91 results on '"C. Marcelo Aldaz"'
Search Results
2. Supplementary Methods and Figures 1-5 from Genomic Alterations during the In Situ to Invasive Ductal Breast Carcinoma Transition Shaped by the Immune System
- Author
-
Kornelia Polyak, Joe Gray, Catherine Wu, Joon Jeong, So Yeon Park, C. Marcelo Aldaz, Bojana Jovanović, Jennifer Eng, Guillaume Thibault, Young Hwan Chang, Koei Chin, Sachet A. Shukla, Carlos R. Gil Del Alcazar, and Anne Trinh
- Abstract
S1. Overview of patient cohort. S2. Whole exome sequencing of matched DCISand IDC transition. S3. Mutationalprofiling of the DCIS-to-IDC transition. S4.Phenotypic immune properties of patient cohort. S5.Immune-related genomicchanges and neoantigen load.
- Published
- 2023
- Full Text
- View/download PDF
3. Data from Genomic Alterations during the In Situ to Invasive Ductal Breast Carcinoma Transition Shaped by the Immune System
- Author
-
Kornelia Polyak, Joe Gray, Catherine Wu, Joon Jeong, So Yeon Park, C. Marcelo Aldaz, Bojana Jovanović, Jennifer Eng, Guillaume Thibault, Young Hwan Chang, Koei Chin, Sachet A. Shukla, Carlos R. Gil Del Alcazar, and Anne Trinh
- Abstract
The drivers of ductal carcinoma in situ (DCIS) to invasive ductal carcinoma (IDC) transition are poorly understood. Here, we conducted an integrated genomic, transcriptomic, and whole-slide image analysis to evaluate changes in copy-number profiles, mutational profiles, expression, neoantigen load, and topology in 6 cases of matched pure DCIS and recurrent IDC. We demonstrate through combined copy-number and mutational analysis that recurrent IDC can be genetically related to its pure DCIS despite long latency periods and therapeutic interventions. Immune “hot” and “cold” tumors can arise as early as DCIS and are subtype-specific. Topologic analysis showed a similar degree of pan-leukocyte-tumor mixing in both DCIS and IDC but differ when assessing specific immune subpopulations such as CD4 T cells and CD68 macrophages. Tumor-specific copy-number aberrations in MHC-I presentation machinery and losses in 3p, 4q, and 5p are associated with differences in immune signaling in estrogen receptor (ER)-negative IDC. Common oncogenic hotspot mutations in genes including TP53 and PIK3CA are predicted to be neoantigens yet are paradoxically conserved during the DCIS-to-IDC transition, and are associated with differences in immune signaling. We highlight both tumor and immune-specific changes in the transition of pure DCIS to IDC, including genetic changes in tumor cells that may have a role in modulating immune function and assist in immune escape, driving the transition to IDC.Implications:We demonstrate that the in situ to IDC evolutionary bottleneck is shaped by both tumor and immune cells.
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Table S1 from Breast Cancer Molecular Signatures as Determined by SAGE: Correlation with Lymph Node Status
- Author
-
C. Marcelo Aldaz, Aysegul Sahin, Steve Horvath, Tao Shi, Sally Gaddis, Maria I. Nunez, Yuhui Hu, Jeffrey A. Drake, Kathleen A. Hawkins, Hongxia Sun, and Martín C. Abba
- Abstract
Supplementary Table S1 from Breast Cancer Molecular Signatures as Determined by SAGE: Correlation with Lymph Node Status
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Materials 1 from Identification of Modulated Genes by Three Classes of Chemopreventive Agents at Preneoplastic Stages in a p53-Null Mouse Mammary Tumor Model
- Author
-
C. Marcelo Aldaz, Daniel Medina, Powel H. Brown, Reid P. Bissonnette, Jamal Hill, Frances S. Kittrell, Sally Gaddis, Carla C. Levy, Yuhui Hu, and Martín C. Abba
- Abstract
Supplementary Materials 1 from Identification of Modulated Genes by Three Classes of Chemopreventive Agents at Preneoplastic Stages in a p53-Null Mouse Mammary Tumor Model
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Table S2 from Breast Cancer Molecular Signatures as Determined by SAGE: Correlation with Lymph Node Status
- Author
-
C. Marcelo Aldaz, Aysegul Sahin, Steve Horvath, Tao Shi, Sally Gaddis, Maria I. Nunez, Yuhui Hu, Jeffrey A. Drake, Kathleen A. Hawkins, Hongxia Sun, and Martín C. Abba
- Abstract
Supplementary Table S2 from Breast Cancer Molecular Signatures as Determined by SAGE: Correlation with Lymph Node Status
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Table S3 from Breast Cancer Molecular Signatures as Determined by SAGE: Correlation with Lymph Node Status
- Author
-
C. Marcelo Aldaz, Aysegul Sahin, Steve Horvath, Tao Shi, Sally Gaddis, Maria I. Nunez, Yuhui Hu, Jeffrey A. Drake, Kathleen A. Hawkins, Hongxia Sun, and Martín C. Abba
- Abstract
Supplementary Table S3 from Breast Cancer Molecular Signatures as Determined by SAGE: Correlation with Lymph Node Status
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Tables 1-7 from Genomic Alterations during the In Situ to Invasive Ductal Breast Carcinoma Transition Shaped by the Immune System
- Author
-
Kornelia Polyak, Joe Gray, Catherine Wu, Joon Jeong, So Yeon Park, C. Marcelo Aldaz, Bojana Jovanović, Jennifer Eng, Guillaume Thibault, Young Hwan Chang, Koei Chin, Sachet A. Shukla, Carlos R. Gil Del Alcazar, and Anne Trinh
- Abstract
Summary of Supplementary Tables Supplementary Table S1. Clinicopathological characteristics of patient cohort Supplementary Table S2. Antibody panel for cyclic IF Supplementary Table S3. Summary of mutations and neoantigen expression in the recurrence cohort (Related to 1C, 6A) Supplementary Table S4. Summary of altered pathways in DCIS/IDC (Fig 1F) Supplementary Table S5. Enriched gene sets between DCIS and IDC (Fig 3A) Supplementary Table S6. Identified chromosomal regions associated with immune changes (Related to Fig. 5) Supplementary Table S7. Mutational, neoantigen and rsSNP frequency of commonly mutated genes in TCGA and Abba cohorts (related to Fig 6C,E)
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Materials 3 from Identification of Modulated Genes by Three Classes of Chemopreventive Agents at Preneoplastic Stages in a p53-Null Mouse Mammary Tumor Model
- Author
-
C. Marcelo Aldaz, Daniel Medina, Powel H. Brown, Reid P. Bissonnette, Jamal Hill, Frances S. Kittrell, Sally Gaddis, Carla C. Levy, Yuhui Hu, and Martín C. Abba
- Abstract
Supplementary Materials 3 from Identification of Modulated Genes by Three Classes of Chemopreventive Agents at Preneoplastic Stages in a p53-Null Mouse Mammary Tumor Model
- Published
- 2023
- Full Text
- View/download PDF
10. Data from Breast Cancer Molecular Signatures as Determined by SAGE: Correlation with Lymph Node Status
- Author
-
C. Marcelo Aldaz, Aysegul Sahin, Steve Horvath, Tao Shi, Sally Gaddis, Maria I. Nunez, Yuhui Hu, Jeffrey A. Drake, Kathleen A. Hawkins, Hongxia Sun, and Martín C. Abba
- Abstract
Global gene expression measured by DNA microarray platforms have been extensively used to classify breast carcinomas correlating with clinical characteristics, including outcome. We generated a breast cancer Serial Analysis of Gene Expression (SAGE) high-resolution database of ∼2.7 million tags to perform unsupervised statistical analyses to obtain the molecular classification of breast-invasive ductal carcinomas in correlation with clinicopathologic features. Unsupervised statistical analysis by means of a random forest approach identified two main clusters of breast carcinomas, which differed in their lymph node status (P = 0.01); this suggested that lymph node status leads to globally distinct expression profiles. A total of 245 (55 up-modulated and 190 down-modulated) transcripts were differentially expressed between lymph node (+) and lymph node (−) primary breast tumors (fold change, ≥2; P < 0.05). Various lymph node (+) up-modulated transcripts were validated in independent sets of human breast tumors by means of real-time reverse transcription-PCR (RT-PCR). We validated significant overexpression of transcripts for HOXC10 (P = 0.001), TPD52L1 (P = 0.007), ZFP36L1 (P = 0.011), PLINP1 (P = 0.013), DCTN3 (P = 0.025), DEK (P = 0.031), and CSNK1D (P = 0.04) in lymph node (+) breast carcinomas. Moreover, the DCTN3 (P = 0.022) and RHBDD2 (P = 0.002) transcripts were confirmed to be overexpressed in tumors that recurred within 6 years of follow-up by real-time RT-PCR. In addition, meta-analysis was used to compare SAGE data associated with lymph node (+) status with publicly available breast cancer DNA microarray data sets. We have generated evidence indicating that the pattern of gene expression in primary breast cancers at the time of surgical removal could discriminate those tumors with lymph node metastatic involvement using SAGE to identify specific transcripts that behave as predictors of recurrence as well. (Mol Cancer Res 2007;5(9):881–90)
- Published
- 2023
- Full Text
- View/download PDF
11. Related Article from Identification of Modulated Genes by Three Classes of Chemopreventive Agents at Preneoplastic Stages in a p53-Null Mouse Mammary Tumor Model
- Author
-
C. Marcelo Aldaz, Daniel Medina, Powel H. Brown, Reid P. Bissonnette, Jamal Hill, Frances S. Kittrell, Sally Gaddis, Carla C. Levy, Yuhui Hu, and Martín C. Abba
- Abstract
Related Article from Identification of Modulated Genes by Three Classes of Chemopreventive Agents at Preneoplastic Stages in a p53-Null Mouse Mammary Tumor Model
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Table S2 from A Molecular Portrait of High-Grade Ductal Carcinoma In Situ
- Author
-
C. Marcelo Aldaz, Aysegul A. Sahin, Marcos R. Estecio, Jianjun Shen, Sally Gaddis, Yoko Takata, Matias Butti, Ezequiel Lacunza, Yi Zhong, Jaeho Lee, Yue Lu, Ting Gong, and Martin C. Abba
- Abstract
Supplementary Table S2: List of mutated genes, Indels and Copy number variations detected in DCIS samples.
- Published
- 2023
- Full Text
- View/download PDF
13. Data from From Mice to Humans
- Author
-
C. Marcelo Aldaz, Daniel Medina, Keith Baggerly, Aysegul Sahin, Sally Gaddis, Li Deng, Martin C. Abba, Frances Kittrell, Jeffrey Drake, Hongxia Sun, and Yuhui Hu
- Abstract
Genetically engineered mouse mammary cancer models have been used over the years as systems to study human breast cancer. However, much controversy exists on the utility of such models as valid equivalents to the human cancer condition. To perform an interspecies gene expression comparative study in breast cancer we used a mouse model that most closely resembles human breast carcinogenesis. This system relies on the transplant of p53 null mouse mammary epithelial cells into the cleared mammary fat pads of syngeneic hosts. Serial analysis of gene expression (SAGE) was used to obtain gene expression profiles of normal and tumor samples from this mouse mammary cancer model (>300,000 mouse mammary-specific tags). The resulting mouse data were compared with 25 of our human breast cancer SAGE libraries (>2.5 million human breast-specific tags). We observed significant similarities in the deregulation of specific genes and gene families when comparing mouse with human breast cancer SAGE data. A total of 72 transcripts were identified as commonly deregulated in both species. We observed a systematic and significant down-regulation in all of the tumors from both species of various cytokines, including CXCL1 (GRO1), LIF, interleukin 6, and CCL2. All of the mouse and most human mammary tumors also displayed decreased expression of genes known to inhibit cell proliferation, including NFKBIA (IKBα), GADD45B, and CDKN1A (p21); transcription-related genes such as CEBP, JUN, JUNB, and ELF1; and apoptosis-related transcripts such as IER3 and GADD34/PPP1R15A. Examples of overexpressed transcripts in tumors from both species include proliferation-related genes such as CCND1, CKS1B, and STMN1 (oncoprotein 18); and genes related to other functions such as SEPW1, SDFR1, DNCI2, and SP110. Importantly, abnormal expression of several of these genes has not been associated previously with breast cancer. The consistency of these observations was validated in independent mouse and human mammary cancer sets.This is the first interspecies comparison of mammary cancer gene expression profiles. The comparative analysis of mouse and human SAGE mammary cancer data validates this p53 null mouse tumor model as a useful system closely resembling human breast cancer development and progression. More importantly, these studies are allowing us to identify relevant biomarkers of potential use in human studies while leading to a better understanding of specific mechanisms of human breast carcinogenesis.
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Table and Figure Legends from A Molecular Portrait of High-Grade Ductal Carcinoma In Situ
- Author
-
C. Marcelo Aldaz, Aysegul A. Sahin, Marcos R. Estecio, Jianjun Shen, Sally Gaddis, Yoko Takata, Matias Butti, Ezequiel Lacunza, Yi Zhong, Jaeho Lee, Yue Lu, Ting Gong, and Martin C. Abba
- Abstract
Supplementary Table and Figure Legends: Legend for Supplementary Tables S1-S5 and Supplementary Figures S1-S4.
- Published
- 2023
- Full Text
- View/download PDF
15. Supplementary Figure S3 from A Molecular Portrait of High-Grade Ductal Carcinoma In Situ
- Author
-
C. Marcelo Aldaz, Aysegul A. Sahin, Marcos R. Estecio, Jianjun Shen, Sally Gaddis, Yoko Takata, Matias Butti, Ezequiel Lacunza, Yi Zhong, Jaeho Lee, Yue Lu, Ting Gong, and Martin C. Abba
- Abstract
Supplementary Figure S3: TP53 pathway activity in invasive breast carcinomas from the TCGA-BRCA dataset.
- Published
- 2023
- Full Text
- View/download PDF
16. Supplementary Table 3 from From Mice to Humans
- Author
-
C. Marcelo Aldaz, Daniel Medina, Keith Baggerly, Aysegul Sahin, Sally Gaddis, Li Deng, Martin C. Abba, Frances Kittrell, Jeffrey Drake, Hongxia Sun, and Yuhui Hu
- Abstract
Supplementary Table 3 from From Mice to Humans
- Published
- 2023
- Full Text
- View/download PDF
17. Supplementary Table 1 from From Mice to Humans
- Author
-
C. Marcelo Aldaz, Daniel Medina, Keith Baggerly, Aysegul Sahin, Sally Gaddis, Li Deng, Martin C. Abba, Frances Kittrell, Jeffrey Drake, Hongxia Sun, and Yuhui Hu
- Abstract
Supplementary Table 1 from From Mice to Humans
- Published
- 2023
- Full Text
- View/download PDF
18. Supplementary Figure S4 from A Molecular Portrait of High-Grade Ductal Carcinoma In Situ
- Author
-
C. Marcelo Aldaz, Aysegul A. Sahin, Marcos R. Estecio, Jianjun Shen, Sally Gaddis, Yoko Takata, Matias Butti, Ezequiel Lacunza, Yi Zhong, Jaeho Lee, Yue Lu, Ting Gong, and Martin C. Abba
- Abstract
Supplementary Figure S4: SOX10, SOX15 and SOX17 mRNA expression among normal and invasive breast carcinomas from TCGA dataset classified according the intrinsic subtypes.
- Published
- 2023
- Full Text
- View/download PDF
19. Supplementary Figure S1 from A Molecular Portrait of High-Grade Ductal Carcinoma In Situ
- Author
-
C. Marcelo Aldaz, Aysegul A. Sahin, Marcos R. Estecio, Jianjun Shen, Sally Gaddis, Yoko Takata, Matias Butti, Ezequiel Lacunza, Yi Zhong, Jaeho Lee, Yue Lu, Ting Gong, and Martin C. Abba
- Abstract
Supplementary Figure S1: Chromosome regions more frequently affected in DCIS samples as determined by Exome-Seq data analysis using Control-FREEC software.
- Published
- 2023
- Full Text
- View/download PDF
20. Supplementary Figure S2 from A Molecular Portrait of High-Grade Ductal Carcinoma In Situ
- Author
-
C. Marcelo Aldaz, Aysegul A. Sahin, Marcos R. Estecio, Jianjun Shen, Sally Gaddis, Yoko Takata, Matias Butti, Ezequiel Lacunza, Yi Zhong, Jaeho Lee, Yue Lu, Ting Gong, and Martin C. Abba
- Abstract
Supplementary Figure S2: Network analysis of transcripts associated with each DCIS clusters.
- Published
- 2023
- Full Text
- View/download PDF
21. Supplementary Table S3 from A Molecular Portrait of High-Grade Ductal Carcinoma In Situ
- Author
-
C. Marcelo Aldaz, Aysegul A. Sahin, Marcos R. Estecio, Jianjun Shen, Sally Gaddis, Yoko Takata, Matias Butti, Ezequiel Lacunza, Yi Zhong, Jaeho Lee, Yue Lu, Ting Gong, and Martin C. Abba
- Abstract
Supplementary Table S3: Differentially expressed transcripts and PARADIGM inferences of the top 300 most variable integrated pathway activities between normal and DCIS samples.
- Published
- 2023
- Full Text
- View/download PDF
22. Supplementary Table S1 from A Molecular Portrait of High-Grade Ductal Carcinoma In Situ
- Author
-
C. Marcelo Aldaz, Aysegul A. Sahin, Marcos R. Estecio, Jianjun Shen, Sally Gaddis, Yoko Takata, Matias Butti, Ezequiel Lacunza, Yi Zhong, Jaeho Lee, Yue Lu, Ting Gong, and Martin C. Abba
- Abstract
Supplementary Table S1: Description of samples employed for Exome-Seq, RNA-Seq and RRBS analysis.
- Published
- 2023
- Full Text
- View/download PDF
23. Supplementary Table 2 from From Mice to Humans
- Author
-
C. Marcelo Aldaz, Daniel Medina, Keith Baggerly, Aysegul Sahin, Sally Gaddis, Li Deng, Martin C. Abba, Frances Kittrell, Jeffrey Drake, Hongxia Sun, and Yuhui Hu
- Abstract
Supplementary Table 2 from From Mice to Humans
- Published
- 2023
- Full Text
- View/download PDF
24. Supplementary Table S5 from A Molecular Portrait of High-Grade Ductal Carcinoma In Situ
- Author
-
C. Marcelo Aldaz, Aysegul A. Sahin, Marcos R. Estecio, Jianjun Shen, Sally Gaddis, Yoko Takata, Matias Butti, Ezequiel Lacunza, Yi Zhong, Jaeho Lee, Yue Lu, Ting Gong, and Martin C. Abba
- Abstract
Supplementary Table S5: Differentially methylated CpGi regions among normal and DCIS samples and their correlation with gene expression.
- Published
- 2023
- Full Text
- View/download PDF
25. WWOX P47T loss-of-function mutation induces epilepsy, progressive neuroinflammation, and cerebellar degeneration in mice phenocopying human SCAR12
- Author
-
Tabish Hussain, Kevin Sanchez, Jennifer Crayton, Dhurjhoti Saha, Collene Jeter, Yue Lu, Martin Abba, Ryan Seo, Jeffrey L Noebels, Laura Fonken, and C Marcelo Aldaz
- Abstract
WWOX gene loss-of-function (LoF) has been associated with neuropathologies resulting in developmental, epileptic, and ataxic phenotypes of varying severity based on the level of WWOX dysfunction. WWOX gene biallelic germline variant p.Pro47Thr (P47T) has been causally associated with a new form of autosomal recessive cerebellar ataxia with epilepsy and intellectual disability (SCAR12). This mutation affects the WW1 protein binding domain of WWOX, impairing its ability to interact with canonical proline-proline-X-tyrosine motifs in partner proteins. We generated a mutant knock-in mouse model of Wwox P47T that phenocopies SCAR12. WwoxP47T/P47T mice displayed epilepsy, profound social behavior and cognition deficits, and poor motor coordination, and unlike KO models that survive only for 1 month, live beyond 1 year of age. These deficits progressed with age, and mice became practically immobile, suggesting severe cerebellar dysfunction. WwoxP47T/P47T mice exhibited signs of progressive neuroinflammation with elevated astro-microgliosis that increased with age. The cerebellar cortex displayed significantly reduced molecular and granular layer thickness and a strikingly reduced number of Purkinje cells with degenerated dendrites. Transcriptome profiling from various brain regions from these Wwox LoF mice highlighted widespread changes in neuronal and glial pathways, enrichment of bioprocesses related to neuroinflammation and severe cerebellar dysfunction, activation of pathways compatible with compensatory neurogenesis along with major suppression of gene networks associated with excitability, neuronal cell differentiation and brain development. Our results show significant pathobiological effects and potential mechanisms through which WWOX LoF leads to epilepsy, cerebellar neurodegeneration, neuroinflammation, and ataxia. Additionally, the mouse model described here will be a useful tool for the study of WWOX in common neurodegenerative conditions in which it has been identified as a novel risk factor.
- Published
- 2022
- Full Text
- View/download PDF
26. Genomic Alterations during the In Situ to Invasive Ductal Breast Carcinoma Transition Shaped by the Immune System
- Author
-
Joe W. Gray, Young Hwan Chang, Koei Chin, Anne Trinh, Carlos R. Gil Del Alcazar, C. Marcelo Aldaz, Jennifer Eng, Joon Jeong, Bojana Jovanovic, Sachet A. Shukla, Kornelia Polyak, Catherine J. Wu, Guillaume Thibault, and So Yeon Park
- Subjects
0301 basic medicine ,In situ ,Cancer Research ,biology ,CD68 ,Estrogen receptor ,Ductal carcinoma ,Major histocompatibility complex ,body regions ,Transcriptome ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,skin and connective tissue diseases ,neoplasms ,Molecular Biology ,Gene - Abstract
The drivers of ductal carcinoma in situ (DCIS) to invasive ductal carcinoma (IDC) transition are poorly understood. Here, we conducted an integrated genomic, transcriptomic, and whole-slide image analysis to evaluate changes in copy-number profiles, mutational profiles, expression, neoantigen load, and topology in 6 cases of matched pure DCIS and recurrent IDC. We demonstrate through combined copy-number and mutational analysis that recurrent IDC can be genetically related to its pure DCIS despite long latency periods and therapeutic interventions. Immune “hot” and “cold” tumors can arise as early as DCIS and are subtype-specific. Topologic analysis showed a similar degree of pan-leukocyte-tumor mixing in both DCIS and IDC but differ when assessing specific immune subpopulations such as CD4 T cells and CD68 macrophages. Tumor-specific copy-number aberrations in MHC-I presentation machinery and losses in 3p, 4q, and 5p are associated with differences in immune signaling in estrogen receptor (ER)-negative IDC. Common oncogenic hotspot mutations in genes including TP53 and PIK3CA are predicted to be neoantigens yet are paradoxically conserved during the DCIS-to-IDC transition, and are associated with differences in immune signaling. We highlight both tumor and immune-specific changes in the transition of pure DCIS to IDC, including genetic changes in tumor cells that may have a role in modulating immune function and assist in immune escape, driving the transition to IDC. Implications: We demonstrate that the in situ to IDC evolutionary bottleneck is shaped by both tumor and immune cells.
- Published
- 2020
- Full Text
- View/download PDF
27. Wwox Binding to the Murine Brca1-BRCT Domain Regulates Timing of Brip1 and CtIP Phospho-Protein Interactions with This Domain at DNA Double-Strand Breaks, and Repair Pathway Choice
- Author
-
Dongju Park, Mehdi Gharghabi, Colleen R. Reczek, Rebecca Plow, Charles Yungvirt, C. Marcelo Aldaz, and Kay Huebner
- Subjects
Mammals ,Endodeoxyribonucleases ,endocrine system diseases ,DNA Repair ,BRCA1 Protein ,Organic Chemistry ,DNA Helicases ,Cell Cycle Proteins ,General Medicine ,DNA ,end-resection at DSBs ,Wwox deficiency ,Chk2 inhibition ,IR and platinum treatment resistance ,Catalysis ,Computer Science Applications ,Inorganic Chemistry ,Mice ,WW Domain-Containing Oxidoreductase ,Animals ,DNA Breaks, Double-Stranded ,Physical and Theoretical Chemistry ,Carrier Proteins ,Homologous Recombination ,Molecular Biology ,Spectroscopy - Abstract
Wwox-deficient human cells show elevated homologous recombination, leading to resistance to killing by double-strand break-inducing agents. Human Wwox binds to the Brca1 981-PPLF-984 Wwox-binding motif, likely blocking the pChk2 phosphorylation site at Brca1-S988. This phosphorylation site is conserved across mammalian species; the PPLF motif is conserved in primates but not in rodents. We now show that murine Wwox does not bind Brca1 near the conserved mouse Brca1 phospho-S971 site, leaving it open for Chk2 phosphorylation and Brca1 activation. Instead, murine Wwox binds to Brca1 through its BRCT domain, where pAbraxas, pBrip1, and pCtIP, of the A, B, and C binding complexes, interact to regulate double-strand break repair pathway response. In Wwox-deficient mouse cells, the Brca1-BRCT domain is thus accessible for immediate binding of these phospho-proteins. We confirm elevated homologous recombination in Wwox-silenced murine cells, as in human cells. Wwox-deficient murine cells showed increased ionizing radiation-induced Abraxas, Brca1, and CtIP foci and long resected single-strand DNA, early after ionizing radiation. Wwox deletion increased the basal level of Brca1-CtIP interaction and the expression level of the MRN-CtIP protein complex, key players in end-resection, and facilitated Brca1 release from foci. Inhibition of phospho-Chk2 phosphorylation of Brca1-S971 delays the end-resection; the delay of premature end-resection by combining Chk2 inhibition with ionizing radiation or carboplatin treatment restored ionizing radiation and platinum sensitivity in Wwox-deficient murine cells, as in human cells, supporting the use of murine in vitro and in vivo models in preclinical cancer treatment research.
- Published
- 2022
28. Abstract A012: Repurposing of the macrolide antibiotic clarithromycin for the prevention of lung cancer
- Author
-
Seyed Javad Moghaddam, Tabish Hussain, Melody Zarghooni, Walter V. Velasco, Linda Phan, Michelle I. Savage, Jennifer T. Fox, Shizuko Sei, Powel H. Brown, and C. Marcelo Aldaz
- Subjects
Cancer Research ,Oncology - Abstract
Lung adenocarcinoma (LUAD), particularly K-ras mutant LUAD, is a leading cause of cancer mortality. Therefore, strategies to prevent K-ras-mutant LUAD in its earliest stages in high-risk individuals (e.g., smokers) are urgently needed to reduce the public burden of this fatal disease. We and others have shown that K-ras driven tumorigenesis in lung is intimately linked to chronic inflammation and ultimately, tumor cells immune-escape. The antibiotic clarithromycin (CAM) was identified as one of the most promising candidates for repurposing with demonstrated immunomodulatory and anticancer properties. CAM is widely used and belongs to the macrolide class of antibiotics which are among the safest broad spectrum antimicrobials available. Abundant preclinical and clinical evidence exists demonstrating the in vitro and in vivo anticancer effects of CAM. It has been shown that macrolide antibiotics exert suppression of inflammation without overt immunosuppressive effects mostly through the inhibition of proinflammatory cytokines in vitro and in vivo. In these studies, we tested the lung cancer prevention efficacy of CAM using a Kras mutant lung cancer model. In the CCSPCre; LSL-Kras- G12 D (CC-LR) model, activation of the KrasG12 D mutation takes place in club cells by means of removal of the lox-stop-lox genomic sequence via expression of Cre recombinase under the control of the CCSP promoter. This model is excellent for reproducing the various premalignant to malignant progression steps in the lung. CC-LR mice of both genders were randomly enrolled to four experimental arms comparing three CAM doses: 10mg/kg/day, 50mg/kg/day, and 100mg/kg/day, to vehicle control (H2O). Treatment was administered by oral gavage, 5 times per week for 10 wks., starting at 4 wks. of age. At 14 wks. of age mice were euthanized, lung surface tumors were counted, bronchial lavage fluid, as well as lung samples, were obtained for histological, immunohistochemical (IHC) and qRT-PCR analyses. Clarithromycin treatment led to significant lung cancer prevention efficacy, as determined by lung surface tumor counts. A clear dose response with CAM was observed with a mean lung surface tumor count of 30.2 tumors per mouse for vehicle (n=12 mice), 23.5 for CAM 10mg/kg (n=8), 18.8 for CAM 50mg/kg (n=8) and 13.5 tumors per mouse for the CAM 100mg/kg treatment group (n=12, p=0.0014). A significant decrease in the incidence and multiplicity of premalignant and malignant lesions was also observed in histological analyses. For profiling of the lung immune microenvironment, we analyzed by qRT-PCR the expression of 18 genes identifying various cytokines, cell surface markers and proteins characteristic of specific activation states on the various cell subtypes in the tumor microenvironment. We found significant reduction in the expression of pro-inflammatory cytokines, IL-6, TNF, and IL-1β that are known to have a pro-tumor function in lung tumorigenesis, shown previously by us and other groups. We also found a reduction in the expression of myeloid cell-specific immunosuppressive markers, Arg1 and Fizz 1, that could be due to the reduction in the Gr1+ myeloid cell population, which was detected by means of IHC analyses on the same samples, or due to reprograming of pro-tumor M2 type macrophages toward an anti-tumor M1 phenotype. We also see a trend toward reduction of IL-17 cytokine, which we have previously shown to have an essential role in the promotion of K-ras mutant lung tumors. This was associated with an increase, although not significant, in the expression of anti-tumor Th1-specific transcription factor. However, we see an increase in the expression of PD-1, which could be due to the potential induction of an exhausted T cell phenotype. Taken together, we see a reprograming of the lung microenvironment from a tumor-promoting immunosuppressive phenotype toward an anti-tumor phenotype. This work was supported by NCI PREVENT TORFP75N91019F00131 (TORFP 2019 E-05). Citation Format: Seyed Javad Moghaddam, Tabish Hussain, Melody Zarghooni, Walter V. Velasco, Linda Phan, Michelle I. Savage, Jennifer T. Fox, Shizuko Sei, Powel H. Brown, C. Marcelo Aldaz. Repurposing of the macrolide antibiotic clarithromycin for the prevention of lung cancer [abstract]. In: Proceedings of the Second Biennial NCI Meeting: Translational Advances in Cancer Prevention Agent Development (TACPAD); 2022 Sep 7-9. Philadelphia (PA): AACR; Can Prev Res 2022;15(12 Suppl_2): Abstract nr A012.
- Published
- 2022
- Full Text
- View/download PDF
29. Wwox deletion leads to reduced GABA-ergic inhibitory interneuron numbers and activation of microglia and astrocytes in mouse hippocampus
- Author
-
Martín Carlos Abba, Bharathi Hattiangady, Jaeho Lee, Hyunsuk Kil, Jianjun Shen, Ashok K. Shetty, Tabish Hussain, Maheedhar Kodali, Yoko Takata, Bing Shuai, Sahithi Attaluri, and C. Marcelo Aldaz
- Subjects
Male ,0301 basic medicine ,GABA-ergic interneurons ,Glutamate decarboxylase ,Astrogliosis ,Hippocampal formation ,Hippocampus ,purl.org/becyt/ford/1 [https] ,0302 clinical medicine ,Neural Stem Cells ,ASTROGLIOSIS ,Gliosis ,GABAergic Neurons ,EPILEPSY ,Mice, Knockout ,Microglia ,WWOX ,Bioquímica y Biología Molecular ,Neural stem cell ,Cell biology ,medicine.anatomical_structure ,WW Domain-Containing Oxidoreductase ,Neurology ,Encephalitis ,Female ,MICROGLIOSIS ,Inflammation Mediators ,CIENCIAS NATURALES Y EXACTAS ,Medicina ,Wwox ,Biology ,Article ,lcsh:RC321-571 ,Ciencias Biológicas ,03 medical and health sciences ,Interneurons ,medicine ,Animals ,purl.org/becyt/ford/1.6 [https] ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,Neuroinflammation ,Epilepsy ,GABA-ERGIC INTERNEURONS ,medicine.disease ,Microgliosis ,030104 developmental biology ,nervous system ,Astrocytes ,HIPPOCAMPUS ,biology.protein ,Transcriptome ,030217 neurology & neurosurgery ,Parvalbumin - Abstract
The association of WW domain-containing oxidoreductase WWOX gene loss of function with central nervous system (CNS) related pathologies is well documented. These include spinocerebellar ataxia, epilepsy and mental retardation (SCAR12, OMIM: 614322) and early infantile epileptic encephalopathy (EIEE28, OMIM: 616211) syndromes. However, there is complete lack of understanding of the pathophysiological mechanisms at play. In this study, using a Wwox knockout (Wwox KO) mouse model (2 weeks old, both sexes) and stereological studies we observe that Wwox deletion leads to a significant reduction in the number of hippocampal GABA-ergic (γ-aminobutyric acid) interneurons. Wwox KO mice displayed significantly reduced numbers of calcium-binding protein parvalbumin (PV) and neuropeptide Y (NPY) expressing interneurons in different subfields of the hippocampus in comparison to Wwox wild-type (WT) mice. We also detected decreased levels of Glutamic Acid Decarboxylase protein isoforms GAD65/67 expression in Wwox null hippocampi suggesting lower levels of GABA synthesis. In addition, Wwox deficiency was associated with signs of neuroinflammation such as evidence of activated microglia, astrogliosis, and overexpression of inflammatory cytokines Tnf-a and Il6. We also performed comparative transcriptome-wide expression analyses of neural stem cells grown as neurospheres from hippocampi of Wwox KO and WT mice thus identifying 283 genes significantly dysregulated in their expression. Functional annotation of transcriptome profiling differences identified ‘neurological disease’ and ‘CNS development related functions’ to be significantly enriched. Several epilepsy-related genes were found differentially expressed in Wwox KO neurospheres. This study provides the first genotype-phenotype observations as well as potential mechanistic clues associated with Wwox loss of function in the brain., Facultad de Ciencias Médicas, Centro de Investigaciones Inmunológicas Básicas y Aplicadas
- Published
- 2019
- Full Text
- View/download PDF
30. MALINC1 an Immune-Related Long Non-Coding RNA Associated with Early-Stage Breast Cancer Progression
- Author
-
María Laura Fabre, Romina Canzoneri, Agustina Gurruchaga, Jaeho Lee, Pradeep Tatineni, Hyunsuk Kil, Ezequiel Lacunza, C. Marcelo Aldaz, and Martín Carlos Abba
- Subjects
Cancer Research ,Oncology ,skin and connective tissue diseases ,MALINC1 ,lncRNA ,breast cancer ,DCIS - Abstract
Long non-coding RNAs are increasingly being recognized as cancer biomarkers in various malignancies, acting as either tumor suppressors or oncogenes. The long non-coding MALINC1 intergenic RNA was identified as significantly upregulated in breast ductal carcinoma in situ. The aim of this study was to characterize MALINC1 expression, localization, and phenotypic and molecular effects in non-invasive and invasive breast cancer cells. We determined that MALINC1 is an estrogen–estrogen receptor-modulated lncRNA enriched in the cytoplasmic fraction of luminal A/B breast cancer cells that is associated with worse overall survival in patients with primary invasive breast carcinomas. Transcriptomic studies in normal and DCIS cells identified the main signaling pathways modulated by MALINC1, which mainly involve bioprocesses related to innate and adaptive immune responses, extracellular matrix remodeling, cell adhesion, and activation of AP-1 signaling pathway. We determined that MALINC1 induces premalignant phenotypic changes by increasing cell migration in normal breast cells. Moreover, high MALINC1 expression in invasive carcinomas was associated with a pro-tumorigenic immune environment and a favorable predicted response to immunotherapy both in luminal and basal-like subtypes compared with low-MALINC1-expression tumors. We conclude that MALINC1 behaves as an oncogenic and immune-related lncRNA involved with early-stage breast cancer progression.
- Published
- 2022
- Full Text
- View/download PDF
31. Genomic Alterations during the
- Author
-
Anne, Trinh, Carlos R, Gil Del Alcazar, Sachet A, Shukla, Koei, Chin, Young Hwan, Chang, Guillaume, Thibault, Jennifer, Eng, Bojana, Jovanović, C Marcelo, Aldaz, So Yeon, Park, Joon, Jeong, Catherine, Wu, Joe, Gray, and Kornelia, Polyak
- Subjects
body regions ,Carcinoma, Intraductal, Noninfiltrating ,Immune System ,Carcinoma, Ductal, Breast ,Humans ,Breast Neoplasms ,Female ,Genomics ,skin and connective tissue diseases ,neoplasms ,Article - Abstract
The drivers of ductal carcinoma in situ (DCIS) to invasive ductal carcinoma (IDC) transition are poorly understood. Here, we conducted an integrated genomic, transcriptomic, and whole-slide image analysis to evaluate changes in copy number profiles, mutational profiles, expression, neoantigen load and topology in 6 cases of matched pure DCIS and recurrent IDC. We demonstrate through combined copy number and mutational analysis that recurrent IDC can be genetically related to its pure DCIS despite long latency periods and therapeutic interventions. Immune “hot” and “cold” tumors can arise as early as DCIS and are subtype-specific. Topologic analysis showed a similar degree of pan-leukocyte-tumor mixing in both DCIS and IDC but differ when assessing specific immune subpopulations such as CD4 T-cells and CD68 macrophages. Tumor-specific copy number aberrations in MHC-I presentation machinery and losses in 3p,4q, and 5p are associated with differences in immune signaling in estrogen receptor (ER) negative IDC. Common oncogenic hotspot mutations in genes including TP53 and PIK3CA are predicted to be neoantigens yet are paradoxically conserved during the DCIS-to-IDC transition, and are associated with differences in immune signaling. We highlight both tumor and immune-specific changes in the transition of pure DCIS to IDC, including genetic changes in tumor cells that may have a role in modulating immune function and assist in immune escape, driving the transition to IDC.
- Published
- 2020
32. Cigarette Smoke and Nicotine-Containing E-cigarette Vapor Downregulate Lung WWOX Expression Which is Associated with Increased Severity of Murine ARDS
- Author
-
Roberto Machado, Viswanathan Natarajan, Lorraine B. Ware, Zhenguo Zeng, Raheel Khan, C. Marcelo Aldaz, Jeffrey R. Jacobson, Alexander Moshensky, Sunit Singla, Weiguo Chen, Steven M. Dudek, and Laura Crotty-Alexander
- Subjects
WWOX ,Lung ,medicine.diagnostic_test ,business.industry ,Chromosomal fragile site ,Wild type ,Lung injury ,Flow cytometry ,Andrology ,medicine.anatomical_structure ,Downregulation and upregulation ,medicine ,business ,Barrier function - Abstract
RATIONALEA history of chronic cigarette smoking is known to increase risk for ARDS, but the corresponding risks associated with chronic e-cigarette use are largely unknown. The chromosomal fragile site gene, WWOX, is highly susceptible to genotoxic stress from environmental exposures, and thus an interesting candidate gene for the study of exposure-related lung disease.METHODS AND RESULTSLungs harvested from current versus former/never smokers exhibited a 47% decrease in WWOX mRNA levels. Exposure to nicotine-containing e-cigarette vapor resulted in an average 57% decrease in WWOX mRNA levels relative to vehicle treated controls. In separate studies, endothelial (EC)-specific WWOX KO versus wild type mice were examined under ARDS-producing conditions. EC WWOX KO mice exhibited significantly greater levels of vascular leak and histologic lung injury. ECs were isolated from digested lungs of untreated EC WWOX KO mice using sorting by flow cytometry for CD31+CD45- cells. These were grown in culture, confirmed to be WWOX-deficient by RT-PCR and Western blotting, and analyzed by electric cell impedance sensing (ECIS) as well as a FITC dextran transwell assay for their barrier properties during MRSA or LPS exposure. WWOX KO ECs demonstrated significantly greater declines in barrier function relative to wild type cells during either MRSA or LPS treatment as measured by both ECIS and the transwell assay.CONCLUSIONThe increased risk for ARDS observed in chronic smokers may be mechanistically linked, at least in part, to lung WWOX downregulation, and this phenomenon may also manifest in the near future in chronic users of e-cigarettes.
- Published
- 2020
- Full Text
- View/download PDF
33. WWOX Loss of Function in Neurodevelopmental and Neurodegenerative Disorders
- Author
-
Tabish Hussain and C. Marcelo Aldaz
- Subjects
Central Nervous System ,WWOX ,Cerebellum ,Cell type ,autism ,Review ,Biology ,multiple sclerosis ,Catalysis ,lcsh:Chemistry ,Inorganic Chemistry ,Mice ,spinocerebellar ataxia ,Loss of Function Mutation ,medicine ,ADHD ,Animals ,Humans ,Genetic Predisposition to Disease ,Physical and Theoretical Chemistry ,lcsh:QH301-705.5 ,Molecular Biology ,Germ-Line Mutation ,Spectroscopy ,Loss function ,Cerebellar ataxia ,Tumor Suppressor Proteins ,Organic Chemistry ,Neurodegeneration ,neurodegeneration ,Neurodegenerative Diseases ,General Medicine ,WOREE ,medicine.disease ,Computer Science Applications ,epileptic encephalopathy ,medicine.anatomical_structure ,WW Domain-Containing Oxidoreductase ,lcsh:Biology (General) ,lcsh:QD1-999 ,Neurodevelopmental Disorders ,intellectual disability ,Cerebellar cortex ,Spinocerebellar ataxia ,medicine.symptom ,Alzheimer’s disease ,Neuroscience - Abstract
The WWOX gene was initially discovered as a putative tumor suppressor. More recently, its association with multiple central nervous system (CNS) pathologies has been recognized. WWOX biallelic germline pathogenic variants have been implicated in spinocerebellar ataxia type 12 (SCAR12; MIM:614322) and in early infantile epileptic encephalopathy (EIEE28; MIM:616211). WWOX germline copy number variants have also been associated with autism spectrum disorder (ASD). All identified germline genomic variants lead to partial or complete loss of WWOX function. Importantly, large-scale genome-wide association studies have also identified WWOX as a risk gene for common neurodegenerative conditions such as Alzheimer’s disease (AD) and multiple sclerosis (MS). Thus, the spectrum of CNS disorders associated with WWOX is broad and heterogeneous, and there is little understanding of potential mechanisms at play. Exploration of gene expression databases indicates that WWOX expression is comparatively higher in the human cerebellar cortex than in other CNS structures. However, RNA in-situ hybridization data from the Allen Mouse Brain Atlas show that specific regions of the basolateral amygdala (BLA), the medial entorhinal cortex (EC), and deep layers of the isocortex can be singled out as brain regions with specific higher levels of Wwox expression. These observations are in close agreement with single-cell RNA-seq data which indicate that neurons from the medial entorhinal cortex, Layer 5 from the frontal cortex as well as GABAergic basket cells and granule cells from cerebellar cortex are the specific neuronal subtypes that display the highest Wwox expression levels. Importantly, the brain regions and cell types in which WWOX is most abundantly expressed, such as the EC and BLA, are intimately linked to pathologies and syndromic conditions in turn associated with this gene, such as epilepsy, intellectual disability, ASD, and AD. Higher Wwox expression in interneurons and granule cells from cerebellum points to a direct link to the described cerebellar ataxia in cases of WWOX loss of function. We now know that total or partial impairment of WWOX function results in a wide and heterogeneous variety of neurodegenerative conditions for which the specific molecular mechanisms remain to be deciphered. Nevertheless, these observations indicate an important functional role for WWOX in normal development and function of the CNS. Evidence also indicates that disruption of WWOX expression at the gene or protein level in CNS has significant deleterious consequences.
- Published
- 2020
- Full Text
- View/download PDF
34. Abstract PO-059: The genomic landscape of the in situ to invasive ductal breast carcinoma transition shaped by the immune system
- Author
-
Bojana Jovanovic, Jennifer Eng, Joe W. Gray, Anne Trinh, Young Hwan Chen, Kornelia Polyak, Catherine J. Wu, Joon Jong, Sachet A. Shukla, C. Marcelo Aldaz, Guillaume Thibault, So Yeon Park, Koei Chin, and Carlos R. Gil Del Alcazar
- Subjects
In situ ,Cancer Research ,Transition (genetics) ,Cancer ,Context (language use) ,Biology ,Ductal carcinoma ,medicine.disease ,body regions ,Immune system ,Oncology ,Cancer research ,medicine ,Invasive Ductal Breast Carcinoma ,skin and connective tissue diseases ,neoplasms ,Exome - Abstract
Background: The transition from ductal carcinoma in situ (DCIS) to invasive ductal carcinoma (IDC) is an evolutionary bottleneck where progression occurs only in 30% of patients. Whilst the genetic drivers of this transition remain poorly understood, we have previously shown that immune escape is a key event. In this study, we profile the evolutionary trajectory of matched pure DCIS and IDC in the context of the immune microenvironment. Methods: We have evaluated changes in copy number profiles, mutational profiles, expression and neoantigen load in 6 cases of matched pure DCIS and IDC using exome and RNA sequencing. We have integrated this information with topologic assessment of H&E images and cyclic immunofluorescence. Results: We provide evidence for an evolutionary bottleneck during DCIS to IDC in matched patient samples, showing that copy number aberrations are early events, but low overlap in mutational profiles. Variation in immune composition and spatial orientation can arise as early as in DCIS and are subtype specific. Tumor-specific copy number changes including loss of MHC-I presentation machinery or changes at cytokine rich loci specifically in ER− tumors could contribute to a more immunosuppressive environment in IDC. Oncogenic hotspot mutations can present as neoantigens yet are paradoxically conserved during the DCIS-to-IDC transition. We suggest these mutations have a secondary immune-modulatory function or may be present in normal tissue, escaping immune surveillance as early as in DCIS. Conclusions: We show both genomic and microenvironmental differences in matched pure DCIS and recurrent IDC, highlighting that progression is shaped by both tumor and immune system at this evolutionary bottleneck. Citation Format: Anne Trinh, Carlos R. Gil Del Alcazar, Sachet A. Shukla, Koei Chin, Young Hwan Chen, Guillaume Thibault, Jennifer Eng, Bojana Jovanovic, C. Marcelo Aldaz, So Yeon Park, Joon Jong, Catherine Wu, Joe Gray, Kornelia Polyak. The genomic landscape of the in situ to invasive ductal breast carcinoma transition shaped by the immune system [abstract]. In: Proceedings of the AACR Virtual Special Conference on Tumor Heterogeneity: From Single Cells to Clinical Impact; 2020 Sep 17-18. Philadelphia (PA): AACR; Cancer Res 2020;80(21 Suppl):Abstract nr PO-059.
- Published
- 2020
- Full Text
- View/download PDF
35. Delineating WWOX Protein Interactome by Tandem Affinity Purification-Mass Spectrometry: Identification of Top Interactors and Key Metabolic Pathways Involved
- Author
-
Martín Carlos Abba, C. Marcelo Aldaz, Junjie Chen, Tabish Hussain, and Jaeho Lee
- Subjects
Bioquímica ,0301 basic medicine ,WWOX ,Tap-ms ,Cancer Research ,Interactome ,Endosome ,Computational biology ,lcsh:RC254-282 ,DNA-binding protein ,purl.org/becyt/ford/1 [https] ,Ciencias Biológicas ,03 medical and health sciences ,symbols.namesake ,0302 clinical medicine ,Ciencias Naturales ,purl.org/becyt/ford/1.6 [https] ,Ciencias Exactas ,Original Research ,Tandem affinity purification ,Protein transport ,Chemistry ,WW domains ,Bioquímica y Biología Molecular ,Golgi apparatus ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,3. Good health ,Transport protein ,Metabolic pathway ,030104 developmental biology ,Oncology ,Metabolic pathways ,030220 oncology & carcinogenesis ,symbols ,CIENCIAS NATURALES Y EXACTAS - Abstract
It has become clear from multiple studies that WWOX (WW domain-containing oxidoreductase) operates as a “non-classical” tumor suppressor of significant relevance in cancer progression. Additionally, WWOX has been recognized for its role in a much wider array of human pathologies including metabolic conditions and central nervous system related syndromes. A myriad of putative functional roles has been attributed to WWOX mostly through the identification of various binding proteins. However, the reality is that much remains to be learned on the key relevant functions of WWOX in the normal cell. Here we employed a Tandem Affinity Purification-Mass Spectrometry (TAP-MS) approach in order to better define direct WWOX protein interactors and by extension interaction with multiprotein complexes under physiological conditions on a proteomic scale. This work led to the identification of both well-known, but more importantly novel high confidence WWOX interactors, suggesting the involvement of WWOX in specific biological and molecular processes while delineating a comprehensive portrait of WWOX protein interactome. Of particular relevance is WWOX interaction with key proteins from the endoplasmic reticulum (ER), Golgi, late endosomes, protein transport, and lysosomes networks such as SEC23IP, SCAMP3, and VOPP1. These binding partners harbor specific PPXY motifs which directly interact with the amino-terminal WW1 domain of WWOX. Pathway analysis of WWOX interactors identified a significant enrichment of metabolic pathways associated with proteins, carbohydrates, and lipids breakdown. Thus, suggesting that WWOX likely plays relevant roles in glycolysis, fatty acid degradation and other pathways that converge primarily in Acetyl-CoA generation, a fundamental molecule not only as the entry point to the tricarboxylic acid (TCA) cycle for energy production, but also as the key building block for de novo synthesis of lipids and amino acids. Our results provide a significant lead on subsets of protein partners and enzymatic complexes with which full-length WWOX protein interacts with in order to carry out its metabolic and other biological functions while also becoming a valuable resource for further mechanistic studies., Facultad de Ciencias Médicas, Centro de Investigaciones Inmunológicas Básicas y Aplicadas
- Published
- 2018
- Full Text
- View/download PDF
36. Abstracts from the 3rd Conference on Aneuploidy and Cancer: Clinical and Experimental Aspects
- Author
-
J. Xu, N. Page, Ruediger Hehlmann, Marthe Løvf, Victor Guryev, Sarah Grasedieck, J. Stein, Paola Cavaliere, Diana C.J. Spierings, S. Bhattacharya, Caroline Jansson, Allan Bradley, Andrew L. Trinh, Zuzana Storchova, Mat Bloomfield, G. Stein, Tristan V. de Jong, Nicolaas C. Baudoin, Jonas M. SveeStrømme, H. Ding, J. Vecerova, Xue Gong, Christina Raftopoulou, Nancy Halsema, Nataliya Huleyuk, Rolf Inge Skotheim, Jordi Camps, Mathew Bloomfield, Christof Börgermann, Anita Sveen, Steven Horne, Bjarne Johannessen, Julian Swoboda, Vladimir P. Baklaushev, A. Fritz, Anders Valind, N. Donnelly, Henry H.Q. Heng, Aracelli Acevedo-Colina, Peter H. Duesberg, A. A. Stepanenko, Rainer Engers, C. Kruse, Mark D. Vincent, Yi-Hong Zhou, Lars Bullinger, Sunyoung Hwang, Fani-Marlen Roumelioti, Vladimir P. Chekhonin, R. Berezney, Martha R. Stampfer, Batoul Y. Abdallah, Guo Liu, C. Tye, David Porubsky, Jenny Karlsson, James C. Garbe, Verena Passerini, Oksana A. Kovaleva, Xinhe Huang, Andrej Schevchenko, N. Sehgal, Frank G. Rücker, Milena Dürrbaum, Karina Hoekstra-Wakker, Daniela Cimini, David Gisselsson, Ellen K. Silbergeld, Stanislav Avdieiev, Bianca Habermann, S V Andreieva, Anke van den Berg, Hans Tobias Gustafsson, Daniele Mandrioli, Jonathan R. Pollack, A. Voskanyan, Floris Foijer, Josef Dietz, Thomas Liehr, Stefan Biesterfeld, Athel Cornish-Bowden, Melissa J. Perry, Fiorella Belpoggi, Christine J. Ye, B. Stojkovicz, Lukas Vrba, Peter M. Lansdorp, Maria Colomé Tatché, Ciara O’Sullivan, Ragnhild A. Lothe, Mirjam E. Belderbos, Hinke G. Kazemier, Eveline S. J. M. de Bont, Kateryna Korets, Alfred Böcking, David Rasnick, Joshua M. Nicholson, Michelle A. Digman, Isabel Quintanilla, Sen Zhao, Yegor S. Vassetzky, Jennifer A. Marshall Graves, Bernard W. Futscher, Mark A. LaBarge, Aaron Taudt, Leonid Berynskyy, Maria Chiourea, Robert C. Dickson, Dmytro Mykytenko, Andreas M. Hoff, Noah Dephoure, C. Marcelo Aldaz, Bjorn Bakker, Christian Klose, Sarantis Gagos, Alice Fabarius, Eduardo M. Torres, and Kimberly Soto
- Subjects
Genetics ,Philosophy ,Biochemistry (medical) ,Molecular Medicine ,Theology ,Molecular Biology ,Biochemistry ,Genetics (clinical) ,3. Good health - Abstract
Author(s): Cornish-Bowden, Athel; Cornish-Bowden, Athel; Rasnick, David; Heng, Henry H; Horne, Steven; Abdallah, Batoul; Liu, Guo; Ye, Christine J; Bloomfield, Mathew; Vincent, Mark D; Aldaz, C Marcelo; Karlsson, Jenny; Valind, Anders; Jansson, Caroline; Gisselsson, David; Graves, Jennifer A Marshall; Stepanenko, Aleksei A; Andreieva, Svitlana V; Korets, Kateryna V; Mykytenko, Dmytro O; Huleyuk, Nataliya L; Baklaushev, Vladimir P; Kovaleva, Oksana A; Chekhonin, Vladimir P; Vassetzky, Yegor S; Avdieiev, Stanislav S; Bakker, Bjorn; Taudt, Aaron S; Belderbos, Mirjam E; Porubsky, David; Spierings, Diana CJ; de Jong, Tristan V; Halsema, Nancy; Kazemier, Hinke G; Hoekstra-Wakker, Karina; Bradley, Allan; de Bont, Eveline SJM; van den Berg, Anke; Guryev, Victor; Lansdorp, Peter M; Tatche, Maria Colome; Foijer, Floris; Liehr, Thomas; Baudoin, Nicolaas C; Nicholson, Joshua M; Soto, Kimberly; Quintanilla, Isabel; Camps, Jordi; Cimini, Daniela; Durrbaum, M; Donnelly, N; Passerini, V; Kruse, C; Habermann, B; Storchova, Z; Mandrioli, Daniele; Belpoggi, Fiorella; Silbergeld, Ellen K; Perry, Melissa J; Skotheim, Rolf I; Lovf, Marthe; Johannessen, Bjarne; Hoff, Andreas M; Zhao, Sen; SveeStromme, Jonas M; Sveen, Anita; Lothe, Ragnhild A; Hehlmann, R; Voskanyan, A; Fabarius, A; Bocking, Alfred; Biesterfeld, Stefan; Berynskyy, Leonid; Borgermann, Christof; Engers, Rainer; Dietz, Josef; Fritz, A; Sehgal, N; Vecerova, J; Stojkovicz, B; Ding, H; Page, N; Tye, C; Bhattacharya, S; Xu, J
- Published
- 2017
- Full Text
- View/download PDF
37. The WWOX Gene Modulates High-Density Lipoprotein and Lipid Metabolism
- Author
-
Mohammad Abu Odeh, M.V. Prasad Linga Reddy, Rami I. Aqeilan, Daphna Weissglas-Volkov, Elina Nikkola, Muhannad Abu-Remaileh, Zaidoun Salah, Isabelle Ruel, Päivi Pajukanta, Hyunsuk Kil, Jacques Genest, Carlo M. Croce, C. Marcelo Aldaz, Zuhier Awan, Jaeho Lee, Hong Y. Choi, Iulia Iatan, and Mete Civelek
- Subjects
WWOX ,Genetics ,medicine.medical_specialty ,Microarray ,Cholesterol ,Lipid metabolism ,Biology ,chemistry.chemical_compound ,High-density lipoprotein ,Endocrinology ,chemistry ,ANGPTL4 ,ABCA1 ,Internal medicine ,medicine ,biology.protein ,lipids (amino acids, peptides, and proteins) ,Cardiology and Cardiovascular Medicine ,Genetics (clinical) ,Lipoprotein - Abstract
Background— Low levels of high-density lipoprotein (HDL) cholesterol constitutes a major risk factor for atherosclerosis. Recent studies from our group reported a genetic association between the WW domain-containing oxidoreductase ( WWOX ) gene and HDL cholesterol levels. Here, through next-generation resequencing, in vivo functional studies and gene microarray analyses, we investigated the role of WWOX in HDL and lipid metabolism. Methods and Results— Using next-generation resequencing of the WWOX region, we first identified 8 variants significantly associated and perfectly segregating with the low-HDL trait in 2 multigenerational French Canadian dyslipidemic families. To understand in vivo functions of WWOX, we used liver-specific Wwox hep−/− and total Wwox −/− mice models, where we found decreased ApoA-I and Abca1 levels in hepatic tissues. Analyses of lipoprotein profiles in Wwox −/− , but not Wwox hep−/− littermates, also showed marked reductions in serum HDL cholesterol concentrations, concordant with the low-HDL findings observed in families. We next obtained evidence of a sex-specific effect in female Wwox hep−/− mice, where microarray analyses revealed an increase in plasma triglycerides and altered lipid metabolic pathways. We further identified a significant reduction in ApoA-I and Lpl and an upregulation in Fas , Angptl4 , and Lipg , suggesting that the effects of Wwox involve multiple pathways, including cholesterol homeostasis, ApoA-I/ABCA1 pathway, and fatty acid biosynthesis/triglyceride metabolism. Conclusions— Our data indicate that WWOX disruption alters HDL and lipoprotein metabolism through several mechanisms and may account for the low-HDL phenotype observed in families expressing the WWOX variants. These findings thus describe a novel gene involved in cellular lipid homeostasis, which effects may impact atherosclerotic disease development.
- Published
- 2014
- Full Text
- View/download PDF
38. Identification of Modulated Genes by Three Classes of Chemopreventive Agents at Preneoplastic Stages in a p53-Null Mouse Mammary Tumor Model
- Author
-
Jamal Hill, Carla C. Levy, Daniel Medina, Frances S. Kittrell, Reid P. Bissonnette, C. Marcelo Aldaz, Powel H. Brown, Sally Gaddis, Martín Carlos Abba, and Yuhui Hu
- Subjects
Cancer Research ,Tetrahydronaphthalenes ,EGR1 ,Biology ,Article ,Malignant transformation ,Transcriptome ,Mice ,Mammary Glands, Animal ,Gene expression ,Biomarkers, Tumor ,medicine ,Animals ,Anticarcinogenic Agents ,Cyclooxygenase Inhibitors ,Oligonucleotide Array Sequence Analysis ,Mice, Knockout ,Bexarotene ,Mice, Inbred BALB C ,Sulfonamides ,Mammary tumor ,Gene Expression Profiling ,Mammary Neoplasms, Experimental ,Tumor Protein, Translationally-Controlled 1 ,Gefitinib ,FOSL2 ,Molecular biology ,ErbB Receptors ,Gene expression profiling ,Disease Models, Animal ,Oncology ,Celecoxib ,Quinazolines ,Cancer research ,Pyrazoles ,Female ,Tumor Suppressor Protein p53 ,Precancerous Conditions ,medicine.drug - Abstract
Genetically engineered mouse cancer models are among the most useful tools for testing the in vivo effectiveness of the various chemopreventive approaches. The p53-null mouse model of mammary carcinogenesis was previously characterized by us at the cellular, molecular, and pathologic levels. In a companion article, Medina et al. analyzed the efficacy of bexarotene, gefitinib, and celecoxib as chemopreventive agents in the same model. Here we report the global gene expression effects on mammary epithelium of such compounds, analyzing the data in light of their effectiveness as chemopreventive agents. SAGE was used to profile the transcriptome of p53-null mammary epithelium obtained from mice treated with each compound versus controls. This information was also compared with SAGE data from p53-null mouse mammary tumors. Gene expression changes induced by the chemopreventive treatments revealed a common core of 87 affected genes across treatments (P < 0.05). The effective compounds, bexarotene and gefitinib, may exert their chemopreventive activity, at least in part, by affecting a set of 34 genes related to specific cellular pathways. The gene expression signature revealed various genes previously described to be associated with breast cancer, such as the activator protein-1 complex member Fos-like antigen 2 (Fosl2), early growth response 1 (Egr1), gelsolin (Gsn), and tumor protein translationally controlled 1 (Tpt1), among others. The concerted modulation of many of these transcripts before malignant transformation seems to be conducive to predominantly decrease cell proliferation. This study has revealed candidate key pathways that can be experimentally tested in the same model system and may constitute novel targets for future translational research.
- Published
- 2009
- Full Text
- View/download PDF
39. WWOXhypomorphic mice display a higher incidence of B-cell lymphomas and develop testicular atrophy
- Author
-
Jan Parker-Thornburg, Maria I. Nunez, Mark T. Bedford, John H. Ludes-Meyers, Hyunsuk Kil, C. Marcelo Aldaz, and Claudio J. Conti
- Subjects
Male ,WWOX ,Cancer Research ,Lymphoma, B-Cell ,Tumor suppressor gene ,Mice, Transgenic ,Biology ,medicine.disease_cause ,Article ,law.invention ,Mice ,law ,Cell Line, Tumor ,Testis ,Gene expression ,Genetics ,medicine ,Animals ,B cell ,Mice, Knockout ,Chromosomal fragile site ,Embryo, Mammalian ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,WW Domain-Containing Oxidoreductase ,Models, Animal ,Knockout mouse ,Cancer research ,Suppressor ,Female ,Atrophy ,Oxidoreductases ,Carcinogenesis - Abstract
WWOX is a putative tumor suppressor gene encoded within common chromosomal fragile site region FRA16D, in chromosome band 16q23. Multiple studies have demonstrated that WWOX expression is often reduced or lost in various tumor types. WWOX tumor suppressor activity was suggested by re-expressing WWOX in breast, ovarian, and lung tumor cell lines leading to tumor growth inhibition in vivo. To determine whether loss of Wwox gene expression has a role in tumorigenesis, we generated a mouse strain containing a Wwox gene mutated by a gene-trap vector. Homozygous Wwox gene-trap mice (Wwoxgt/gt) had no detectable Wwox protein in most tissues examined, although, a low level could be detected in a minority of tissues. Because of these observations, we concluded that these mice are Wwox hypomorphs. Remarkably, Wwox hypomorphic mice are viable in contrast to the recently reported postnatal lethality of Wwox knockout mice. Testes from Wwoxgt/gt males had high numbers of atrophic seminiferous tubules and reduced fertility when compared with wild-type counterparts. We observed that the Wwoxgt/gt mice had a significantly shorter lifespan, and female hypomorphs had a higher incidence of spontaneous B-cell lymphomas. In conclusion, we describe a novel Wwox hypomorphic mouse model that overcomes postnatal lethality that was recently observed in Wwox knockout mice. Therefore, tumorigenesis studies using this model more closely recapitulates the loss of WWOX expression observed in human cancers. Importantly, our observation that Wwox hypomorphs had an increased incidence of B-cell lymphomas supports a role of Wwox as a tumor suppressor. © 2007 Wiley-Liss, Inc.
- Published
- 2007
- Full Text
- View/download PDF
40. Breast Cancer Molecular Signatures as Determined by SAGE: Correlation with Lymph Node Status
- Author
-
Steve Horvath, Yuhui Hu, Tao Shi, Kathleen A. Hawkins, C. Marcelo Aldaz, Martín Carlos Abba, Hongxia Sun, Sally Gaddis, Aysegul Sahin, Jeffrey A. Drake, and Maria I. Nunez
- Subjects
Cancer Research ,Databases, Factual ,Transcription, Genetic ,Breast Neoplasms ,Biology ,Article ,Breast cancer ,Gene expression ,medicine ,Humans ,Serial analysis of gene expression ,Molecular Biology ,Lymph node ,DNA Primers ,Neoplasm Staging ,Oligonucleotide Array Sequence Analysis ,Regulation of gene expression ,Reverse Transcriptase Polymerase Chain Reaction ,medicine.disease ,Fold change ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,Oncology ,CSNK1D ,Lymphatic Metastasis ,Cancer research ,Female ,Lymph Nodes ,DNA microarray - Abstract
Global gene expression measured by DNA microarray platforms have been extensively used to classify breast carcinomas correlating with clinical characteristics, including outcome. We generated a breast cancer Serial Analysis of Gene Expression (SAGE) high-resolution database of ∼2.7 million tags to perform unsupervised statistical analyses to obtain the molecular classification of breast-invasive ductal carcinomas in correlation with clinicopathologic features. Unsupervised statistical analysis by means of a random forest approach identified two main clusters of breast carcinomas, which differed in their lymph node status (P = 0.01); this suggested that lymph node status leads to globally distinct expression profiles. A total of 245 (55 up-modulated and 190 down-modulated) transcripts were differentially expressed between lymph node (+) and lymph node (−) primary breast tumors (fold change, ≥2; P < 0.05). Various lymph node (+) up-modulated transcripts were validated in independent sets of human breast tumors by means of real-time reverse transcription-PCR (RT-PCR). We validated significant overexpression of transcripts for HOXC10 (P = 0.001), TPD52L1 (P = 0.007), ZFP36L1 (P = 0.011), PLINP1 (P = 0.013), DCTN3 (P = 0.025), DEK (P = 0.031), and CSNK1D (P = 0.04) in lymph node (+) breast carcinomas. Moreover, the DCTN3 (P = 0.022) and RHBDD2 (P = 0.002) transcripts were confirmed to be overexpressed in tumors that recurred within 6 years of follow-up by real-time RT-PCR. In addition, meta-analysis was used to compare SAGE data associated with lymph node (+) status with publicly available breast cancer DNA microarray data sets. We have generated evidence indicating that the pattern of gene expression in primary breast cancers at the time of surgical removal could discriminate those tumors with lymph node metastatic involvement using SAGE to identify specific transcripts that behave as predictors of recurrence as well. (Mol Cancer Res 2007;5(9):881–90)
- Published
- 2007
- Full Text
- View/download PDF
41. Epidermal hyperplasia and oral carcinoma in mice overexpressing the transcription factor ATF3 in basal epithelial cells
- Author
-
Stacey Arantes, C. Marcelo Aldaz, Mark J. McArthur, Michael C. MacLeod, Aijin Wang, and Claudio J. Conti
- Subjects
Cancer Research ,DNA, Complementary ,Time Factors ,Genotype ,Mice, Transgenic ,Biology ,Outer root sheath ,Plant Roots ,Mice ,Basal (phylogenetics) ,Carcinoma ,medicine ,Animals ,Humans ,Oral mucosa ,Molecular Biology ,Activating Transcription Factor 3 ,Epidermis (botany) ,Hyperplasia ,medicine.disease ,Hair follicle ,Gene Expression Regulation, Neoplastic ,Keratin 5 ,medicine.anatomical_structure ,Cancer research ,Keratin-5 ,Keratins ,Mouth Neoplasms ,Tumor Suppressor Protein p53 ,Transcription Factors - Abstract
ATF3 is a highly conserved eukaryotic transcription factor that is ubiquitously upregulated transcriptionally during cellular responses to a variety of stresses, in particular DNA damage. However, the role of ATF3 in the DNA damage response is unclear. Transgenic mice that overexpress human ATF3 in basal epithelial cells under the control of the bovine keratin 5 (K5) promoter were constructed and characterized for epidermal alterations. Strong, nuclear expression of the exogenous ATF3 protein was seen in basal cells of the epidermis, hair follicles, and oral mucosa. Hyperplastic changes in the K5-expressing, outer root sheath (ORS) cells of the hair follicle were observed in young mice, resulting in multiple layers of ORS cells in the mature follicle and large aberrantly shaped follicles. Mild hyperplasia of the interfollicular epidermis was also noted, increasing with age. However, no epidermal tumors were identified in BK5.ATF3 mice observed for 16 mo. At 16 mo of age, most transgenic mice exhibited multi-focal areas of hyperplasia and dysplasia in the oral mucosa, with cellular atypia and underlying acute inflammatory changes. Neoplastic lesions were also seen in the oral cavity of BK5.ATF3 mice, including oral squamous cell carcinoma (60% incidence) and basal cell tumors with follicular differentiation (70% incidence), but not in non-transgenic FVB/N littermates. Heterogeneous nuclear expression (or stabilization) of p53 protein was seen in some oral dysplasias, with a patchy distribution primarily in the least differentiated layers of the lesions. This represents the first indication that ATF3 may have oncogenic properties in epithelial cells.
- Published
- 2007
- Full Text
- View/download PDF
42. A Molecular Portrait of High-Grade Ductal Carcinoma In Situ
- Author
-
Sally Gaddis, Jaeho Lee, Yue Lu, Yoko Takata, Matias Butti, Marcos R. Estecio, Ting Gong, Yi Zhong, Aysegul A. Sahin, Ezequiel Lacunza, C. Marcelo Aldaz, Martín Carlos Abba, and Jianjun Shen
- Subjects
Antigens, Differentiation, T-Lymphocyte ,Cancer Research ,Pathology ,RNA, Untranslated ,T-Lymphocytes, Regulatory ,CDH1 ,purl.org/becyt/ford/1 [https] ,CTLA-4 Antigen ,Breast ,RNA, Neoplasm ,skin and connective tissue diseases ,Exome ,HOTAIR ,DNA, Neoplasm ,Bioquímica y Biología Molecular ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,Ductal Carcinoma in Situ ,Oncology ,Female ,CIENCIAS NATURALES Y EXACTAS ,medicine.medical_specialty ,DCIS ,Breast Neoplasms ,Biology ,Article ,Ciencias Biológicas ,Gene Expresssion ,Breast cancer ,Carcinoma Ductal ,Lymphocytes, Tumor-Infiltrating ,Breast Cancer ,Carcinoma ,medicine ,Humans ,Neoplasm Invasiveness ,RNA, Messenger ,purl.org/becyt/ford/1.6 [https] ,neoplasms ,Gene Expression Profiling ,Cancer ,Ductal carcinoma ,DNA Methylation ,medicine.disease ,Carcinoma, Intraductal, Noninfiltrating ,Tumor progression ,Ciencias Médicas ,Mutation ,Cancer research ,biology.protein ,Transcriptome ,Genes, Neoplasm - Abstract
Ductal carcinoma in situ (DCIS) is a noninvasive precursor lesion to invasive breast carcinoma. We still have no understanding on why only some DCIS lesions evolve to invasive cancer whereas others appear not to do so during the life span of the patient. Here, we performed full exome (tumor vs. matching normal), transcriptome, and methylome analysis of 30 pure high-grade DCIS (HG-DCIS) and 10 normal breast epithelial samples. Sixty-two percent of HG-DCIS cases displayed mutations affecting cancer driver genes or potential drivers. Mutations were observed affecting PIK3CA (21% of cases), TP53 (17%), GATA3 (7%), MLL3 (7%) and single cases of mutations affecting CDH1, MAP2K4, TBX3, NF1, ATM, and ARID1A. Significantly, 83% of lesions displayed numerous large chromosomal copy number alterations, suggesting they might precede selection of cancer driver mutations. Integrated pathway-based modeling analysis of RNA-seq data allowed us to identify two DCIS subgroups (DCIS-C1 and DCIS-C2) based on their tumor-intrinsic subtypes, proliferative, immune scores, and in the activity of specific signaling pathways. The more aggressive DCIS-C1 (highly proliferative, basal-like, or ERBB2 + ) displayed signatures characteristic of activated Treg cells (CD4 + /CD25 + /FOXP3 + ) and CTLA4 + /CD86 + complexes indicative of a tumor-associated immunosuppressive phenotype. Strikingly, all lesions showed evidence of TP53 pathway inactivation. Similarly, ncRNA and methylation profiles reproduce changes observed postinvasion. Among the most significant findings, we observed upregulation of lncRNA HOTAIR in DCIS-C1 lesions and hypermethylation of HOXA5 and SOX genes. We conclude that most HG-DCIS lesions, in spite of representing a preinvasive stage of tumor progression, displayed molecular profiles indistinguishable from invasive breast cancer., Centro de Investigaciones Inmunológicas Básicas y Aplicadas
- Published
- 2015
43. Quantitative high-throughput measurement of gene expression with sub-zeptomole sensitivity by capillary electrophoresis
- Author
-
K. Leslie Powell, Howard D. Thames, Nikki Liburd, Lea Spyres, Stacey Arantes, Michael C. MacLeod, Ella Bedford, Earl F. Walborg, Mahmoud Rouabhia, C. Marcelo Aldaz, David L. Mitchell, and Sally Gaddis
- Subjects
Keratinocytes ,Biophysics ,Gene Expression ,Bronchi ,Respiratory Mucosa ,Biology ,Sensitivity and Specificity ,Biochemistry ,Genome ,Cell Line ,Capillary electrophoresis ,Gene expression ,Humans ,RNA, Messenger ,Mammary Glands, Human ,Molecular Biology ,Gene ,Detection limit ,Messenger RNA ,Electrophoresis, Capillary ,Epithelial Cells ,Cell Biology ,Fibroblasts ,Genes, p53 ,Fluorescence ,Molecular biology ,Orders of magnitude (mass) ,Female - Abstract
Microarray technologies have provided the ability to monitor the expression of whole genomes rapidly. However, concerns persist with regard to quantitation and reproducibility, and the detection limits for individual genes in particular arrays are generally unknown. This article describes a semiautomated PCR-based technology, Q-RAGE, which rapidly provides measurements of mRNA abundance with extremely high sensitivity using fluorescent detection of specific products separated by capillary electrophoresis. A linear relationship between template concentration and fluorescent signal can be demonstrated down to template concentrations in the low aM region, corresponding to approximately 0.04 zmol (24 molecules) per reaction. The technique is shown to be quantitative over five orders of magnitude of template concentration, and average mRNA abundances of approximately 0.01 molecule per cell can be detected. A single predefined set of 320 primers provides 90–95% coverage of all eukaryotic genomes. Analysis of a set of 19 p53-regulated genes in untreated cultures of normal human epithelial cells, derived from three different tissues, revealed a 600-fold range of apparent constitutive expression levels. For most of the genes assayed, good correlations were observed among the expression levels in normal mammary, bronchial, and epidermal epithelial cells.
- Published
- 2005
- Full Text
- View/download PDF
44. Characterization of the tumor suppressor gene WWOX in primary human oral squamous cell carcinomas
- Author
-
Dawidson Assis Gomes, Marcus Vinicius Gomez, Alvimar Afonso Barbosa, Luiz De Marco, C. Marcelo Aldaz, Ricardo Santiago Gomez, Paôlla Freitas Perdigão, Flávio Juliano Pimenta, and Marco Aurélio Romano-Silva
- Subjects
WWOX ,Cancer Research ,DNA, Complementary ,Transcription, Genetic ,Tumor suppressor gene ,Biology ,medicine.disease_cause ,Article ,Exon ,medicine ,Humans ,RNA, Messenger ,Mouth neoplasm ,Base Sequence ,Tumor Suppressor Proteins ,Chromosomal fragile site ,Cancer ,medicine.disease ,Immunohistochemistry ,Candidate Tumor Suppressor Gene ,Gene Expression Regulation, Neoplastic ,stomatognathic diseases ,WW Domain-Containing Oxidoreductase ,Oncology ,Carcinoma, Squamous Cell ,Cancer research ,Mouth Neoplasms ,Oxidoreductases ,Carcinogenesis - Abstract
Oral squamous cell carcinoma (OSCC) is the most common malignant neoplasm of the oral cavity, representing ~90% of all oral carcinomas and accounting for 3–5% of all malignancies. The WWOX gene (WW-domain containing oxidoreductase) is a candidate tumor suppressor gene located at 16q23.3–24.1, spanning the second most common fragile site, FRA16D. In this report, the role of the WWOX gene was investigated in 20 tumors and 10 normal oral mucosas, and we demonstrated an altered WWOX gene in 50% (10/20) of OSCCs. Using nested RT-PCR, mRNA transcription was altered in 35% of the tumors, with the complete absence of transcripts in 2 samples as well as absence of exons 6–8 (2 tumors), exon 7 (1 tumor), exon 7 and exon 6–8 (1 tumor) and partial loss of exons 8 and 9 (1 tumor). To determine if the aberrant transcripts were translated, Western blots were performed in all samples; however, only the normal protein was detected. By immunohistochemistry, a reduction in Wwox protein expression was observed, affecting 40% of the tumors when compared with normal mucosa. In addition, a novel somatic mutation (S329F) was found. The presence of alterations in mRNA transcription correlated with the reduced expression of Wwox protein in the tumors. These results show that the WWOX gene is frequently altered in OSCC and may contribute to the carcinogenesis processes in oral cancer. ' 2005 Wiley-Liss, Inc.
- Published
- 2005
- Full Text
- View/download PDF
45. Expression of common chromosomal fragile site genes, WWOX/FRA16D and FHIT/FRA3B is downregulated by exposure to environmental carcinogens, UV, and BPDE but not by IR
- Author
-
C. Marcelo Aldaz, Elangovan Thavathiru, Michael C. MacLeod, and John H. Ludes-Meyers
- Subjects
WWOX ,Cancer Research ,Cell cycle checkpoint ,Ultraviolet Rays ,DNA damage ,7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide ,Down-Regulation ,Biology ,medicine.disease_cause ,Article ,FHIT ,Cell Line, Tumor ,Radiation, Ionizing ,medicine ,Humans ,Molecular Biology ,Chromosome Fragile Sites ,Tumor Suppressor Proteins ,Chromosomal fragile site ,Cell cycle ,Molecular biology ,Carcinogens, Environmental ,Acid Anhydride Hydrolases ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,WW Domain-Containing Oxidoreductase ,Chromosome Fragile Site ,Tumor Suppressor Protein p53 ,Oxidoreductases ,Carcinogenesis - Abstract
Common chromosomal fragile sites are unstable genomic loci susceptible to breakage, rearrangement, and are highly recombinogenic. Frequent alterations at these loci in tumor cells led to the hypothesis that they may contribute to cancer development. The two most common chromosomal fragile sites FRA16D and FRA3B which harbor WWOX and FHIT genes, respectively, are frequently altered in human cancers. Here we report that environmental carcinogens, ultraviolet (UV) light, and Benzo[a]pyrene diol epoxide (BPDE), significantly downregulate expression of both genes. On the other hand, we observe that ionizing radiation (IR) does not affect expression of these genes, suggesting that the effect of repression exerted by UV and BPDE is not just a consequence of DNA damage but may be a result of different signaling pathways triggered by specific DNA lesions. Such downregulation correlates with an induction of an S-phase delay in the cell cycle. Treatment of UV-irradiated cells with caffeine abrogates the S-phase delay while concomitantly overcoming the repression phenomenon. This suggests the involvement of unique cell cycle checkpoint mechanisms in the observed repression. Therefore, it is hypothesized that protracted downregulation of the putative tumor suppressor genes WWOX and FHIT by environmental carcinogens may constitute an additional mechanism of relevance in the initiation of tumorigenesis.
- Published
- 2005
- Full Text
- View/download PDF
46. From Mice to Humans
- Author
-
Daniel Medina, C. Marcelo Aldaz, Martín Carlos Abba, Hongxia Sun, Yuhui Hu, Sally Gaddis, Frances S. Kittrell, Aysegul Sahin, Keith A. Baggerly, Jeffrey A. Drake, and Li Deng
- Subjects
Regulation of gene expression ,Genetics ,Cancer Research ,Cancer Model ,Cancer ,Biology ,medicine.disease ,medicine.disease_cause ,Gene expression profiling ,Breast cancer ,Oncology ,medicine ,Cancer research ,Gene family ,Serial analysis of gene expression ,Carcinogenesis - Abstract
Genetically engineered mouse mammary cancer models have been used over the years as systems to study human breast cancer. However, much controversy exists on the utility of such models as valid equivalents to the human cancer condition. To perform an interspecies gene expression comparative study in breast cancer we used a mouse model that most closely resembles human breast carcinogenesis. This system relies on the transplant of p53 null mouse mammary epithelial cells into the cleared mammary fat pads of syngeneic hosts. Serial analysis of gene expression (SAGE) was used to obtain gene expression profiles of normal and tumor samples from this mouse mammary cancer model (>300,000 mouse mammary-specific tags). The resulting mouse data were compared with 25 of our human breast cancer SAGE libraries (>2.5 million human breast-specific tags). We observed significant similarities in the deregulation of specific genes and gene families when comparing mouse with human breast cancer SAGE data. A total of 72 transcripts were identified as commonly deregulated in both species. We observed a systematic and significant down-regulation in all of the tumors from both species of various cytokines, including CXCL1 (GRO1), LIF, interleukin 6, and CCL2. All of the mouse and most human mammary tumors also displayed decreased expression of genes known to inhibit cell proliferation, including NFKBIA (IKBα), GADD45B, and CDKN1A (p21); transcription-related genes such as CEBP, JUN, JUNB, and ELF1; and apoptosis-related transcripts such as IER3 and GADD34/PPP1R15A. Examples of overexpressed transcripts in tumors from both species include proliferation-related genes such as CCND1, CKS1B, and STMN1 (oncoprotein 18); and genes related to other functions such as SEPW1, SDFR1, DNCI2, and SP110. Importantly, abnormal expression of several of these genes has not been associated previously with breast cancer. The consistency of these observations was validated in independent mouse and human mammary cancer sets.This is the first interspecies comparison of mammary cancer gene expression profiles. The comparative analysis of mouse and human SAGE mammary cancer data validates this p53 null mouse tumor model as a useful system closely resembling human breast cancer development and progression. More importantly, these studies are allowing us to identify relevant biomarkers of potential use in human studies while leading to a better understanding of specific mechanisms of human breast carcinogenesis.
- Published
- 2004
- Full Text
- View/download PDF
47. Differential expression in SAGE: accounting for normal between-library variation
- Author
-
Jeffrey S. Morris, Keith A. Baggerly, Li Deng, and C. Marcelo Aldaz
- Subjects
Statistics and Probability ,Inference ,Biology ,Sensitivity and Specificity ,Biochemistry ,Genetic variation ,Statistics ,Econometrics ,Test statistic ,Differential expression ,Molecular Biology ,Gene Library ,Expressed Sequence Tags ,Models, Statistical ,Models, Genetic ,Gene Expression Profiling ,SAGE ,Genetic Variation ,Reproducibility of Results ,Sampling (statistics) ,Sequence Analysis, DNA ,Computer Science Applications ,Computational Mathematics ,Variation (linguistics) ,Normal variation ,Computational Theory and Mathematics ,Algorithms - Abstract
Motivation: In contrasting levels of gene expression between groups of SAGE libraries, the libraries within each group are often combined and the counts for the tag of interest summed, and inference is made on the basis of these larger ‘pseudolibraries’. While this captures the sampling variability inherent in the procedure, it fails to allow for normal variation in levels of the gene between individuals within the same group, and can consequently overstate the significance of the results. The effect is not slight: between-library variation can be hundreds of times the within-library variation. Results: We introduce a beta-binomial sampling model that correctly incorporates both sources of variation. We show how to fit the parameters of this model, and introduce a test statistic for differential expression similar to a two-sample t-test. Contact: kabagg@mdanderson.org Supplementary information http://bioinformatics.mdanderson.org/ Includes Matlab and R code for fitting the model. * To whom correspondence should be addressed.
- Published
- 2003
- Full Text
- View/download PDF
48. Celecoxib and difluoromethylornithine in combination have strong therapeutic activity against UV-induced skin tumors in mice
- Author
-
C. Marcelo Aldaz, Ronald A. Lubet, Jaye L. Viner, Susan M. Fischer, and Claudio J. Conti
- Subjects
Keratinocytes ,Cancer Research ,medicine.medical_specialty ,Pathology ,Eflornithine ,Neoplasms, Radiation-Induced ,Skin Neoplasms ,Ultraviolet Rays ,Ratón ,medicine.medical_treatment ,Urology ,Antineoplastic Agents ,Apoptosis ,Ornithine Decarboxylase ,medicine.disease_cause ,Mice ,Oral administration ,medicine ,Animals ,Enzyme Inhibitors ,Anticarcinogen ,Skin ,Mice, Hairless ,Sulfonamides ,Chemotherapy ,business.industry ,General Medicine ,Ornithine Decarboxylase Inhibitors ,medicine.disease ,Isoenzymes ,Celecoxib ,Cyclooxygenase 2 ,Prostaglandin-Endoperoxide Synthases ,Disease Progression ,Pyrazoles ,Drug Therapy, Combination ,Skin cancer ,Carcinogenesis ,business ,Cell Division ,medicine.drug - Abstract
The cyclooxygenase-2 (COX-2) inhibitor celecoxib and the ornithine decarboxylase (ODC) inhibitor difluoromethylornithine (DFMO) were each previously shown to prevent skin tumor development when administered throughout the course of UV irradiation. This raised the question of whether maintenance or continued growth of existing tumors required prostaglandins, the product of COX, or polyamines, the product of ODC. To address this question, SKH hairless mice were irradiated 3 times/week with 90 mJ/cm(2); this dose was increased 10% weekly to a maximum of 175 mJ/cm(2). UV was stopped at 27 weeks, at which time there were an average of 5 papillomas/mouse. The mice were then placed in one of four treatment groups: group 1, no treatment; group 2, 0.4% DFMO in the drinking water; group 3, 500 p.p.m. celecoxib in the diet (AIN76); group 4, both DFMO and celecoxib. The control group continued to produce new tumors in a nearly linear manner such that by week 31 the tumor number had nearly doubled, i.e. approximately 10 tumors/mouse. The group receiving DFMO showed significant tumor regression, losing an average of 1 tumor/mouse/week, such that 50% of the tumors remained at week 31. The celecoxib group showed a 25% reduction in tumor number. The group receiving the combination of celecoxib and DFMO showed the greatest regression, with an 89% reduction in tumor number compared with the control group. There was also a corresponding reduction in the size of the tumors. To determine whether tumor regression was permanent or required continued treatment, all treatments were stopped at 31 weeks. Over the next 4 weeks, tumors reappeared at the same rate in all treatment groups. It is concluded that the combination of celecoxib and DFMO are potent therapeutic agents for skin cancer, although the benefits are lost with the cessation of treatment.
- Published
- 2003
- Full Text
- View/download PDF
49. Serial analysis of gene expression in normal p53 null mammary epithelium
- Author
-
Sally Gaddis, Yuhui Hu, Frances S. Kittrell, C. Marcelo Aldaz, Daniel Medina, and Rachael L. Daniel
- Subjects
Cancer Research ,Biology ,medicine.disease_cause ,Transcriptome ,Mice ,Mammary Glands, Animal ,Gene expression ,Genetics ,medicine ,Animals ,Humans ,Serial analysis of gene expression ,Molecular Biology ,Expressed Sequence Tags ,Mice, Inbred BALB C ,Gene Expression Profiling ,Epithelial Cells ,Genes, p53 ,Null allele ,Epithelium ,Gene expression profiling ,medicine.anatomical_structure ,Gene Expression Regulation ,Mammary Epithelium ,Cytochrome P-450 CYP1B1 ,Cancer research ,Female ,Aryl Hydrocarbon Hydroxylases ,Carcinogenesis - Abstract
Much evidence has accumulated implicating the p53 gene as of importance in breast carcinogenesis. However, much still remains to be uncovered on the specific downstream pathways influenced by this important activator/repressor of transcription. This study investigated the effects of a p53 null genotype on the transcriptome of 'normal' mouse mammary epithelium using a unique in vivo model of preneoplastic transformation. We used SAGE for the comparative analysis of p53 wild type (wt) and null mammary epithelium unexposed and exposed to hormonal stimulation. Analysis of the hormone exposed samples provided a comprehensive view of the dramatic changes in gene expression as consequence of the functional differentiation of the mammary epithelium in an in vivo system. We detected the dysregulation in p53(null) epithelium of
- Published
- 2002
- Full Text
- View/download PDF
50. Karyotypic evolutions of cancer species in rats during the long latent periods after injection of nitrosourea
- Author
-
Amanda McCormack, Daniele Mandrioli, Christian Fiala, C. Marcelo Aldaz, Mathew Bloomfield, and Peter H. Duesberg
- Subjects
medicine.medical_specialty ,Individual clonal karyotypes of cancers ,Karyotype arrays ,Aneuploidy ,Biology ,medicine.disease_cause ,Biochemistry ,medicine ,Genetics ,Genetics(clinical) ,Molecular Biology ,Carcinogens function as aneuploidogens ,Genetics (clinical) ,Biochemistry, medical ,Research ,Biochemistry (medical) ,Cytogenetics ,Cancer ,Karyotype ,medicine.disease ,Phenotype ,Human genetics ,Single-step origins of cancer karyotypes ,Persistent non-clonal hyperplasias ,Molecular Medicine ,Carcinogenesis ,Clonal selection - Abstract
Background A century of research has established that cancers arise from tissues exposed to carcinogens only after long latencies of years to decades and have individual clonal karyotypes. Since speciation from known precursors also depends on long latencies and new species also have individual karyotypes, we and others have recently proposed that carcinogenesis is a form of speciation. According to this theory karyotypic evolutions generate new cancer species from normal cells as follows: Carcinogens induce aneuploidy (Figure 1). By unbalancing thousands of genes aneuploidy automatically destabilizes the karyotype and thus catalyzes random karyotypic variations. Selections of variants with proliferative phenotypes form non-clonal hyperplasias with persistently varying karyotypes. Very rare karyotypic variations form new cancer species with individual clonal karyotypes. Despite destabilization by the resulting congenital aneuploidies, cancer karyotypes are stabilized within narrow margins of variation by clonal selections for cancer-specific autonomy. Because all non-cancerous aneuploidies are unstable, all aneusomies of prospective cancers are joined in single-steps, rather than gradually. Since this mechanism is very inefficient, it predicts long latent periods from carcinogens to cancers and individual clonal cancer karyotypes. Results Here we have tested the predicted roles of karyotypic evolutions during the time course of carcinogenesis in an established experimental system. In this system injection of nitrosourea induces in female rats non-invasive mammary hyperplasias (“tumors”) after two or more months, and invasive carcinomas after six or more months. Accordingly four specific predictions were tested: (1) Invasive cancers are late and carry individual clonal karyotypes and phenotypes, (2) Persistent hyperplasias carry non-clonal karyotypes, (3) Non-clonal hyperplasias generate clonal cancers spontaneously but rarely, (4) Cancer-karyotypes arise with all individual clonal aneusomies in single-steps. All four predictions were experimentally confirmed. Conclusions Our results along with the literature reveal a coherent karyotypic mechanism of carcinogenesis: Carcinogens induce aneuploidy. The inherent instability of aneuploidy automatically catalyzes new karyotypic variations. Aneuploid karyotypes with proliferative phenotypes form varying non-clonal hyperplasias. Rare variations form cancer species with individual clonal karyotypes, which are stabilized by clonal selection for autonomy. The low odds of this mechanism explain the long latencies of carcinogenesis, the individuality and karyotypic clonality of cancers.
- Published
- 2014
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.