library ,Bromodomain ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Histone ,chemistry ,030220 oncology & carcinogenesis ,biology.protein ,Epigenetics ,030304 developmental biology - Abstract
With the advent of automatic cell imaging and machine learning, high-content phenotypic screening has become the approach of choice for drug discovery because it can extract drug-specific multi-layered data, which could be compared to known profiles. In the field of epigenetics, such screening approaches have suffered from a lack of tools sensitive to selective epigenetic perturbations. Here we describe a novel approach, Microscopic Imaging of Epigenetic Landscapes (MIEL), which captures the nuclear staining patterns of epigenetic marks (e.g., acetylated and methylated histones) and employs machine learning to accurately distinguish between such patterns. We validated the fidelity and robustness of the MIEL platform across multiple cells lines using dose-response curves. We employed MIEL to uncover the mechanism by which bromodomain inhibitors synergize with temozolomide-mediated killing of human glioblastoma lines. To explore alternative, non-cytotoxic, glioblastoma treatment, we screen the Prestwick chemical library and documented the power of MIEL platform to identify epigenetically active drugs and accurately rank them according to their ability to produce epigenetic and transcriptional alterations consistent with the induction of glioblastoma differentiation.
- Published
- 2019
- Full Text
- View/download PDF
19. Capzimin is a potent and specific inhibitor of proteasome isopeptidase Rpn11
- Author
-
Gavin Magnuson, Seth M. Cohen, Yuyong Ma, Brock T. Brown, Christian Perez, Ian Pass, Kevin Nguyen, Kyle P. Carter, Andrew L. Mackinnon, Stefan Vasile, Sonja Hess, Jing Li, Anthony B. Pinkerton, Tanya R. Yakushi, Eduard Sergienko, Layton H. Smith, Eigo Suyama, Sharon A. Colayco, Francesco Parlati, Thomas D.Y. Chung, Raymond J. Deshaies, and Amy E. Palmer
- Subjects
0301 basic medicine ,Proteases ,Proteasome Endopeptidase Complex ,Biology ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Structure-Activity Relationship ,0302 clinical medicine ,medicine ,Humans ,Molecular Biology ,Multiple myeloma ,Dose-Response Relationship, Drug ,Molecular Structure ,Bortezomib ,Cell Biology ,medicine.disease ,Carfilzomib ,030104 developmental biology ,Proteostasis ,chemistry ,Proteasome ,Biochemistry ,030220 oncology & carcinogenesis ,Cancer cell ,Unfolded protein response ,Cancer research ,Quinolines ,Trans-Activators ,Proteasome Inhibitors ,medicine.drug - Abstract
The proteasome is a vital cellular machine that maintains protein homeostasis, which is of particular importance in multiple myeloma and possibly other cancers. Targeting of proteasome 20S peptidase activity with bortezomib and carfilzomib has been widely used to treat myeloma. However, not all patients respond to these compounds, and those who do eventually suffer relapse. Therefore, there is an urgent and unmet need to develop new drugs that target proteostasis through different mechanisms. We identified quinoline-8-thiol (8TQ) as a first-in-class inhibitor of the proteasome 19S subunit Rpn11. A derivative of 8TQ, capzimin, shows >5-fold selectivity for Rpn11 over the related JAMM proteases and >2 logs selectivity over several other metalloenzymes. Capzimin stabilized proteasome substrates, induced an unfolded protein response, and blocked proliferation of cancer cells, including those resistant to bortezomib. Proteomic analysis revealed that capzimin stabilized a subset of polyubiquitinated substrates. Identification of capzimin offers an alternative path to develop proteasome inhibitors for cancer therapy.
- Published
- 2016
20. TR-FRET-Based High-Throughput Screening Assay for Identification of UBC13 Inhibitors
- Author
-
Charitha Madiraju, Shu-ichi Matsuzawa, John C. Reed, Tram A. Ngo, Eduard Sergienko, Stefan Vasile, Kate Welsh, Ian Pass, Paul Diaz, P.H.C. Godoi, and Michael Cuddy
- Subjects
Cell signaling ,biology ,High-throughput screening ,Ubiquitination ,Chemosensitizer ,Small Molecule Libraries ,Ubiquitin-conjugating enzyme ,Biochemistry ,Article ,High-Throughput Screening Assays ,Analytical Chemistry ,Chemical library ,chemistry.chemical_compound ,Förster resonance energy transfer ,Ubiquitin ,chemistry ,Ubiquitin-Conjugating Enzymes ,Fluorescence Resonance Energy Transfer ,biology.protein ,Molecular Medicine ,Polyubiquitin ,Biotechnology - Abstract
UBC13 is a noncanonical ubiquitin conjugating enzyme (E2) that has been implicated in a variety of cellular signaling processes due to its ability to catalyze formation of lysine 63-linked polyubiquitin chains on various substrates. In particular, UBC13 is required for signaling by a variety of receptors important in immune regulation, making it a candidate target for inflammatory diseases. UBC13 is also critical for double-strand DNA repair and thus a potential radiosensitizer and chemosensitizer target for oncology. The authors developed a high-throughput screening (HTS) assay for UBC13 based on the method of time-resolved fluorescence resonance energy transfer (TR-FRET). The TR-FRET assay combines fluorochrome (Fl)-conjugated ubiquitin (fluorescence acceptor) with terbium (Tb)-conjugated ubiquitin (fluorescence donor), such that the assembly of mixed chains of Fl- and Tb-ubiquitin creates a robust TR-FRET signal. The authors defined conditions for optimized performance of the TR-FRET assay in both 384- and 1536-well formats. Chemical library screens (total 456 865 compounds) were conducted in high-throughput mode using various compound collections, affording superb Z' scores (typically >0.7) and thus validating the performance of the assays. Altogether, the HTS assays described here are suitable for large-scale, automated screening of chemical libraries in search of compounds with inhibitory activity against UBC13.
- Published
- 2012
- Full Text
- View/download PDF
21. The Novel Adaptor Protein Tks4 (SH3PXD2B) Is Required for Functional Podosome Formation
- Author
-
Sara A. Courtneidge, Fang Wen, Ian Pass, Pilar Cejudo-Martin, Paul Andrew Bromann, and Matthew D. Buschman
- Subjects
Podosome ,Biology ,Cell Membrane Structures ,Cell Line ,Extracellular matrix ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cell Movement ,Animals ,Humans ,Cloning, Molecular ,Phosphorylation ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,030304 developmental biology ,0303 health sciences ,Signal transducing adaptor protein ,Tyrosine phosphorylation ,Articles ,Cell Biology ,Phosphate-Binding Proteins ,Lipid Metabolism ,Phosphoproteins ,Protein Structure, Tertiary ,Cell biology ,chemistry ,030220 oncology & carcinogenesis ,Invadopodia ,Tyrosine kinase ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Metastatic cancer cells have the ability to both degrade and migrate through the extracellular matrix (ECM). Invasiveness can be correlated with the presence of dynamic actin-rich membrane structures called podosomes or invadopodia. We showed previously that the adaptor protein tyrosine kinase substrate with five Src homology 3 domains (Tks5)/Fish is required for podosome/invadopodia formation, degradation of ECM, and cancer cell invasion in vivo and in vitro. Here, we describe Tks4, a novel protein that is closely related to Tks5. This protein contains an amino-terminal Phox homology domain, four SH3 domains, and several proline-rich motifs. In Src-transformed fibroblasts, Tks4 is tyrosine phosphorylated and predominantly localized to rosettes of podosomes. We used both short hairpin RNA knockdown and mouse embryo fibroblasts lacking Tks4 to investigate its role in podosome formation. We found that lack of Tks4 resulted in incomplete podosome formation and inhibited ECM degradation. Both phenotypes were rescued by reintroduction of Tks4, whereas only podosome formation, but not ECM degradation, was rescued by overexpression of Tks5. The tyrosine phosphorylation sites of Tks4 were required for efficient rescue. Furthermore, in the absence of Tks4, membrane type-1 matrix metalloproteinase (MT1-MMP) was not recruited to the incomplete podosomes. These findings suggest that Tks4 and Tks5 have overlapping, but not identical, functions, and implicate Tks4 in MT1-MMP recruitment and ECM degradation.
- Published
- 2009
- Full Text
- View/download PDF
22. A role for the podosome/invadopodia scaffold protein Tks5 in tumor growth in vivo
- Author
-
Sara A. Courtneidge, Barbara Blouw, Ian Pass, Begoña Díaz, and Darren F. Seals
- Subjects
Histology ,Podosome ,Angiogenesis ,Biology ,Transfection ,Article ,Pathology and Forensic Medicine ,Metastasis ,src Homology Domains ,Mice ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Humans ,Neovascularization, Pathologic ,Microfilament Proteins ,Cell Biology ,General Medicine ,Phosphate-Binding Proteins ,Phosphoproteins ,medicine.disease ,Immunohistochemistry ,Extravasation ,Cell biology ,Adaptor Proteins, Vesicular Transport ,Tumor progression ,Invadopodia ,Cancer cell ,NIH 3T3 Cells ,Cell Surface Extensions - Abstract
Podosomes and invadopodia are electron-dense, actin-rich protrusions located on the ventral side of the cellular membrane. They are detected in various types of normal cells, but also in human cancer cells and in Src-transformed fibroblasts. Previously we have shown that the scaffold protein Tks5 (tyrosine kinase substrate 5) co-localizes to podosomes/invadopodia in different human cancer cells and in Src-transformed NIH-3T3 cells. Upon reduced expression of Tks5 podosome formation is decreased, which leads to diminished gelatin degradation in vitro in various human cancer cell lines. It is unclear, however, whether cancer cells need podosomes for tumor growth and metastasis in vivo. To test this idea, we evaluated the ability of Src-transformed NIH-3T3 cells, showing stable reduced expression of Tks5 and podosome formation (Tks5 KD), to form subcutaneous tumors in mice. We demonstrate that decreased expression of Tks5 correlated with reduced tumor growth at this site. In addition, we generated lung metastases from these cells following tail vein injection. The lungs of mice injected i.v. with the Tks5 KD showed smaller-sized metastases, but there was no difference in the number of lesions compared to the controls, indicating that podosomes may not be required for extravasation from the blood stream into the lung parenchyma. Independent of the microenvironment however, the reduced tumor growth correlated with decreased tumor vascularization. Our data potentially implicate a novel role of podosomes as mediators of tumor angiogenesis and support further exploration of how podosome formation and Tks5 expression contribute to tumor progression.
- Published
- 2008
- Full Text
- View/download PDF
23. Cell-Based High-Throughput Luciferase Reporter Gene Assays for Identifying and Profiling Chemical Modulators of Endoplasmic Reticulum Signaling Protein, IRE1
- Author
-
John C. Reed, Shu-ichi Matsuzawa, Nicholas D. P. Cosford, Gavin Magnuson, Michael R. Jackson, Ian Pass, Juan Rong, Michelle Sauer, Tram A. Ngo, Fu-Yue Zeng, Christian A. Hassig, and Paul Diaz
- Subjects
X-Box Binding Protein 1 ,XBP1 ,MAP Kinase Signaling System ,Regulatory Factor X Transcription Factors ,Biology ,Protein Serine-Threonine Kinases ,Biochemistry ,Analytical Chemistry ,Chemical library ,Small Molecule Libraries ,chemistry.chemical_compound ,Genes, Reporter ,Cell Line, Tumor ,High-Throughput Screening Assays ,Endoribonucleases ,Humans ,Luciferase ,Luciferases ,Transcription factor ,Neurons ,Reporter gene ,Endoplasmic reticulum ,Endoplasmic Reticulum Stress ,Cell biology ,DNA-Binding Proteins ,Enzyme Activation ,chemistry ,Unfolded protein response ,Molecular Medicine ,Thapsigargin ,Biotechnology ,HeLa Cells ,Transcription Factors - Abstract
Endoplasmic reticulum (ER) stress activates three distinct signal transducers on the ER membrane. Inositol-requiring protein 1 (IRE1), the most conserved signal transducer, plays a key role in ER stress-mediated signaling. During ER stress, IRE1 initiates two discrete signaling cascades: the "adaptive" signaling cascade mediated by the XBP1 pathway and the "alarm" signaling cascade mediated by stress-activated protein kinase pathways. Fine-tuning of the balance between the adaptive and alarm signals contributes significantly to cellular fate under ER stress. Thus, we propose that the design of high-throughput screening (HTS) assays to selectively monitor IRE1 mediated-signaling would be desirable for drug discovery. To this end, we report the generation of stable human neural cell lines and development of cell-based HTS luciferase (Luc) reporter gene assays for the identification of pathway-specific chemical modulators of IRE1. We implemented a cell-based Luc assay using a chimeric CHOP-Gal4 transcription factor in 384-well format for monitoring IRE1 kinase-mediated p38MAPK activation and an unfolded response pathway element (URPE)-Luc cell-based assay in 1536-well format for monitoring IRE1's RNase-mediated activation of XBP1. Chemical library screening was successfully conducted with both the CHOP/Gal4-Luc cells and UPRE-Luc engineered cells. The studies demonstrate the feasibility of using these HTS assays for discovery of pathway-selective modulators of IRE1.
- Published
- 2015
24. The adaptor protein Tks5/Fish is required for podosome formation and function, and for the protease-driven invasion of cancer cells
- Author
-
Rebecca Gordon, Sara A. Courtneidge, James H. Resau, Lia Tesfay, Melissa Woodrow, Eduardo F. Azucena, Ian Pass, and Darren F. Seals
- Subjects
Cancer Research ,Podosome ,Invadopodium ,Breast Neoplasms ,Biology ,Cell Line ,src Homology Domains ,Extracellular matrix ,Mice ,Cell Line, Tumor ,Neoplasms ,Animals ,Humans ,Neoplasm Invasiveness ,RNA, Small Interfering ,Melanoma ,Signal transducing adaptor protein ,Cell Biology ,Extracellular Matrix ,Protein Structure, Tertiary ,Cell biology ,Adaptor Proteins, Vesicular Transport ,src-Family Kinases ,Microscopy, Fluorescence ,Oncology ,Cell culture ,Invadopodia ,Cancer cell ,NIH 3T3 Cells ,Chickens ,Peptide Hydrolases ,Proto-oncogene tyrosine-protein kinase Src - Abstract
SummaryTks5/Fish is a scaffolding protein with five SH3 domains and one PX domain. In Src-transformed cells, Tks5/Fish localizes to podosomes, discrete protrusions of the ventral membrane. We generated Src-transformed cells with reduced Tks5/Fish levels. They no longer formed podosomes, did not degrade gelatin, and were poorly invasive. We detected Tks5/Fish expression in podosomes in invasive cancer cells, as well as in human breast cancer and melanoma samples. Tks5/Fish expression was also required for protease-driven matrigel invasion in human cancer cells. Finally, coexpression of Tks5/Fish and Src in epithelial cells resulted in the appearance of podosomes. Thus, Tks5/Fish appears to be required for podosome formation, for degradation of the extracellular matrix, and for invasion of some cancer cells.
- Published
- 2005
- Full Text
- View/download PDF
25. The Src Substrate Tks5, Podosomes (Invadopodia), and Cancer Cell Invasion
- Author
-
Sara A. Courtneidge, E.F. Azucena, Ian Pass, Lia Tesfay, and Darren F. Seals
- Subjects
Matrigel ,Small interfering RNA ,Podosome ,Chemistry ,PX domain ,Biochemistry ,Protein Structure, Tertiary ,Substrate Specificity ,Cell biology ,Adaptor Proteins, Vesicular Transport ,Mice ,src-Family Kinases ,Cell Line, Tumor ,Invadopodia ,Cancer cell ,Genetics ,Animals ,Humans ,Female ,Neoplasm Invasiveness ,Cell Surface Extensions ,Molecular Biology ,Tyrosine kinase ,Protein Binding ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Some years ago, we employed a screen of phage cDNA expression libraries to identify novel substrates of the protein tyrosine kinase Src. One of these, Tks5 (previously known as Fish), is a large scaffolding protein with an amino-terminal PX domain and five SH3 domains. In normal fibroblasts, Tks5 is cytoplasmic, but the protein is found in podosomes when the cells are transformed with Src. Using short interfering RNA technology, we have shown that Tks5 is required for podosome formation. Furthermore, cells with reduced Tks5 expression are poorly invasive through Matrigel. Tks5 is expressed and localized to podosomes in invasive human cancer cell lines and in tumor tissue, particularly breast cancers and melanomas. In these cells too, Tks5 is required for invasion. Our future work will focus on the identification of the binding partners of Tks5 that are responsible for podosome formation and invasion, and on determining the role of Tks5 in animal models of metastasis.
- Published
- 2005
- Full Text
- View/download PDF
26. Interfacial kinetic analysis of the tumour suppressor phosphatase, PTEN: evidence for activation by anionic phospholipids
- Author
-
Ian Pass, C. Peter Downes, Steven Walker, and George McConnachie
- Subjects
Anions ,Phosphatase ,Phospholipid ,Biochemistry ,Catalysis ,chemistry.chemical_compound ,Phosphatidylinositol Phosphates ,Phosphatidylcholine ,Tensin ,PTEN ,Molecular Biology ,Phospholipids ,C2 domain ,chemistry.chemical_classification ,biology ,Hydrolysis ,Tumor Suppressor Proteins ,PTEN Phosphohydrolase ,Substrate (chemistry) ,Cell Biology ,Phosphoric Monoester Hydrolases ,Kinetics ,Enzyme ,chemistry ,biology.protein ,Research Article - Abstract
We investigated the kinetic behaviour and substrate specificity of PTEN (phosphatase and tensin homologue deleted on chromosome 10) using unilamellar vesicles containing substrate lipids in a background of phosphatidylcholine. PTEN displays the characteristics expected of an interfacial enzyme, since the rate of enzyme activity is dependent on the surface concentration of the substrate lipids used (mol fraction), as well as the bulk concentration. Surface-dilution analysis revealed the catalytic efficiency of PTEN for PtdIns(3,4,5) P (3) to be 200-fold greater than for either PtdIns(3,4) P (2) or PtdIns(3,5) P (2), and 1000-fold greater than for PtdIns3 P. The interfacial K (m) value of PTEN for PtdIns(3,4,5) P (3) was very low, reflecting the small proportions of this lipid that are present in cellular membranes. The catalytic-centre activity ( k (cat)) for PtdIns(3,4,5) P (3) was at least 200-fold greater than that for the water-soluble substrate Ins(1,3,4,5) P (4). The preference for lipid substrates may result from an interfacial activation of the enzyme, rather than processive catalysis of vesicular substrates. Moreover, both PtdIns(4,5) P (2) and univalent salts stimulated the activity of PTEN for PtdIns(3,4,5) P (3), but profoundly inhibited activity against Ins(1,3,4,5) P (4). The stimulatory effect of PtdIns(4,5) P (2) was greater in magnitude and more potent in comparison with other anionic phospholipid species. A mutation in the lipid-binding C2 domain (M-CBR3) that is biologically inactive did not alter overall catalytic efficiency in this model, but decreased the efficiency of the interfacial binding step, demonstrating its importance in the catalytic mechanism of PTEN.
- Published
- 2003
- Full Text
- View/download PDF
27. The Adaptor Protein Fish Associates with Members of the ADAMs Family and Localizes to Podosomes of Src-transformed Cells
- Author
-
Sara A. Courtneidge, Therese M. Roth, Lisa M. Maurer, Clare L. Abram, Ian Pass, Daniel Salinsky, and Darren F. Seals
- Subjects
Podosome ,Invadopodium ,Biology ,Biochemistry ,SH3 domain ,src Homology Domains ,Mice ,Animals ,Molecular Biology ,Cell Line, Transformed ,Membrane Proteins ,Metalloendopeptidases ,Signal transducing adaptor protein ,3T3 Cells ,Cell Biology ,PX domain ,Phosphate-Binding Proteins ,Phosphoproteins ,Fusion protein ,Cell biology ,Genes, src ,COS Cells ,Invadopodia ,Protein Binding ,Signal Transduction ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Fish is a scaffolding protein and Src substrate. It contains an amino-terminal Phox homology (PX) domain and five Src homology 3 (SH3) domains, as well as multiple motifs for binding both SH2 and SH3 domain-containing proteins. We have determined that the PX domain of Fish binds 3-phosphorylated phosphatidylinositols (including phosphatidylinositol 3-phosphate and phosphatidylinositol 3,4-bisphosphate). Consistent with this, a fusion protein of green fluorescent protein and the Fish PX domain localized to punctate structures similar to endosomes in normal fibroblasts. However, the full-length Fish protein was largely cytoplasmic, suggesting that its PX domain may not be able to make intermolecular interactions in unstimulated cells. In Src-transformed cells, we observed a dramatic re-localization of some Fish molecules to actin-rich structures called podosomes; the PX domain was both necessary and sufficient to effect this translocation. We used a phage display screen with the fifth SH3 domain of Fish and isolated ADAM19 as a binding partner. Subsequent analyses in mammalian cells demonstrated that Fish interacts with several members of the ADAMs family, including ADAMs 12, 15, and 19. In Src-transformed cells, ADAM12 co-localized with Fish in podosomes. Because members of the ADAMs family have been implicated in growth factor processing, as well as cell adhesion and motility, Fish could be acting as an adaptor molecule that allows Src to impinge on these processes.
- Published
- 2003
- Full Text
- View/download PDF
28. Identification of a selective inhibitor of murine intestinal alkaline phosphatase (ML260) by concurrent ultra-high throughput screening against human and mouse isozymes
- Author
-
Russell Dahl, Sonoko Narisawa, Shenghua Shi, Craig Rosenstein, Stefan Vasile, Ying Su, Jiwen Zou, Santhi Ganji, Ian Pass, Anton Cheltsov, Tina Kiffer-Moreira, José Luis Millán, Carlton Gasior, Anthony B. Pinkerton, Flávia Godoy Iano, Brock Brown, Justin Rascon, Jonathan D. Kaunitz, Eduard Sergienko, Marc Hoylaerts, Robert Ardecky, Michael P Hedrick, Thomas D.Y. Chung, Ekaterina V. Bobkova, and Anita H. Forster
- Subjects
High-throughput screening ,1.1 Normal biological development and functioning ,Medicinal & Biomolecular Chemistry ,Clinical Biochemistry ,Pharmaceutical Science ,Biochemistry ,Isozyme ,Article ,Inflammatory bowel disease ,Intestinal alkaline phosphatase ,Dephosphorylation ,Mice ,Medicinal and Biomolecular Chemistry ,Underpinning research ,Drug Discovery ,medicine ,Animals ,Humans ,Protein Isoforms ,2.1 Biological and endogenous factors ,Secretion ,Enzyme Inhibitors ,Aetiology ,Molecular Biology ,Sulfonamides ,biology ,Chemistry ,Inhibitors ,Organic Chemistry ,Pharmacology and Pharmaceutical Sciences ,biology.organism_classification ,Alkaline Phosphatase ,Molecular biology ,In vitro ,Enzymes ,body regions ,Enzyme Activation ,medicine.anatomical_structure ,Molecular Medicine ,Alkaline phosphatase ,Acetanilides ,Germ cell ,Bacteria - Abstract
Alkaline phosphatase (AP) isozymes are present in a wide range of species from bacteria to man and are capable of dephosphorylation and transphosphorylation of a wide spectrum of substrates in vitro. In humans, four AP isozymes have been identified—one tissue-nonspecific (TNAP) and three tissue-specific—named according to the tissue of their predominant expression: intestinal (IAP), placental (PLAP) and germ cell (GCAP) APs. Modulation of activity of the different AP isozymes may have therapeutic implications in distinct diseases and cellular processes. For instance, changes in the level of IAP activity can affect gut mucosa tolerance to microbial invasion due to the ability of IAP to detoxify bacterial endotoxins, alter the absorption of fatty acids and affect ectopurinergic regulation of duodenal bicarbonate secretion. To identify isozyme selective modulators of the human and mouse IAPs, we developed a series of murine duodenal IAP (Akp3-encoded dIAP isozyme), human IAP (hIAP), PLAP, and TNAP assays. High throughput screening and subsequent SAR efforts generated a potent inhibitor of dIAP, ML260, with specificity for the Akp3-, compared to the Akp5- and Akp6-encoded mouse isozymes.
- Published
- 2014
29. Antagonism of PI 3-kinase-dependent signalling pathways by the tumour suppressor protein, PTEN
- Author
-
Lisa Patel, Deborah Bennett, G. McConnachie, Alexander Gray, Ian Pass, C P Downes, Nick R. Leslie, and Colin H. Macphee
- Subjects
Phosphatase ,Protein tyrosine phosphatase ,Biology ,Phosphatidylinositol 3-Kinases ,Biochemistry ,Cell biology ,chemistry.chemical_compound ,chemistry ,biology.protein ,Tensin ,PTEN ,Inositol ,Phosphatidylinositol ,Signal transduction - Abstract
The tumour suppressor protein, PTEN (phosphatase and tensin homologue deleted on chromosome 10) is a member of the mixed function, serine/threonine/tyrosine phosphatase subfamily of protein phosphatases. Its physiological substrates, however, are primarily 3-phosphorylated inositol phospholipids, which are products of phosphoinositide 3-kinases. PTEN thus antagonizes PI 3-kinase-dependent signalling pathways, which explains to a large extent its tumour suppressor status. We have examined the kinetic behaviour, substrate specificity and regulation of PTEN both in vitro and in a variety of cellular models. Although PTEN can utilize both phosphatidylinositol 3,4,5-trisphosphate [PtdIns-(3,4,5)P3] and its water-soluble headgroup, inositol 1,3,4,5-tetrakisphosphate, as substrates, it displays classical features of interfacial catalysis, which greatly favour the lipid substrate (by as much as 1000-fold as judged by Kcat/Km values). Expression of PTEN in U87 cells (which lack endogenous PTEN) and measuring the levels of all known 3-phosphorylated lipids suggests that phosphatidylinositol 3,4-bisphosphate and PtdIns(3,4,5)P3 are both substrates, but that phosphatidylinositol 3-phosphate and phosphatidyl-inositol 3,5-bisphosphate are not. PTEN binds to several PDZ-domain-containing proteins via a consensus sequence at its extreme C-terminus. Disruption of targeting to PDZ-domain proteins selectively blocks some PTEN functions, but not others, suggesting the existence of spatially localized, functionally dedicated pools of signalling lipids. We have also shown recently that PTEN expression is controlled at the transcriptional level and is profoundly upregulated by peroxisome proliferator activated receptor γ agonists, thereby providing possible implications for these drugs in diabetes, inflammation and cancer.
- Published
- 2001
- Full Text
- View/download PDF
30. Targeting mutants of PTEN reveal distinct subsets of tumour suppressor functions
- Author
-
Nick R. LESLIE, Deborah BENNETT, Alex GRAY, Ian PASS, Khe HOANG-XUAN, and C. Peter DOWNES
- Subjects
Cell Biology ,Molecular Biology ,Biochemistry - Abstract
The tumour suppressor protein PTEN (phosphatase and tensin homolog deleted on chromosome 10) is a lipid phosphatase which can antagonize the phosphoinositide 3-kinase (PI 3-kinase) signalling pathway, promoting apoptosis and inhibiting cell-cycle progression and cell motility. We show that very little cellular PTEN is associated with the plasma membrane, but that artificial membrane-targeting of PTEN enhances its inhibition of signalling to protein kinase B (PKB). Evidence for potential targeting of PTEN to the membrane through PDZ domain-mediated protein–protein interactions led us to use a PTEN enzyme with a deletion of the C-terminal PDZ-binding sequence, that retains full phosphatase activity against soluble substrates, and to analyse the efficiency of this mutant in different cellular assays. The extreme C-terminal PDZ-binding sequence was dispensable for the efficient down-regulation of cellular PtdIns(3,4,5)P3 levels and a number of PI 3-kinase-dependent signalling activities, including PKB and p70S6K. However, the PDZ-binding sequence was required for the efficient inhibition of cell spreading. The data show that a PTEN mutation, similar to those found in some tumours, affects some functions of the protein but not others, and implicate the deregulation of PTEN-dependent processes other than PKB activation in the development of some tumours. Significantly, this hypothesis is supported by data showing low levels of PKB phosphorylation in a glioblastoma sample carrying a mutation in the extreme C-terminus of PTEN compared with tumours carrying phosphatase-inactivating mutations of the enzyme. Our data show that deregulation of PKB is not a universal feature of tumours carrying PTEN mutations and implicate other processes that may be deregulated in these tumours.
- Published
- 2001
- Full Text
- View/download PDF
31. Analysis of the cellular functions of PTEN using catalytic domain and C-terminal mutations: differential effects of C-terminal deletion on signalling pathways downstream of phosphoinositide 3-kinase
- Author
-
Elaine A. Orchiston, C. Peter Downes, Nick R. Leslie, Ian Pass, and Alexander Gray
- Subjects
Phosphoinositide 3-kinase ,biology ,Membrane ruffling ,Phosphatase ,Cell Biology ,Biochemistry ,Cell biology ,Lipid phosphatase activity ,biology.protein ,PTEN ,Tensin ,Molecular Biology ,Protein kinase B ,Platelet-derived growth factor receptor - Abstract
The tumour suppressor protein, PTEN (phosphatase and tensin homolog deleted on chromosome 10), is a phosphatase that can dephosphorylate tyrosine-containing peptides, Shc, focal adhesion kinase and phosphoinositide substrates. In cellular assays, PTEN has been shown to antagonize the PI-3K-dependent activation of protein kinase B (PKB) and to inhibit cell spreading and motility. It is currently unclear, however, whether PTEN accomplishes these effects through its lipid- or protein-phosphatase activity, although strong evidence has demonstrated the importance of the latter for tumour suppression by PTEN. By using a PTEN G129E (Gly129 → Glu) mutant that has lost its lipid phosphatase activity, while retaining protein phosphatase activity, we demonstrated a requirement for the lipid phosphatase activity of PTEN in the regulation of PKB activity, cell viability and membrane ruffling. We also made a small C-terminal deletion of PTEN, removing a putative PDZ (PSD95, Dlg and ZO1)-binding motif, with no detectable effect on the phosphatase activity of the protein expressed in HEK293 cells (human embryonic kidney 293 cells) assayed in vitro. Surprisingly, expression of this mutant revealed differential requirements for the C-terminus in the different functional assays. Wild-type and C-terminally deleted PTEN appeared to be equally active in down-regulating PKB activity, but this mutant enzyme had no effect on platelet-derived growth factor (PDGF)-induced membrane ruffling and was only partially active in a cell viability assay. These results stress the importance of the lipid phosphatase activity of PTEN in the regulation of several signalling pathways. They also identify a mutation, similar to mutations that occur in some human tumours, which removes the effect of PTEN on membrane ruffling but not that on PKB.
- Published
- 2000
- Full Text
- View/download PDF
32. Tks5-dependent, nox-mediated generation of reactive oxygen species is necessary for invadopodia formation
- Author
-
Sara A. Courtneidge, Begoña Díaz, Manuela Quintavalle, Gidon Shani, Diana Anderson, and Ian Pass
- Subjects
inorganic chemicals ,Transfection ,Biochemistry ,Article ,Cell Line ,chemistry.chemical_compound ,Mice ,Cell Line, Tumor ,Animals ,Humans ,Phosphorylation ,RNA, Small Interfering ,Molecular Biology ,chemistry.chemical_classification ,Feedback, Physiological ,Reactive oxygen species ,NADPH oxidase ,biology ,NADPH Oxidases ,Tyrosine phosphorylation ,Cell Biology ,Phosphate-Binding Proteins ,Cytochrome b Group ,Phosphoproteins ,Cell biology ,Adaptor Proteins, Vesicular Transport ,chemistry ,Invadopodia ,biology.protein ,cardiovascular system ,Cell Surface Extensions ,Reactive Oxygen Species ,Tyrosine kinase ,Nicotinamide adenine dinucleotide phosphate ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Invadopodia are actin-rich membrane protrusions of cancer cells that facilitate pericellular proteolysis and invasive behavior. We show here that reactive oxygen species (ROS) generated by the NADPH (reduced form of nicotinamide adenine dinucleotide phosphate) oxidase (Nox) system are necessary for invadopodia formation and function. Knockdown of the invadopodia protein Tks5 [tyrosine kinase substrate with five Src homology 3 (SH3) domains], which is structurally related to the Nox component p47 phox , reduces total ROS abundance in cancer cells. Furthermore, Tks5 and p22 phox can associate with each other, suggesting that Tks5 is part of the Nox complex. Tyrosine phosphorylation of Tks5 and Tks4, but not other Src substrates, is reduced by Nox inhibition. We propose that Tks5 facilitates the production of ROS necessary for invadopodia formation, and that in turn ROS modulate Tks5 tyrosine phosphorylation in a positive feedback loop.
- Published
- 2009
33. Nck adaptor proteins link Tks5 to invadopodia actin regulation and ECM degradation
- Author
-
San San Xu, Peter Lock, Ian Pass, Sara A. Courtneidge, T. T.I. Stacey, Anne M. Verhagen, and Stanley S. Stylli
- Subjects
Podosome ,Invadopodium ,Blotting, Western ,Fluorescent Antibody Technique ,macromolecular substances ,Biology ,Transfection ,src Homology Domains ,Mice ,Neoplasms ,Tumor Cells, Cultured ,NCK2 ,Animals ,Humans ,Immunoprecipitation ,RNA, Small Interfering ,Adaptor Proteins, Signal Transducing ,Oncogene Proteins ,Signal transducing adaptor protein ,Cell Biology ,PX domain ,Phosphate-Binding Proteins ,Phosphoproteins ,Actins ,Cell biology ,Extracellular Matrix ,Invadopodia ,Cell Surface Extensions ,Extracellular Matrix Degradation ,Proto-oncogene tyrosine-protein kinase Src ,Research Article - Abstract
Invadopodia are actin-based projections enriched with proteases, which invasive cancer cells use to degrade the extracellular matrix (ECM). The Phox homology (PX)-Src homology (SH)3 domain adaptor protein Tks5 (also known as SH3PXD2A) cooperates with Src tyrosine kinase to promote invadopodia formation but the underlying pathway is not clear. Here we show that Src phosphorylates Tks5 at Y557, inducing it to associate directly with the SH3-SH2 domain adaptor proteins Nck1 and Nck2 in invadopodia. Tks5 mutants unable to bind Nck show reduced matrix degradation-promoting activity and recruit actin to invadopodia inefficiently. Conversely, Src- and Tks5-driven matrix proteolysis and actin assembly in invadopodia are enhanced by Nck1 or Nck2 overexpression and inhibited by Nck1 depletion. We show that clustering at the plasma membrane of the Tks5 inter-SH3 region containing Y557 triggers phosphorylation at this site, facilitating Nck recruitment and F-actin assembly. These results identify a Src-Tks5-Nck pathway in ECM-degrading invadopodia that shows parallels with pathways linking several mammalian and pathogen-derived proteins to local actin regulation.
- Published
- 2009
34. Tumor suppressor and anti-inflammatory actions of PPARgamma agonists are mediated via upregulation of PTEN
- Author
-
Lisa Patel, C. Peter Downes, Phil Coxon, Ian Pass, Stephen A. Smith, and Colin H. Macphee
- Subjects
Tumor suppressor gene ,Anti-Inflammatory Agents ,Receptors, Cytoplasmic and Nuclear ,Antineoplastic Agents ,General Biochemistry, Genetics and Molecular Biology ,Rosiglitazone ,Downregulation and upregulation ,Tumor Cells, Cultured ,PTEN ,Humans ,Genes, Tumor Suppressor ,Protein kinase A ,biology ,Agricultural and Biological Sciences(all) ,Pioglitazone ,Cell growth ,Biochemistry, Genetics and Molecular Biology(all) ,Tumor Suppressor Proteins ,PTEN Phosphohydrolase ,Cell migration ,Phosphoric Monoester Hydrolases ,Up-Regulation ,Thiazoles ,Nuclear receptor ,Monocyte differentiation ,Cancer research ,biology.protein ,Thiazolidinediones ,General Agricultural and Biological Sciences ,Transcription Factors - Abstract
The PTEN tumor suppressor gene modulates several cellular functions, including cell migration, survival, and proliferation [1] by antagonizing phosphatidylinositol 3-kinase (PI 3-kinase)-mediated signaling cascades. Mechanisms by which the expression of PTEN is regulated are, however, unclear. The ligand-activated nuclear receptor peroxisome proliferator-activated receptor gamma (PPARgamma) [2] has been shown to regulate differentiation and/or cell growth in a number of cell types [3, 4, 5], which has led to the suggestion that PPARgamma, like PTEN [1, 6], could act as a tumor suppressor. PPARgamma has also been implicated in anti-inflammatory responses [7, 8], although downstream mediators of these effects are not well defined. Here, we show that the activation of PPARgamma by its selective ligand, rosiglitazone, upregulates PTEN expression in human macrophages, Caco2 colorectal cancer cells, and MCF7 breast cancer cells. This upregulation correlated with decreased PI 3-kinase activity as measured by reduced phosphorylation of protein kinase B. One consequence of this was that rosiglitazone treatment reduced the proliferation rate of Caco2 and MCF7 cells. Antisense-mediated disruption of PPARgamma expression prevented the upregulation of PTEN that normally accompanies monocyte differentiation and reduced the proportion of macrophages undergoing apoptosis, while electrophoretic mobility shift assays showed that PPARgamma is able to bind two response elements in the genomic sequence upstream of PTEN. Our results demonstrate a role for PPARgamma in regulating PI 3-kinase signaling by modulating PTEN expression in inflammatory and tumor-derived cells.
- Published
- 2001
35. Functional Evaluation of the Benzodiazepine Cholecystokinin Type-B Receptor Antagonists L-365,260, L-740,093, and YM022
- Author
-
John Dunlop, Chris Ennis, Neil Brammer, Ian Pass, and Non Evans
- Subjects
education.field_of_study ,Benzodiazepine ,medicine.drug_class ,digestive, oral, and skin physiology ,Population ,Pharmacology ,Neurotransmission ,Biology ,digestive system ,Cholecystokinin receptor ,medicine.anatomical_structure ,Neuromodulation ,Cholecystokinin B receptor ,medicine ,education ,Receptor ,hormones, hormone substitutes, and hormone antagonists ,Cholecystokinin - Abstract
Two identified receptor subtypes, designated type-A and type-B (1), mediate the physiological actions of the brain/gut peptide cholecystokinin (CCK). In the central nervous system the CCK-B receptor represents the predominant receptor population and a central role for CCK in mediating neurotransmission and/or neuromodulation has been proposed (2). Additionally, the CCK-B receptor has been implicated in a number of psychiatric disorders including panic attacks (3) and anxiety (4). Consequently, this receptor subtype has received considerable attention as a therapeutic target with a number of selective, high-affinity ligands now available. In particular, a number of molecules incorporating a benzodiazepine template have been developed as selective high-affinity CCK-B receptor antagonists. Examples of these are L-365,260 (5), L-740,093 (6) and YM022 (7) which represent three structurally similar (Fig. 1) members of this class.
- Published
- 1997
- Full Text
- View/download PDF
36. Discovery and Characterization of Chemical Inhibitors of UBC13
- Author
-
Charitha Madiraju, Thomas D.Y. Chung, Carina Wimer, Chris Hassig, Robert Ardecky, John C. Reed, Chih-Wen Shu, Michael Cuddy, Anthony B. Pinkerton, Craig M. Tamble, Tram A. Ngo, Eduard Sergienko, Correa Ricardo, Shu-ichi Matsuzawa, Ian Pass, Yasuko Matsuzawa, and Paul Diaz
- Subjects
Protein sumoylation ,Toll-like receptor ,DNA repair ,Immunology ,SUMO protein ,Cell Biology ,Hematology ,Ubiquitin-conjugating enzyme ,Biology ,Biochemistry ,Ubiquitin ,biology.protein ,Neddylation ,Signal transduction - Abstract
Abstract 2950 Poly-ubiquitination of signaling proteins via lysine 63 (K63)-linked chains is recognized as a critical post-translational modification involved in activation of NF-kB and stress kinases in the context of signaling by Tumor Necrosis Factor Receptors (TNFRs), Toll-like receptors (TLRs), and antigen receptors. UBC13 is a K63-specific ubiquitin conjugating enzyme that partners with TNFR-associated factors (TRAFs) to mediate K63-linked ubiquitination. Gene ablation studies have shown UBC13 is required for NF-kB signaling induced by a variety of stimuli in specific types of immune cells, making it a potential target for certain cancers, autoimmune and inflammatory diseases. UBC13 operates together with obligatory cofactors, either UEV1A in the cytosol or MMS2 in the nucleus. The nuclear function of UBC13 is evolutionarily conserved, where it plays a critical role in double strand DNA repair, making UBC13 a potential chemo- and radio-sensitizer target for oncology. To identify chemical inhibitors of UBC13, we developed a HTS assay measuring UBC13-UEV1A enzymatic activity by TR-FRET, screening altogether ∼450,000 diverse compounds. Hit compounds were characterized using a rigorous testing funnel consisting of (a) informatics filtering against a database of > 100 HTS campaigns conducted with the same libraries, to eliminate promiscuous compounds; (b) counter-screens against E1, another cysteine-dependent enzyme (caspase-3), and against an irrelevant target formatted as a TR-FRET assay; and (c) ordering compounds from fresh powders and demonstrating reproducible concentration-dependent inhibition of UBC13. The surviving hits were then analyzed by cell-based assays for suppression of TRAF6 ubiquitination but not Mdm2-mediated ubiquitination of p53, resulting in 14 promising hits that included two chemical series. While suppressing TRAF6 ubiquitination (UBC13-dependent) in cells, these compounds did not interfere with either SUMOylation (UBC9-dependent) or NEDDylation (UBC12-dependent) of cellular proteins. UBC13 inhibitors also suppressed NF-kB activity (measured using stably integrated NF-kB-driven luciferase reporter gene) induced by PKC activators (Carma/Bcl-10/MALT pathway) and DNA damaging agent (Doxorubicin) but not by TNF-a. Investigations of the bioactivity of these UBC13 inhibitory compounds and their analogs will be described for a variety of hematolymphoid malignancies. (Supported by NIH R03-MH085677, NIH U54–005033, and by a fellowship grant from International Myeloma Foundation). Disclosures: No relevant conflicts of interest to declare.
- Published
- 2012
- Full Text
- View/download PDF
37. Characterisation of a PTEN homologue from Drosophila
- Author
-
G. McConnachie, Nick R. Leslie, C P Downes, and Ian Pass
- Subjects
biology ,biology.protein ,PTEN ,Drosophila (subgenus) ,biology.organism_classification ,Biochemistry ,Cell biology - Published
- 2001
- Full Text
- View/download PDF
38. PTEN, A Tumour Suppresser Is A 3 Phosphorylated Phosphoinositide 3 Phosphatase
- Author
-
C. Peter Downes, Ian Pass, and Ian H. Batty
- Subjects
biology ,Chemistry ,Phosphatase ,Cancer research ,biology.protein ,PTEN ,Phosphorylation ,Biochemistry - Published
- 1999
- Full Text
- View/download PDF
39. INFLAMMATORY DISORDERS ASSOCIATED WITH HELICOBACTER PYLORI IN THE ROUX-EN-Y BYPASS GASTRIC POUCH
- Author
-
Luiz Claudio Lopes CHAVES, Isabela Klautau Leite Chaves BORGES, Maíra Danielle Gomes de SOUZA, Ian Passos SILVA, Lyz Bezerra SILVA, Marcelo Alexandre Prado MAGALHÃES, Allan Herbert Feliz FONSECA, and Josemberg Marins CAMPOS
- Subjects
Helicobacter pylori ,Gastrointestinal diseases ,Gastric Bypass ,Obesity ,Surgery ,RD1-811 ,Diseases of the digestive system. Gastroenterology ,RC799-869 - Abstract
ABSTRACT Background: The prevalence of Helicobacter pylori in obese candidates for bariatric surgery and its role in the emergence of inflammatory lesions after surgery has not been well established. Aim: To identify the incidence of inflammatory lesions in the stomach after bariatric surgery and to correlate it with H. pylori infection. Methods: This is a prospective study with 216 patients undergoing Roux-en-Y gastric bypass. These patients underwent histopathological endoscopy to detect H. pylori prior to surgery. Positive cases were treated with antibiotics and a proton inhibitor pump followed by endoscopic follow-up in the 6th and 12th month after surgery. Results: Most patients were female (68.1%), with grade III obesity (92.4%). Preoperative endoscopy revealed gastritis in 96.8%, with H. pylori infection in 40.7% (88/216). A biopsy was carried out in 151 patients, revealing H. pylori in 60/151, related to signs of inflammation in 90% (54/60). In the 6th and 12th month after surgery, the endoscopy and the histopathological exam showed a normal gastric pouch in 84% of patients and the incidence of H. pylori was 11% and 16%, respectively. The presence of inflammation was related to H. pylori infection (p
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.