241 results on '"Stuart K. Calderwood"'
Search Results
2. Cancer extracellular vesicles, tumoroid models, and tumor microenvironment
- Author
-
Takanori, Eguchi, Mona, Sheta, Masanori, Fujii, and Stuart K, Calderwood
- Subjects
Exosome ,3D tumoroid models ,Cancer Research ,Tumor microenvironment ,Cancer stem cells ,Neoplasms ,Humans ,Tissue Distribution ,Extracellular vesicles ,Exosomes ,Metastatic niche - Abstract
Cancer extracellular vesicles (EVs), or exosomes, promote tumor progression through enhancing tumor growth, initiating epithelial-to-mesenchymal transition, remodeling the tumor microenvironment, and preparing metastatic niches. Three-dimensionally (3D) cultured tumoroids / spheroids aim to reproduce some aspects of tumor behavior in vitro and show increased cancer stem cell properties. These properties are transferred to their EVs that promote tumor growth. Moreover, recent tumoroid models can be furnished with aspects of the tumor microenvironment, such as vasculature, hypoxia, and extracellular matrix. This review summarizes tumor tissue culture and engineering platforms compatible with EV research. For example, the combination experiments of 3D-tumoroids and EVs have revealed multifunctional proteins loaded in EVs, such as metalloproteinases and heat shock proteins. EVs or exosomes are able to transfer their cargo molecules to recipient cells, whose fates are often largely altered. In addition, the review summarizes approaches to EV labeling technology using fluorescence and luciferase, useful for studies on EV-mediated intercellular communication, biodistribution, and metastatic niche formation.
- Published
- 2022
3. Publisher Correction: Extracellular Hsp90α stimulates a unique innate gene profile in microglial cells with simultaneous activation of Nrf2 and protection from oxidative stress
- Author
-
Yuka Okusha, Benjamin J. Lang, Ayesha Murshid, Thiago J. Borges, Kristina M. Holton, Joanne Clark-Matott, Sachin Doshi, Tsuneya Ikezu, and Stuart K. Calderwood
- Subjects
Cell Biology ,Biochemistry - Published
- 2022
4. Data from Targeting Cdc37 Inhibits Multiple Signaling Pathways and Induces Growth Arrest in Prostate Cancer Cells
- Author
-
Stuart K. Calderwood, Mary Ann Stevenson, and Phillip J. Gray
- Abstract
Members of the 90-kDa heat shock protein (HSP90) family are known to bind and stabilize intermediates in a wide variety of cell signaling pathways and contribute to their dysregulation in cancer. An important intracellular cofactor for HSP90 is Cdc37, a protein with a broad role in fostering the activities of protein kinases. By targeting Cdc37 using RNA interference, we have shown that the loss of Cdc37 function induces irreversible growth arrest in androgen receptor-positive and -negative prostate carcinoma cells. In contrast to HSP90-directed agents, Cdc37 targeting seems to affect cancer cells through a distinct mechanism and does not significantly deplete the intracellular levels of most known HSP90 client proteins. Instead, Cdc37 depletion inhibits cellular kinase activity and flux through growth-promoting signal transduction cascades. We show that the loss of Cdc37 leads to reduced activity of the Erk, Akt, mTOR, and androgen-induced pathways. We have also discovered synergistic interactions between Cdc37 inactivation and the HSP90-inhibitory anticancer drug 17-(allylamino)-17-demethoxygeldanamycin (17AAG). These interactions involve enhanced degradation of proteins essential for growth and inhibition of 17AAG-induced expression of the antiapoptotic HSP70. Thus, Cdc37 is essential for maintaining prostate tumor cell growth and may represent a novel target in the search for multitargeted therapies based on the HSP90 chaperone system. [Cancer Res 2007;67(24):11942–50]
- Published
- 2023
5. Supplementary Figure 1 from Targeting Cdc37 Inhibits Multiple Signaling Pathways and Induces Growth Arrest in Prostate Cancer Cells
- Author
-
Stuart K. Calderwood, Mary Ann Stevenson, and Phillip J. Gray
- Abstract
Supplementary Figure 1 from Targeting Cdc37 Inhibits Multiple Signaling Pathways and Induces Growth Arrest in Prostate Cancer Cells
- Published
- 2023
6. Data Supplement from Hsp70–Bag3 Interactions Regulate Cancer-Related Signaling Networks
- Author
-
Michael Y. Sherman, Shinichi Takayama, Jason E. Gestwicki, Jennifer N. Rauch, Z.T. Young, Xiaokai Li, Mikel Garcia-Marcos, Irina A. Zamulaeva, Nina V. Orlova, Svetlana G. Smirnova, Olga N. Matchuk, Suryaram Gummuluru, Hu Li, Stuart K. Calderwood, Jianlin Gong, Vladimir L. Gabai, and Teresa A. Colvin
- Abstract
Supplemental Figure S2. Effect of Hsp70 or Bag3 depletion in HCT116 cells on signaling pathways.
- Published
- 2023
7. The XIth International Online Symposium on Heat Shock Proteins in Biology and Medicine
- Author
-
Stuart K, Calderwood, Elizabeth, Repasky, Len, Neckers, and Lawrence, Hightower
- Subjects
Neoplasms ,Humans ,Medicine ,Cell Biology ,Biology ,Meeting Review ,Biochemistry ,Heat-Shock Proteins ,Heat-Shock Response - Abstract
Single cell and multicellular organisms encounter physical stress from their environment as well as behavioral stress experienced in more complex organisms. As these stresses can present an existential threat, organisms respond with a coordinated response at the tissue and cellular level, the heat shock response (HSR) and this was the major theme of the symposium. Much of the meeting was concentrated on the heat shock proteins (HSPs), the effector molecules of the response. The balance between the potency of the HSR and the experience of stress naturally plays a key role in the etiology of many disease. Roles in cancer, the immune response, cell metabolism and aging were discussed at length at the meeting. Finally, a major goal of this field is to enhance the HSR in pathological conditions where it becomes inadequate or over stimulated and important findings regarding pharmacological approaches to modulating the HSR were discussed.
- Published
- 2022
8. Cdc37 as a Co-chaperone to Hsp90
- Author
-
Thomas L. Prince, Benjamin J. Lang, Yuka Okusha, Takanori Eguchi, and Stuart K. Calderwood
- Published
- 2022
9. Cdc37 as a Co-chaperone to Hsp90
- Author
-
Thomas L, Prince, Benjamin J, Lang, Yuka, Okusha, Takanori, Eguchi, and Stuart K, Calderwood
- Abstract
The co-chaperone p50/Cdc37 is an important partner for Hsp90, assisting in molecular chaperone activities, particularly with regard to the regulation of protein kinases. Analysis of the structure of Hsp90-Cdc37-kinase complexes demonstrates the way in which Cdc37 interacts with and controls the folding of a large proportion of intracellular protein kinases. This co-chaperone thus stands at the hub of a multitude of intracellular signaling networks. Indeed, the influence of Cdc37 reaches beyond the housekeeping pathways of protein folding into the regulation of a wide range of cellular processes. This co-chaperone has attracted attention as a potential intermediate in carcinogenesis. Cdc37 is an attractive potential target in cancer due to (1) high expression in a number of tumor types and (2) control of multiple signaling pathways. These properties indicate (3) a potential for selectivity due to its elevated expression in malignant cells and (4) robustness, as the co-chaperone may control multiple growth signaling pathways and thus be less prone to evolution of resistance than less versatile oncoproteins. Cdc37 may also be involved in other aspects of pathophysiology and has been shown to be secreted in exosomes. Protein aggregation disorders have been linked to age-related declines in molecular chaperones and co-chaperones. Cdc37 also appears to be a potential agent in longevity due to its links to protein folding and autophagy, and it will be informative to study the role of Cdc37 maintenance/decline in aging organisms.
- Published
- 2022
10. SCAND1 Reverses Epithelial-to-Mesenchymal Transition (EMT) and Suppresses Prostate Cancer Growth and Migration
- Author
-
Takanori Eguchi, Eva Csizmadia, Hotaka Kawai, Mona Sheta, Kunihiro Yoshida, Thomas L. Prince, Barbara Wegiel, and Stuart K. Calderwood
- Subjects
epithelial-to-mesenchymal transition (EMT) ,hybrid E/M ,partial EMT ,SCAND1 ,MZF1 ,SCAN zinc finger transcription factors ,gene expression ,cancer prognosis ,collective migration ,metastasis ,General Medicine - Abstract
Epithelial–mesenchymal transition (EMT) is a reversible cellular program that transiently places epithelial (E) cells into pseudo-mesenchymal (M) cell states. The malignant progression and resistance of many carcinomas depend on EMT activation, partial EMT, or hybrid E/M status in neoplastic cells. EMT is activated by tumor microenvironmental TGFβ signal and EMT-inducing transcription factors, such as ZEB1/2, in tumor cells. However, reverse EMT factors are less studied. We demonstrate that prostate epithelial transcription factor SCAND1 can reverse the cancer cell mesenchymal and hybrid E/M phenotypes to a more epithelial, less invasive status and inhibit their proliferation and migration in DU-145 prostate cancer cells. SCAND1 is a SCAN domain-containing protein and hetero-oligomerizes with SCAN-zinc finger transcription factors, such as MZF1, for accessing DNA and the transcriptional co-repression of target genes. We found that SCAND1 expression correlated with maintaining epithelial features, whereas the loss of SCAND1 was associated with mesenchymal phenotypes of tumor cells. SCAND1 and MZF1 were mutually inducible and coordinately included in chromatin with hetero-chromatin protein HP1γ. The overexpression of SCAND1 reversed hybrid E/M status into an epithelial phenotype with E-cadherin and β-catenin relocation. Consistently, the co-expression analysis in TCGA PanCancer Atlas revealed that SCAND1 and MZF1 expression was negatively correlated with EMT driver genes, including CTNNB1, ZEB1, ZEB2 and TGFBRs, in prostate adenocarcinoma specimens. In addition, SCAND1 overexpression suppressed tumor cell proliferation by reducing the MAP3K-MEK-ERK signaling pathway. Of note, in a mouse tumor xenograft model, SCAND1 overexpression significantly reduced Ki-67(+) and Vimentin(+) tumor cells and inhibited migration and lymph node metastasis of prostate cancer. Kaplan–Meier analysis showed high expression of SCAND1 and MZF1 to correlate with better prognoses in pancreatic cancer and head and neck cancers, although with poorer prognosis in kidney cancer. Overall, these data suggest that SCAND1 induces expression and coordinated heterochromatin-binding of MZF1 to reverse the hybrid E/M status into an epithelial phenotype and, inhibits tumor cell proliferation, migration, and metastasis, potentially by repressing the gene expression of EMT drivers and the MAP3K-MEK-ERK signaling pathway.
- Published
- 2022
11. MicroRNA-570 targets the HSP chaperone network, increases proteotoxic stress and inhibits mammary tumor cell migration
- Author
-
Yuka Okusha, Martin E. Guerrero-Gimenez, Benjamin J. Lang, Thiago J. Borges, Mary A. Stevenson, Andrew W. Truman, and Stuart K. Calderwood
- Subjects
Gene Expression Regulation, Neoplastic ,MicroRNAs ,Multidisciplinary ,Cell Movement ,Untranslated Regions ,Cell Line, Tumor ,Humans ,Neoplasm Invasiveness ,Molecular Chaperones - Abstract
The dynamic network of chaperone interactions known as the chaperome contributes significantly to the proteotoxic cell response and the malignant phenotype. To bypass the inherent redundancy in the network, we have used a microRNA (mir) approach to target multiple members of the chaperome simultaneously. We identified a potent microRNA, miR-570 that could bind the 3′untranslated regions of multiple HSP mRNAs and inhibit HSP synthesis. Transfection of cells with this miR species reduced expression of multiple HSPs, inhibited the heat shock response and reduced tumor cell growth while acted additively in combination with cytotoxic drugs. As overexpression of miR-570 elicited tumor suppressive effects, we inferred that this miR could play a potential role in inhibiting tumorigenesis and cancer cell growth. In accordance with this hypothesis, we determined a significant role for miR-570 in regulating markers of mammary tumor progression, including cell motility and invasion. Our data provide a proof of the principle that the tumor chaperome can be targeted by microRNAs suggesting a potential therapeutic avenue towards cancer therapy.
- Published
- 2022
12. Extracellular Hsp90 and protection of neuronal cells through Nrf2
- Author
-
Ayesha Murshid, Thiago J Borges, Benjamin J. Lang, Stuart K. Calderwood, Yuka Okusha, Takanori Eguchi, and Thomas Prince
- Subjects
Neurons ,Microglia ,biology ,NF-E2-Related Factor 2 ,Chemistry ,Cellular homeostasis ,Biochemistry ,Antioxidants ,Cell biology ,Oxidative Stress ,medicine.anatomical_structure ,Phagocytosis ,Heat shock protein ,Chaperone (protein) ,Extracellular ,medicine ,biology.protein ,Humans ,HSP90 Heat-Shock Proteins ,Transcellular ,Heat shock ,Intracellular ,Molecular Chaperones ,Signal Transduction - Abstract
Heat shock protein 90 (Hsp90), although one of the most essential intracellular chaperones, can also play key roles in the extracellular milieu. Here, we review the properties of extracellular Hsp90 in cellular homeostasis in the heat shock response (HSR), focusing on cells of the central nervous system. Hsp90 can be secreted by microglia as well as other cell types by non-canonical pathways of secretion. The chaperone may then influence the behavior of distant cells and can for instance protect neuronal cells from the oxidative burst accompanying phagocytosis by microglia of beta-amyloid fibrils. A mechanism involving activation of the transcription factor Nrf2, and induction of the antioxidant response is reported. We review the potential role of extracellular Hsp90, Nrf2 and transcellular chaperone signaling in the non-cell-intrinsic HSR.
- Published
- 2021
13. The functions and regulation of heat shock proteins; key orchestrators of proteostasis and the heat shock response
- Author
-
Cristina Bonorino, Martin E Guerrero, Stuart K. Calderwood, Benjamin J. Lang, Thomas Prince, and Yuka Okusha
- Subjects
0301 basic medicine ,Programmed cell death ,Effector ,Health, Toxicology and Mutagenesis ,General Medicine ,010501 environmental sciences ,Cell fate determination ,Biology ,Toxicology ,01 natural sciences ,Cell biology ,Heat stress ,03 medical and health sciences ,030104 developmental biology ,Proteostasis ,Heat shock protein ,Treatment strategy ,Heat shock ,0105 earth and related environmental sciences - Abstract
Cells respond to protein-damaging (proteotoxic) stress by activation of the Heat Shock Response (HSR). The HSR provides cells with an enhanced ability to endure proteotoxic insults and plays a crucial role in determining subsequent cell death or survival. The HSR is, therefore, a critical factor that influences the toxicity of protein stress. While named for its vital role in the cellular response to heat stress, various components of the HSR system and the molecular chaperone network execute essential physiological functions as well as responses to other diverse toxic insults. The effector molecules of the HSR, the Heat Shock Factors (HSFs) and Heat Shock Proteins (HSPs), are also important regulatory targets in the progression of neurodegenerative diseases and cancers. Modulation of the HSR and/or its extended network have, therefore, become attractive treatment strategies for these diseases. Development of effective therapies will, however, require a detailed understanding of the HSR, important features of which continue to be uncovered and are yet to be completely understood. We review recently described and hallmark mechanistic principles of the HSR, the regulation and functions of HSPs, and contexts in which the HSR is activated and influences cell fate in response to various toxic conditions.
- Published
- 2021
14. Stress-Inducible SCAND Factors Suppress the Stress Response and Are Biomarkers for Enhanced Prognosis in Cancers
- Author
-
Mona Sheta, Kunihiro Yoshida, Hideka Kanemoto, Stuart K. Calderwood, and Takanori Eguchi
- Subjects
cell stress response ,heat shock factor (HSF) ,SCAN domain (SCAND)-containing proteins ,Organic Chemistry ,heat shock protein 90 (HSP90) ,long noncoding RNA (lncRNA) ,General Medicine ,cancer patient prognosis ,co-expression correlation ,Catalysis ,Computer Science Applications ,Inorganic Chemistry ,ZSCAN6 ,Kaplan-Meier plot ,MZF1/ZSCAN6 ,Kaplan–Meier plot ,MZF1 ,Physical and Theoretical Chemistry ,Molecular Biology ,Spectroscopy - Abstract
The cell stress response is an essential system present in every cell for responding and adapting to environmental stimulations. A major program for stress response is the heat shock factor (HSF)–heat shock protein (HSP) system that maintains proteostasis in cells and promotes cancer progression. However, less is known about how the cell stress response is regulated by alternative transcription factors. Here, we show that the SCAN domain (SCAND)-containing transcription factors (SCAN-TFs) are involved in repressing the stress response in cancer. SCAND1 and SCAND2 are SCAND-only proteins that can hetero-oligomerize with SCAN-zinc finger transcription factors, such as MZF1(ZSCAN6), for accessing DNA and transcriptionally co-repressing target genes. We found that heat stress induced the expression of SCAND1, SCAND2, and MZF1 bound to HSP90 gene promoter regions in prostate cancer cells. Moreover, heat stress switched the transcript variants’ expression from long noncoding RNA (lncRNA-SCAND2P) to protein-coding mRNA of SCAND2, potentially by regulating alternative splicing. High expression of HSP90AA1 correlated with poorer prognoses in several cancer types, although SCAND1 and MZF1 blocked the heat shock responsiveness of HSP90AA1 in prostate cancer cells. Consistent with this, gene expression of SCAND2, SCAND1, and MZF1 was negatively correlated with HSP90 gene expression in prostate adenocarcinoma. By searching databases of patient-derived tumor samples, we found that MZF1 and SCAND2 RNA were more highly expressed in normal tissues than in tumor tissues in several cancer types. Of note, high RNA expression of SCAND2, SCAND1, and MZF1 correlated with enhanced prognoses of pancreatic cancer and head and neck cancers. Additionally, high expression of SCAND2 RNA was correlated with better prognoses of lung adenocarcinoma and sarcoma. These data suggest that the stress-inducible SCAN-TFs can function as a feedback system, suppressing excessive stress response and inhibiting cancers.
- Published
- 2023
15. Extracellular Hsp90α stimulates a unique innate gene profile in microglial cells with simultaneous activation of Nrf2 and protection from oxidative stress
- Author
-
Yuka Okusha, Benjamin J. Lang, Ayesha Murshid, Thiago J. Borges, Kristina M. Holton, Joanne Clark-Matott, Sachin Doshi, Tsuneya Ikezu, and Stuart K. Calderwood
- Subjects
Lipopolysaccharides ,Amyloid beta-Peptides ,NF-E2-Related Factor 2 ,NF-kappa B ,Neurodegenerative Diseases ,Cell Biology ,Nitric Oxide ,Biochemistry ,Publisher Correction ,Oxidative Stress ,Culture Media, Conditioned ,Humans ,Original Article ,HSP90 Heat-Shock Proteins ,Microglia ,Heat-Shock Proteins - Abstract
Delivery of exogenous heat shock protein 90α (Hsp90α) and/or its induced expression in neural tissues has been suggested as a potential strategy to combat neurodegenerative disease. However, within a neurodegenerative context, a pro-inflammatory response to extracellular Hsp90α (eHsp90α) could undermine strategies to use it for therapeutic intervention. The aim of this study was to investigate the biological effects of eHsp90α on microglial cells, the primary mediators of inflammatory responses in the brain. Transcriptomic profiling by RNA-seq of primary microglia and the cultured EOC2 microglial cell line treated with eHsp90α showed the chaperone to stimulate activation of innate immune responses in microglia that were characterized by an increase in NF-kB-regulated genes. Further characterization showed this response to be substantially lower in amplitude than the effects of other inflammatory stimuli such as fibrillar amyloid-β (fAβ) or lipopolysaccharide (LPS). Additionally, the toxicity of conditioned media obtained from microglia treated with fAβ was attenuated by addition of eHsp90α. Using a co-culture system of microglia and hippocampal neuronal cell line HT22 cells separated by a chamber insert, the neurotoxicity of medium conditioned by microglia treated with fAβ was reduced when eHsp90α was also added. Mechanistically, eHsp90α was shown to activate Nrf2, a response which attenuated fAβ-induced nitric oxide production. The data thus suggested that eHsp90α protects against fAβ-induced oxidative stress. We also report eHsp90α to induce expression of macrophage receptor with collagenous structure (Marco), which would permit receptor-mediated endocytosis of fAβ. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1007/s12192-022-01279-9.
- Published
- 2022
16. Host expression system modulates recombinant Hsp70 activity through post‐translational modifications
- Author
-
Nitika, Thiago J. Borges, Benjamin J. Lang, Stuart K. Calderwood, Donald Wolfgeher, Cristina Bonorino, Maurício Menegatti Rigo, Ayesha Murshid, and Andrew W. Truman
- Subjects
0301 basic medicine ,biology ,Chemistry ,In silico ,Cell Biology ,biology.organism_classification ,Biochemistry ,Article ,Cell biology ,Pichia pastoris ,Hsp70 ,law.invention ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,law ,030220 oncology & carcinogenesis ,Heat shock protein ,Foldase ,Recombinant DNA ,Phosphorylation ,Molecular Biology ,Intracellular - Abstract
The use of model organisms for recombinant protein production results in the addition of model-specific post-translational modifications (PTMs) that can affect the structure, charge, and function of the protein. The 70-kDa heat shock proteins (Hsp70) were originally described as intracellular chaperones, with ATPase and foldase activity. More recently, new extracellular activities of Hsp70 proteins (e.g., as immunomodulators) have been identified. While some studies indicate an inflammatory potential for extracellular Hsp70 proteins, others suggest an immunosuppressive activity. We hypothesized that the production of recombinant Hsp70 in different expression systems would result in the addition of different PTMs, perhaps explaining at least some of these opposing immunological outcomes. We produced and purified Mycobacterium tuberculosis DnaK from two different systems, Escherichia coli and Pichia pastoris, and analyzed by mass spectrometry of the protein preparations, investigating the impact of PTMs in an in silico and in vitro perspective. The comparisons of DnaK structures in silico highlighted that electrostatic and topographical differences exist that are dependent upon the expression system. Production of DnaK in the eukaryotic system dramatically affected its ATPase activity and significantly altered its ability to downregulate MHC II and CD86 expression on murine dendritic cells (DCs). Phosphatase treatment of DnaK indicated that some of these differences related specifically to phosphorylation. Altogether, our data indicate that PTMs are an important characteristic of the expression system, with differences that impact interactions of Hsps with their ligands and subsequent functional activities. DATABASE: Mass spectrometry proteomic data are available in the PRIDE database under the accession number PXD011583.
- Published
- 2020
17. BRCA1-BARD1 regulates transcription through modulating topoisomerase IIβ
- Author
-
Matthew J. Schellenberg, Keunsoo Kang, You Mie Lee, Benjamin P C Chen, Heeyoun Bunch, Dong-Hyung Cho, Anh T. Q. Cong, Jaehyeon Jeong, Doo Sin Jo, Donguk Kim, Deukyeong Kim, and Stuart K. Calderwood
- Subjects
endocrine system diseases ,Transcription, Genetic ,QH301-705.5 ,DNA damage ,Ubiquitin-Protein Ligases ,topoisomerase IIβ ,Immunology ,stimulus-inducible transcriptional activation ,RNA polymerase II ,Ataxia Telangiectasia Mutated Proteins ,BRCA1-BARD1 complex ,General Biochemistry, Genetics and Molecular Biology ,DNA Strand Break ,Immediate-Early Proteins ,transcription-coupled DNA break ,Transcription (biology) ,Humans ,Biology (General) ,Phosphorylation ,skin and connective tissue diseases ,Poly-ADP-Ribose Binding Proteins ,Gene ,Research Articles ,Early Growth Response Protein 1 ,Regulation of gene expression ,biology ,BRCA1 Protein ,General Neuroscience ,Topoisomerase ,Tumor Suppressor Proteins ,Research ,Ubiquitination ,Cell biology ,DNA Topoisomerases, Type II ,HEK293 Cells ,Gene Expression Regulation ,Mutation ,biology.protein ,Transcription Initiation Site ,gene regulation ,HeLa Cells - Abstract
RNA polymerase II (Pol II)-dependent transcription in stimulus-inducible genes requires topoisomerase IIβ (TOP2B)-mediated DNA strand break and the activation of DNA damage response signalling in humans. Here, we report a novel function of the breast cancer 1 (BRCA1)-BRCA1-associated ring domain 1 (BARD1) complex in this process. We found that BRCA1 is phosphorylated at S1524 by the kinases ataxia-telangiectasia mutated and ATR during gene activation, and that this event is important for productive transcription. Our biochemical and genomic analyses showed that the BRCA1-BARD1 complex interacts with TOP2B in the EGR1 transcription start site and in a large number of protein-coding genes. Intriguingly, the BRCA1-BARD1 complex ubiquitinates TOP2B, which stabilizes TOP2B binding to DNA while BRCA1 phosphorylation at S1524 controls the TOP2B ubiquitination by the complex. Together, these findings suggest the novel function of the BRCA1-BARD1 complex in the regulation of TOP2B and Pol II-mediated gene expression.
- Published
- 2021
18. BRCA1‐BARD1 complex regulates the stability of topoisomerase IIβ in transcription of human immediate early genes
- Author
-
Matthew J. Schellenberg, Donguk Kim, Heeyoun Bunch, Benjamin P C Chen, Dong-Hyung Cho, Keunsoo Kang, Stuart K. Calderwood, Jaehyeon Jeong, Doo Sin Jo, and Anh T. Q. Cong
- Subjects
Transcription (biology) ,Topoisomerase ,Genetics ,biology.protein ,Biology ,BRCA1-BARD1 complex ,Molecular Biology ,Biochemistry ,Gene ,Biotechnology ,Cell biology - Published
- 2021
19. Extracellular Hsp90α Detoxifies β-Amyloid Fibrils Through an NRF2 and Autophagy Dependent Pathway
- Author
-
Woodbury M, Benjamin J. Lang, Choudhury R, Ayesha Murshid, Stuart K. Calderwood, Thiago J Borges, Tsuneya Ikezu, Lay-Hong Ang, Yasmine S, Clark-Matott J, Doshi Sp, and Yuka Okusha
- Subjects
biology ,Microglia ,Amyloid beta ,Chemistry ,Autophagy ,Inflammation ,In vitro ,Cell biology ,medicine.anatomical_structure ,Heat shock protein ,biology.protein ,Extracellular ,medicine ,medicine.symptom ,Beta (finance) - Abstract
We have investigated the role of extracellular Heat shock protein 90alpha (eHsp90α) in conferring protection of neuronal cells against fibrillary amyloid beta (f-Aβ1-42) toxicity mediated by microglial cells. Formation of f-Aβ1-42 plaques leads to neurotoxic inflammation, a critical pathological feature of Alzheimer’s Disease. We observed increased uptake and clearance of internalized f-Aβ1-42 by microglial cells treated with eHsp90α, an effect associated with activation of NRF2 (NF-E2-related factor 2) - mediated autophagy. eHsp90α thus mitigated the neuronal toxicity of f-Aβ1-42-activated microglia. In addition, eHsp90α facilitated f-Aβ1-42 engulfment by microglial cells in vitro. In summary, eHsp90α triggers NRF2-mediated autophagy in microglia and thus protects against the neurotoxic effects of f-Aβ1-42.
- Published
- 2021
20. The 2021 FASEB Virtual Catalyst Conference on Extracellular and Organismal Proteostasis in Health and Disease, February 3-4, 2021
- Author
-
Chen Lior, Veena Prahlad, Stuart K. Calderwood, Evandro A. De-Souza, Dimitra Bourboulia, Maurizio Zanetti, Rebecca C. Taylor, Patricija van Oosten-Hawle, Mark R. Wilson, D. Allan Drummond, Francesca Hodge, Lea Sistonen, Ruth Scherz-Shouval, R. Luke Wiseman, Richard I. Morimoto, Adrienne L. Edkins, Oded Rechavi, and Della C. David
- Subjects
Chemistry ,Disease ,Congresses as Topic ,Biochemistry ,Immunity, Innate ,Article ,Cell biology ,Proteostasis ,immunology [Proteostasis Deficiencies] ,pathology [Proteostasis Deficiencies] ,Stress, Physiological ,ddc:570 ,Genetics ,Extracellular ,Animals ,Humans ,Proteostasis Deficiencies ,Molecular Biology ,Biotechnology - Published
- 2021
21. Arsenic hexoxide has differential effects on cell proliferation and genome-wide gene expression in human primary mammary epithelial and MCF7 cells
- Author
-
Ill Ju Bae, Donguk Kim, Stuart K. Calderwood, Na Yeon Park, Dong-Hyung Cho, Heeyoun Bunch, and Keunsoo Kang
- Subjects
Cell type ,Programmed cell death ,DNA repair ,Science ,Primary Cell Culture ,Gene Expression ,Apoptosis ,Breast Neoplasms ,Toxicology ,Arsenicals ,Article ,Arsenic ,Transcriptome ,Cell growth ,Breast cancer ,Arsenic Trioxide ,Tumor Cells, Cultured ,Humans ,Mammary Glands, Human ,Cell Proliferation ,Multidisciplinary ,Chemistry ,Cell Cycle ,Cell Differentiation ,Cell Cycle Checkpoints ,Genomics ,Cell cycle ,Drug regulation ,Cell biology ,Gene Expression Regulation, Neoplastic ,Cell culture ,Cancer cell ,MCF-7 Cells ,Medicine ,Female - Abstract
Arsenic is reportedly a biphasic inorganic compound for its toxicity and anticancer effects in humans [1, 2]. Recent studies have shown that certain arsenic compounds including arsenic hexoxide (AS4O6; hereafter, AS6) induce programmed cell death and cell cycle arrest in human cancer cells and murine cancer models [3, 4]. However, the mechanisms by which AS6 suppresses cancer cells are incompletely understood. In this study, we report the mechanisms of AS6 through transcriptome analyses. In particular, the cytotoxicity and global gene expression regulation by AS6 were compared in human normal and cancer breast epithelial cells. Using RNA-sequencing and bioinformatics analyses, differentially expressed genes in significantly affected biological pathways in these cell types were validated by real-time quantitative polymerase chain reaction and immunoblotting assays. Our data show markedly differential effects of AS6 on cytotoxicity and gene expression in human mammary epithelial normal cells (HUMEC) and Michigan Cancer Foundation 7 (MCF7), a human mammary epithelial cancer cell line. AS6 selectively arrests cell growth and induces cell death in MCF7 cells without affecting the growth of HUMEC in a dose-dependent manner. AS6 alters the transcription of a large number of genes in MCF7 cells, but much fewer genes in HUMEC. Importantly, we found that the cell proliferation, cell cycle, and DNA repair pathways are significantly suppressed whereas cellular stress response and apoptotic pathways increase in AS6-treated MCF7 cells. Together, we provide the first evidence of differential effects of AS6 on normal and cancerous breast epithelial cells, suggesting that AS6 at moderate concentrations induces cell cycle arrest and apoptosis through modulating genome-wide gene expression, leading to compromised DNA repair and increased genome instability selectively in human breast cancer cells.
- Published
- 2021
22. BRCA1-BARD1 regulates transcription through modulating topoisomerase IIβ
- Author
-
Benjamin P C Chen, Matthew J. Schellenberg, Heeyoun Bunch, Keunsoo Kang, Donguk Kim, Dong-Hyung Cho, Stuart K. Calderwood, Doo Sin Jo, Anh T. Q. Cong, and Jaehyeon Jeong
- Subjects
Regulation of gene expression ,chemistry.chemical_compound ,biology ,Chemistry ,DNA damage ,Transcription (biology) ,Topoisomerase ,Gene expression ,biology.protein ,RNA polymerase II ,Gene ,DNA ,Cell biology - Abstract
RNA polymerase II (Pol II)-dependent transcription in stimulus-inducible genes requires topoisomerase IIβ (TOP2B)-mediated DNA strand break and the activation of DNA damage response signaling in humans. Here, we report a novel function of the breast cancer 1 (BRCA1)-BRCA1 associated ring domain 1 (BARD1) complex, in this process. We found that BRCA1 is phosphorylated at S1524 by the kinases ATM and ATR during gene activation and that this event is essential for productive transcription. Our in vitro biochemical analyses showed TOP2B and BARD1 interaction and colocalization in the EGR1 transcription start site (TSS) and that the BRCA1-BARD1 complex ubiquitinates TOP2B, which appears to stabilize TOP2B protein in the cell and binding to DNA. Intriguingly, BRCA1 phosphorylation at S1524 controls this interaction. In addition, genomic analyses indicated colocalization between TOP2B and BRCA1 in a large number of protein-coding genes. Together, these findings reveal the novel function of the BRCA1-BARD1 complex in gene expression and in the regulation of TOP2B during Pol II transcription.Significance StatementMaintaining genomic integrity against cellular and extracellular genotoxic elements is essential for normal cell growth and function. Recent studies indicated that stimulus-induced transcription provokes topoisomerase IIβ-mediated DNA strand break and DNA damage response signaling, requiring DNA repair to be coupled with transcription. Here, we present a novel role for the BRCA1-BARD1 complex in regulating the transcription of serum-inducible genes and the stability of topoisomerase IIβ. The mechanism involving topoisomerase IIβ ubiquitination by the BRCA1-BARD1 complex and the phosphorylation of BRCA1 S1524 upon transcriptional activation appears to function as a switch to the reaction. Our findings provide the first evidence of functional interaction between the BRCA1-BARD1 complex and topoisomerase IIβ in transcription in humans.
- Published
- 2020
23. Regulation of a Novel Splice Variant of Early Growth Response 4 (EGR4-S) by HER+ Signalling and HSF1 in Breast Cancer
- Author
-
Jeremy M. Drake, Benjamin J. Lang, Martin Eduardo Guerrero-Gimenez, Jack Bolton, Christopher A. Dow, Stuart K. Calderwood, John T. Price, and Chau H. Nguyen
- Subjects
Cancer Research ,Oncology ,skin and connective tissue diseases ,EGR4 ,HSF1 ,HER2 ,breast cancer ,molecular stress - Abstract
The zinc finger transcription factor EGR4 has previously been identified as having a critical role in the proliferation of small cell lung cancer. Here, we have identified a novel, shortened splice variant of this transcription factor (EGR4-S) that is regulated by Heat Shock Factor-1 (HSF1). Our findings demonstrate that the shortened variant (EGR4-S) is upregulated with high EGFR, HER2, and H-Rasv12-expressing breast cell lines, and its expression is inhibited in response to HER pathway inhibitors. Protein and mRNA analyses of HER2+ human breast tumours indicated the novel EGR4-S splice variant to be preferentially expressed in tumour tissue and not detectable in patient-matched normal tissue. Knockdown of EGR4-S in the HER2-amplified breast cancer cell line SKBR3 reduced cell growth, suggesting that EGR4-S supports the growth of HER2+ tumour cells. In addition to chemical inhibitors of the HER2 pathway, EGR4-S expression was also found to be suppressed by chemical stressors and the overexpression of HSF1. Under these conditions, reduced EGR4-S levels were associated with the observed lower cell growth rate, but the augmentation of properties associated with higher metastatic potential. Taken together, these findings identify EGR4-S as a potential biomarker for HER2 pathway activation in human tumours that is regulated by HSF1.
- Published
- 2022
24. Chemogenomic screening identifies the Hsp70 co-chaperone DNAJA1 as a hub for anticancer drug resistance
- Author
-
Jade E. Takakuwa, Andrew W. Truman, Laura E. Knighton, Nitika, Jacob S. Blackman, and Stuart K. Calderwood
- Subjects
0301 basic medicine ,Carcinogenesis ,lcsh:Medicine ,Antineoplastic Agents ,Drug resistance ,medicine.disease_cause ,Article ,Chemical genetics ,Efficacy ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,Chaperones ,Tumor Cells, Cultured ,Humans ,Medicine ,HSP70 Heat-Shock Proteins ,Molecular Targeted Therapy ,Precision Medicine ,lcsh:Science ,Oncogene Proteins ,Multidisciplinary ,biology ,business.industry ,lcsh:R ,HSP40 Heat-Shock Proteins ,Precision medicine ,Co-chaperone ,030104 developmental biology ,Drug Resistance, Neoplasm ,Chaperone (protein) ,Cancer cell ,biology.protein ,Cancer research ,lcsh:Q ,Personalized medicine ,business ,030217 neurology & neurosurgery ,Molecular Chaperones - Abstract
Heat shock protein 70 (Hsp70) is an important molecular chaperone that regulates oncoprotein stability and tumorigenesis. However, attempts to develop anti-chaperone drugs targeting molecules such as Hsp70 have been hampered by toxicity issues. Hsp70 is regulated by a suite of co-chaperone molecules that bring “clients” to the primary chaperone for efficient folding. Rather than targeting Hsp70 itself, here we have examined the feasibility of inhibiting the Hsp70 co-chaperone DNAJA1 as a novel anticancer strategy. We found DNAJA1 to be upregulated in a variety of cancers, suggesting a role in malignancy. To confirm this role, we screened the NIH Approved Oncology collection for chemical-genetic interactions with loss of DNAJA1 in cancer. 41 compounds showed strong synergy with DNAJA1 loss, whereas 18 dramatically lost potency. Several hits were validated using a DNAJA1 inhibitor (116-9e) in castration-resistant prostate cancer cell (CRPC) and spheroid models. Taken together, these results confirm that DNAJA1 is a hub for anticancer drug resistance and that DNAJA1 inhibition is a potent strategy to sensitize cancer cells to current and future therapeutics. The large change in drug efficacy linked to DNAJA1 suggests a personalized medicine approach where tumor DNAJA1 status may be used to optimize therapeutic strategy.
- Published
- 2020
25. Extracellular Vesicles Enriched with Moonlighting Metalloproteinase Are Highly Transmissive, Pro-Tumorigenic, and Trans-Activates Cellular Communication Network Factor (
- Author
-
Yuka, Okusha, Takanori, Eguchi, Manh T, Tran, Chiharu, Sogawa, Kaya, Yoshida, Mami, Itagaki, Eman A, Taha, Kisho, Ono, Eriko, Aoyama, Hirohiko, Okamura, Ken-Ichi, Kozaki, Stuart K, Calderwood, Masaharu, Takigawa, and Kuniaki, Okamoto
- Subjects
body regions ,matrix metalloproteinase ,integumentary system ,CRISPR ,genome editing ,moonlighting metalloproteinase (MMP) ,oncosome ,cellular communication network factor ,protein moonlighting ,CCN2/CTGF ,extracellular vesicles ,Article ,transcription factor - Abstract
Matrix metalloproteinase 3 (MMP3) plays multiple roles in extracellular proteolysis as well as intracellular transcription, prompting a new definition of moonlighting metalloproteinase (MMP), according to a definition of protein moonlighting (or gene sharing), a phenomenon by which a protein can perform more than one function. Indeed, connective tissue growth factor (CTGF, aka cellular communication network factor 2 (CCN2)) is transcriptionally induced as well as cleaved by MMP3. Moreover, several members of the MMP family have been found within tumor-derived extracellular vesicles (EVs). We here investigated the roles of MMP3-rich EVs in tumor progression, molecular transmission, and gene regulation. EVs derived from a rapidly metastatic cancer cell line (LuM1) were enriched in MMP3 and a C-terminal half fragment of CCN2/CTGF. MMP3-rich, LuM1-derived EVs were disseminated to multiple organs through body fluid and were pro-tumorigenic in an allograft mouse model, which prompted us to define LuM1-EVs as oncosomes in the present study. Oncosome-derived MMP3 was transferred into recipient cell nuclei and thereby trans-activated the CCN2/CTGF promoter, and induced CCN2/CTGF production in vitro. TRENDIC and other cis-elements in the CCN2/CTGF promoter were essential for the oncosomal responsivity. The CRISPR/Cas9-mediated knockout of MMP3 showed significant anti-tumor effects such as the inhibition of migration and invasion of tumor cells, and a reduction in CCN2/CTGF promoter activity and fragmentations in vitro. A high expression level of MMP3 or CCN2/CTGF mRNA was prognostic and unfavorable in particular types of cancers including head and neck, lung, pancreatic, cervical, stomach, and urothelial cancers. These data newly demonstrate that oncogenic EVs-derived MMP is a transmissive trans-activator for the cellular communication network gene and promotes tumorigenesis at distant sites.
- Published
- 2020
26. Extracellular Oncosomes Rich in Moonlighting Metalloproteinase (MMP3) Are Transmissive, Pro-Tumorigenic, and Induces Cellular Communication Network Factor 2 (CCN2/CTGF): CRISPR against Cancer
- Author
-
Stuart K. Calderwood, Kisho Ono, Yuka Okusha, Mami Itagaki, Manh Tien Tran, Ken Ichi Kozaki, Hirohiko Okamura, Eman A. Taha, Kuniaki Okamoto, Takanori Eguchi, Eriko Aoyama, Chiharu Sogawa, Masaharu Takigawa, and Kaya Yoshida
- Subjects
Metalloproteinase ,MMP3 ,integumentary system ,Cancer ,Biology ,medicine.disease ,Cell biology ,body regions ,Genome editing ,Ccn2 ctgf ,Extracellular ,medicine ,CRISPR ,Transcription factor ,oncology_oncogenics - Abstract
Matrix metalloproteinase 3 (MMP3) plays multiple roles in pro-tumorigenic proteolysis and in intracellular transcription. These include inducing connective tissue growth factor [CTGF, also known as cellular communication network factor 2 (CCN2)] and prompting a new definition of MMP3 as a moonlighting metalloproteinase. Members of the MMP family have been found within tumor-derived extracellular vesicles (EVs) such as oncosomes or exosomes. We here investigated the roles of MMP3-rich oncosomes in tumor progression, molecular transmission, and gene regulation. MMP3 and CCN2/CTGF were significantly co-expressed in tumor samples derived from patients suffering from colorectal adenocarcinoma. We found that oncosomes derived from a rapidly metastatic colon cancer cells (LuM1) were enriched in MMP3 and a C-terminal half fragment of CCN2/CTGF. MMP3-rich oncosomes were highly transmissive into recipient cells and were pro-tumorigenic in an allograft mouse model. Oncosome-derived MMP3 was transmissive into recipient cell nuclei, trans-activated CCN2/CTGF promoter, and induced CCN2/CTGF production at 1 to 6 hours after the addition of oncosomes to culture media. In addition, CRISPR/Cas9-mediated knockout of MMP3 showed significant anti-tumor effects, including inhibition of migration and invasion of LuM1 cells in vitro, inhibition of tumor growth in vivo, and reduction of CCN2/CTGF and its promoter activity in vitro. These data newly demonstrate that the oncosome-derived moonlighting metalloproteinase promotes metastasis and is pro-tumorigenic at distant sites as well as a transmissive trans-activator for the cellular communication network gene.
- Published
- 2020
27. Antiparkinson Drug Benztropine Suppresses Tumor Growth, Circulating Tumor Cells, and Metastasis by Acting on SLC6A3 and Reducing STAT3
- Author
-
Hotaka Kawai, Masayuki Ishige, Yanyin Lu, Manh Tien Tran, Yuka Okusha, Kilian Trin, Stuart K. Calderwood, Kuniaki Okamoto, Ken Ichi Kozaki, Takanori Eguchi, Norio Sogawa, Eman A. Taha, Masaharu Takigawa, and Chiharu Sogawa
- Subjects
biology ,business.industry ,medicine.disease ,Benztropine ,pharmacology_toxicology ,Metastasis ,Antiparkinson drug ,Circulating tumor cell ,medicine ,biology.protein ,Cancer research ,Tumor growth ,business ,STAT3 - Abstract
Tumor growth, progression, and therapy resistance are crucial factors in the prognosis of cancer. Properties of three-dimensional tumor-like organoids (tumoroids) more closely resemble in vivo tumors compared to two-dimensionally cultured cells and are therefore effectively used for assays and drug screening. We here established a repurposed drug for novel anticancer research and therapeutics using a tumoroid-based screening system. We screened 6 pharmacologically active compounds by using an original tumoroid-based multiplex phenotypic screening system with matrix metalloproteinase 9 (MMP9) promoter-driven fluorescence reporter for the evaluation of both tumoroid formation and progression. The effects of one of the hit compounds were examined on tumor formation and progression in vitro and in vivo. Antiparkinson drug benztropine was the most effective compound uncovered by the screen. Benztropine significantly inhibited in vitro tumoroid formation, cancer cell survival, and MMP9 promoter activity. Benztropine also reduced the activity of oncogenic signaling transducers and trans-activators for MMP9, including STAT3, NF-κB, and β-catenin, and properties of cancer stem cells / cancer-initiating cells. Benztropine and GBR-12935 directly targeted the dopamine transporter DAT/SLC6A3, whose genetic alterations such as amplification were correlated with poor prognosis for cancer patients. Benztropine also inhibited tumor growth, circulating tumor cell (CTC) number, and rate of metastasis in a tumor allograft model in mice. In conclusion, we propose the repurposing of benztropine for anticancer research and therapeutics that can suppress tumor progression, CTC, and metastasis of aggressive cancers by reducing key pro-tumorigenic factors.
- Published
- 2020
28. Immunological Outcomes Mediated Upon Binding of Heat Shock Proteins to Scavenger Receptors SCARF1 and LOX-1, and Endocytosis by Mononuclear Phagocytes
- Author
-
Ayesha Murshid, Thiago J. Borges, Cristina Bonorino, Benjamin J. Lang, and Stuart K. Calderwood
- Subjects
lcsh:Immunologic diseases. Allergy ,0301 basic medicine ,Immunology ,Review ,macrophage ,scavenger receptor ,03 medical and health sciences ,0302 clinical medicine ,Heat shock protein ,Animals ,Humans ,Immunology and Allergy ,Scavenger receptor ,Receptor ,Heat-Shock Proteins ,Scavenger Receptor Class F Member 1 ,Receptors, Scavenger ,Phagocytes ,Innate immune system ,Chemistry ,Antigen processing ,Scavenger Receptors, Class E ,Acquired immune system ,immunity ,Endocytosis ,Scavenger Receptors, Class F ,Cell biology ,030104 developmental biology ,heat shock proteins ,dendritic ,Signal transduction ,lcsh:RC581-607 ,030215 immunology - Abstract
Heat shock proteins (HSP) are a highly abundant class of molecular chaperones that can be released into the extracellular milieu and influence the immune response. HSP release can occur when cells undergo necrosis and exude their contents. However, HSPs are also secreted from intact cells, either in free form or in lipid vesicles including exosomes to react with receptors on adjacent cells. Target cells are able recognize extracellular HSPs through cell surface receptors. These include scavenger receptors (SR) such as class E member oxidized low-density lipoprotein receptor-1 (LOX-1, aka OLR1, Clec8A, and SR-E1) and scavenger receptor class F member 1 (SCARF1, aka SREC1). Both receptors are expressed by dendritic cells (DC) and macrophages. These receptors can bind HSPs coupled to client binding proteins and deliver the chaperone substrate to the pathways of antigen processing in cells. SR are able to facilitate the delivery of client proteins to the proteasome, leading to antigen processing and presentation, and stimulation of adaptive immunity. HSPs may also may be involved in innate immunity through activation of inflammatory signaling pathways in a mechanism dependent on SR and toll-like receptor 4 (TLR4) on DC and macrophages. We will discuss the pathways by which HSPs can facilitate uptake of protein antigens and the receptors that regulate the ensuing immune response.
- Published
- 2020
29. Anti-EGFR antibody cetuximab is secreted by oral squamous cell carcinoma and alters EGF-driven mesenchymal transition
- Author
-
Kisho Ono, Ken ichi Kozaki, Jun Ichi Asaumi, Chiharu Sogawa, Stuart K. Calderwood, Toshifumi Fujiwara, Takanori Eguchi, Kuniaki Okamoto, Jun Murakami, and Soichiro Ibaragi
- Subjects
0301 basic medicine ,Epithelial-Mesenchymal Transition ,Biophysics ,Cetuximab ,Stimulation ,Antibodies, Monoclonal, Humanized ,Biochemistry ,Extracellular Vesicles ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Humans ,Secretion ,Epithelial–mesenchymal transition ,Epidermal growth factor receptor ,neoplasms ,Molecular Biology ,Anti-EGFR antibody therapy ,Epidermal Growth Factor ,biology ,Transition (genetics) ,business.industry ,Mesenchymal stem cell ,Head and neck squamous cell carcinoma ,Cell Biology ,medicine.disease ,Head and neck squamous-cell carcinoma ,digestive system diseases ,ErbB Receptors ,stomatognathic diseases ,030104 developmental biology ,Epithelial-to-mesenchymal transition ,Drug Resistance, Neoplasm ,Carcinoma, Squamous Cell ,Cancer research ,biology.protein ,Mouth Neoplasms ,business ,medicine.drug - Abstract
Genetic amplification, overexpression, and increased signaling from the epidermal growth factor receptor (EGFR) are often found in oral squamous cell carcinoma (OSCC) and thus EGFR is frequently targeted molecularly by the therapeutic antibody cetuximab. We assessed effects of cetuximab in control of EGF-driven malignant traits of OSCC cells. EGF stimulation promoted progression level of mesenchymal traits in OSCC cells, which were attenuated by cetuximab but incompletely. We pursued a potential mechanism underlying such incomplete attenuation of OSCC malignant traits. Cetuximab promoted secretion of EGFR-EVs by OSCC cells and failed to inhibit EGF-driven secretion of EGFR-EVs. Cetuximab was also found to be robustly secreted with the EGFR-EVs by the OSCC cells. Thus, EGF promotes the level of mesenchymal traits of OSCC cells and secretion of EGFR-EVs, which involve cetuximab resistance.
- Published
- 2018
30. Exosome Release of Drugs: Coupling with Epithelial-Mesenchymal Transition
- Author
-
Stuart K. Calderwood, Kisho Ono, Takanori Eguchi, and Kuniaki Okamoto
- Subjects
Coupling (electronics) ,Chemistry ,medicine ,Epithelial–mesenchymal transition ,Hypoxia (medical) ,medicine.symptom ,Exosome ,Tumor immunology ,pharmacology_toxicology ,Cell biology - Abstract
Extracellular vesicles (EVs), such as exosomes or oncosomes are released with molecules unfavorable for survival from cells. In addition, accumulating evidence has shown that tumor cells often eject anti-cancer drugs such as chemotherapeutics and targeted drugs within EVs, a novel mechanism of drug resistance. The EV-releasing, drug resistance phenotype is often coupled with cellular dedifferentiation and transformation, cells undergoing epithelial-mesenchymal transition (EMT) and taking on a cancer stem cell phenotype. Recent studies have shown that the release of EVs is also involved in immunosuppression. The concept of the resistance-associated secretory phenotype (RASP) is reviewed herein.
- Published
- 2019
31. Abstract 2158: Arsenic hexoxide has differential effects on cell proliferation and genome-wide gene expression in human primary mammary epithelial and mcf7 cells
- Author
-
Donguk Kim, Na Yeon Park, Keunsoo Kang, Jaehyeon Jeong, Dong-Hyung Cho, Heeyoun Bunch, Stuart K. Calderwood, and Ill Ju Bae
- Subjects
Cancer Research ,Primary (chemistry) ,Oncology ,chemistry ,Cell growth ,Gene expression ,chemistry.chemical_element ,Biology ,Differential effects ,Genome ,Arsenic ,MCF7 Cells ,Cell biology - Abstract
Arsenic is reportedly a biphasic inorganic compound for its toxicity and anticancer effects in humans. Recent studies have shown that certain arsenic compounds including arsenic hexoxide (AS4O6; hereafter, AS6) induce programmed cell death and cell cycle arrest in human cancer cells and murine cancer models. However, the mechanisms by which AS6 suppresses cancer cells are incompletely understood. In this study, we report the mechanisms of AS6 through transcriptome analyses. In particular, the cytotoxicity and global gene expression regulation by AS6 were compared in human normal and cancer breast epithelial cells. Using RNA-sequencing and bioinformatics analyses, differentially expressed genes in significantly affected biological pathways in these cell types were validated by real-time quantitative polymerase chain reaction and immunoblotting assays. Our data show markedly differential effects of AS6 on cytotoxicity and gene expression in human mammary epithelial normal cells (HUMEC) and Michigan Cancer Foundation 7 (MCF7), a human mammary epithelial cancer cell line. AS6 selectively arrests cell growth and induces cell death in MCF7 cells without affecting the growth of HUMEC in a dose-dependent manner. AS6 alters the transcription of a large number of genes in MCF7 cells, but much fewer genes in HUMEC. Importantly, we found that the cell proliferation, cell cycle, and DNA repair pathways are significantly suppressed whereas cellular stress response and apoptotic pathways increase in AS6-treated MCF7 cells. Together, we provide the first evidence of differential effects of AS6 on normal and cancerous breast epithelial cells, suggesting that AS6 at moderate concentrations induces cell cycle arrest and apoptosis through modulating genome-wide gene expression, leading to compromised DNA repair and increased genome instability selectively in human breast cancer cells. Citation Format: Heeyoun Bunch, Donguk Kim, Na Yeon Park, Jaehyeon Jeong, Keunsoo Kang, Stuart K. Calderwood, Dong-Hyung Cho, Ill Ju Bae. Arsenic hexoxide has differential effects on cell proliferation and genome-wide gene expression in human primary mammary epithelial and mcf7 cells [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 2158.
- Published
- 2021
32. Exploring the nexus of Alzheimer's disease and related dementias with cancer and cancer therapies: A convening of the Alzheimer's Association & Alzheimer's Drug Discovery Foundation
- Author
-
Honglei Chen, Suzanne Craft, Diana W Shineman, Jane A. Driver, Suzana Petanceska, Tim A. Ahles, Mary Ganguli, Howard Fillit, Maria C. Carrillo, Heather M. Snyder, Andrew J. Saykin, Sudha Seshadri, Chung-Chou H. Chang, David S. Knopman, Mary C. Tierney, Stuart K. Calderwood, Philip L. De Jager, and Michael T. Lotze
- Subjects
Gerontology ,Epidemiology ,Population ,Disease ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Developmental Neuroscience ,medicine ,Dementia ,education ,education.field_of_study ,business.industry ,Drug discovery ,Health Policy ,Foundation (evidence) ,Cancer ,medicine.disease ,Psychiatry and Mental health ,030220 oncology & carcinogenesis ,Neurology (clinical) ,Geriatrics and Gerontology ,Alzheimer's disease ,business ,Neuroscience ,Nexus (standard) ,030217 neurology & neurosurgery - Abstract
Recent population studies suggest an intriguing inverse relationship between several types of cancer and neurodegenerative diseases, including Alzheimer's disease. Understanding the intersection of the underlying biology for these two distinct families of diseases with one another may offer novel approaches to identify new therapeutic approaches and possible opportunities to repurpose existing drug candidates. The Alzheimer's Association and the Alzheimer's Drug Discovery Foundation convened a one-day workshop to delve into this discussion. Workshop participants outlined research focus areas, potential collaborations, and partnerships for future action.
- Published
- 2016
33. SCAND1 Suppresses CDC37 Gene Transcription by Repressing MZF1
- Author
-
Takanori Eguchi, Thomas L. Prince, Tien Manh Tran, Chiharu Sogawa, Benjamin J. Lang, and Stuart K. Calderwood
- Subjects
3. Good health - Abstract
Cell division control 37 (CDC37) increases the stability of HSP90 client proteins and is thus essential for numerous intracellular oncogenic signaling pathways, playing a key role in prostate oncogenesis. Notably, elevated expression of CDC37 was found in prostate cancer cells, although the regulatory mechanisms through which CDC37 expression becomes increased are unknown. Here we show both positive and negative regulation of CDC37 gene transcription by two members of the SCAN transcription factor family- MZF1 and SCAND1, respectively. Consensus DNA-binding motifs for myeloid zinc finger 1 (MZF1 / ZSCAN6) were abundant in the CDC37 promoter region. MZF1 became bound to these regulatory sites and trans-activated the CDC37 gene whereas MZF1 depletion decreased CDC37 transcription and reduced tumorigenesis of prostate cancer cells. On the other hand, SCAND1, a zinc-fingerless SCAN box protein that potentially inhibits MZF1, accumulated at MZF1-binding sites in CDC37 gene, negatively regulated CDC37 gene and inhibited tumorigenesis. SCAND1 was abundantly expressed in normal prostate cells but was reduced in prostate cancer cells, suggesting a potential tumor suppressor role of SCAND1 in prostate cancer. These findings indicate that CDC37, a crucial protein in prostate cancer progression, is regulated reciprocally by MZF1 and SCAND1.
- Published
- 2019
34. Regulatory Roles of HSP90-Rich Extracellular Vesicles
- Author
-
Kuniaki Okamoto, Takanori Eguchi, Kazumi Kawata, Stuart K. Calderwood, and Kisho Ono
- Subjects
biology ,Chemistry ,polycyclic compounds ,Extracellular ,biology.protein ,Secretion ,Rab ,Extracellular vesicle ,Exosome ,LRP1 ,Microvesicles ,Receptor tyrosine kinase ,Cell biology - Abstract
HSP90 is an essential protein in protein folding, cancer progression and wound healing. Originally, most studies were focused on the intracellular molecular chaperone role of HSP90. However, more recent studies, including ours, have reported the secretion of HSP90 and novel functions for this protein in the extracellular space (ex-HSP90). Additionally, HSP90 has been found to be a major cargo contained in extracellular vesicles (EV) such as exosomes. HSP90 can directly bind to and promote functions of CD91/LRP1 and receptor tyrosine kinases such as EGF receptor. HSP90 also regulates the recycling of Rab proteins that control the secretion of exosomes. This chapter reviews current knowledge and the future potential of ex-HSP90 and EV-HSP90.
- Published
- 2019
35. BRCA1-BARD1 Regulates Topoisomerase IIβ Stability in Transcription
- Author
-
Dong-Hyung Cho, Benjamin P C Chen, Sanghwa Lee, Heeyoun Bunch, Doo Sin Jo, Stuart K. Calderwood, and Keunsoo Kang
- Subjects
Genome instability ,Regulation of gene expression ,biology ,Transcription (biology) ,DNA damage ,Chemistry ,DNA repair ,Topoisomerase ,biology.protein ,RNA polymerase II ,skin and connective tissue diseases ,Gene ,Cell biology - Abstract
RNA polymerase II (Pol II) dependent transcription in stimulus-inducible genes requires topoisomerase IIβ-mediated DNA strand break and the activation of DNA damage response signaling. Here we report a novel function of DNA repair proteins, involving a breast cancer 1 (BRCA1)-BRCA1 associated RING domain 1 (BARD1) complex, in transcriptional activation of stimulus-inducible genes in humans. We have found that BRCA1 is phosphorylated at S1524 by the kinases ATM and ATR during gene activation and that this phosphorylation event is essential for productive transcription. The major TOPIIβ binding site in the EGR1 TSS was mapped in the promoter between −132 and −14. We showed that BARD1 interacts with and could ubiquitinate TOPIIβ, and that BRCA1 regulates this interaction. Together, our findings indicate that the BRCA1-BARD1 complex controls TOPIIβ stability and gene association during Pol II transcription.
- Published
- 2019
36. Intracellular MMP3 PromotesHSPGene Expression in Collaboration With Chromobox Proteins
- Author
-
Satoshi Kubota, Ken Ichi Kozaki, Stuart K. Calderwood, Takanori Eguchi, and Masaharu Takigawa
- Subjects
0301 basic medicine ,Reporter gene ,Promoter ,Cell Biology ,Biology ,Biochemistry ,Molecular biology ,body regions ,03 medical and health sciences ,030104 developmental biology ,Heat shock protein ,Gene expression ,Heterochromatin protein 1 ,HSF1 ,Molecular Biology ,Gene ,Transcription factor - Abstract
Matrix metalloproteinases (MMPs) are crucial factors in tumor progression, inflammatory/immune responses and tissue development/regeneration. Of note, it has been known that MMPs promote genome instability, epithelial-mesenchymal transition, invasion, and metastasis in tumor progression. We previously reported that human MMP3 could translocate into cellular nuclei and control transcription in human chondrosarcoma-derived cells and in articular cartilage (Eguchi et al. [2008] Mol Cell Biol 28(7):2391-2413); however, further transcriptional target genes and cofactors of intranuclear MMP3 have not been uncovered. In this paper, we used transcriptomics analysis in order to examine novel transcriptional target genes regulated by intracellular MMP3. We found that mRNA levels of HSP family members (HSP70B', HSP72, HSP40/DNAJ, and HSP20/CRYAB) are upregulated by the intracellular MMP3 overload. Bioinformatic analysis predicted several transcription factors that possibly interact with MMP3. Among these factors, heat shock factor 1 (HSF1) cooperated with the MMP3 to activate the HSP70B' gene promoter in reporter gene assays, while a dominant negative HSF1 blocked the role for MMP3 in the trans-activation. The hemopexin-like repeat (PEX) domain of the human MMP3 was essential for transcriptional induction of the HSP70B' gene. In addition, chromobox proteins CBX5/HP1α and CBX3/HP1γ cooperated with the PEX domain in induction of HSP70B' mRNA. Taken together, this study newly clarified that intracellular MMP3 cooperate with CBXs/HP1s in transcriptional promotion of HSP genes.
- Published
- 2016
37. A critical role for topoisomerase IIb and DNA double strand breaks in transcription
- Author
-
Stuart K. Calderwood
- Subjects
0301 basic medicine ,Transcription, Genetic ,DNA repair ,DNA damage ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Transcription (biology) ,Genetics ,DNA Breaks, Double-Stranded ,Point-of-View ,Polymerase ,Transcription bubble ,biology ,Chemistry ,Topoisomerase ,DNA ,Molecular biology ,DNA-Binding Proteins ,DNA Repair Enzymes ,DNA Topoisomerases, Type II ,030104 developmental biology ,biology.protein ,DNA supercoil ,030217 neurology & neurosurgery ,Biotechnology - Abstract
Recent studies have indicated a novel role for topoisomerase IIb in transcription. Transcription of heat shock genes, serum-induced immediate early genes and nuclear receptor-activated genes, each required DNA double strands generated by topoisomerase IIb. Such strand breaks seemed both necessary and sufficient for transcriptional activation. In addition, such transcription was associated with initiation of the DNA damage response pathways, including the activation of the enzymes: ataxia-telangiectasia mutated (ATM), DNA-dependent protein kinase and poly (ADP ribose) polymerase 1. DNA damage response signaling was involved both in transcription and in repair of DNA breaks generated by topoisomerase IIb.
- Published
- 2016
38. Triple knockdown of CDC37, HSP90‐alpha and HSP90‐beta diminishes extracellular vesicles‐driven malignancy events and macrophage M2 polarization in oral cancer
- Author
-
Hitoshi Nagatsuka, Kisho Ono, Soichiro Ibaragi, Ken Ichi Kozaki, Chiharu Sogawa, Masaharu Takigawa, Yanyin Lu, May Wathone Oo, Hotaka Kawai, Kuniaki Okamoto, Akira Sasaki, Manh Tien Tran, Yuka Okusha, Eman A. Taha, Stuart K. Calderwood, Hirohiko Okamura, and Takanori Eguchi
- Subjects
0301 basic medicine ,Histology ,tumour-associated macrophage ,epithelial-mesenchymal transition ,tumour‐associated macrophage ,cdc37 ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Epithelial–mesenchymal transition ,lcsh:QH573-671 ,Gene knockdown ,epithelial‐mesenchymal transition ,biology ,lcsh:Cytology ,Chemistry ,Cancer ,Cell Biology ,oral cancer ,medicine.disease ,Hsp90 ,hsp90 ,Microvesicles ,tetraspanin ,030104 developmental biology ,CDC37 ,030220 oncology & carcinogenesis ,Chaperone (protein) ,Cancer cell ,Cancer research ,biology.protein ,extracellular vesicles ,Research Article - Abstract
Evidence has been accumulating to indicate that extracellular vesicles (EVs), including exosomes, released by cancer cells can foster tumour progression. The molecular chaperones – CDC37, HSP90α and HSP90β play key roles in cancer progression including epithelial-mesenchymal transition (EMT), although their contribution to EVs-mediated cell–cell communication in tumour microenvironment has not been thoroughly examined. Here we show that triple depletion of the chaperone trio attenuates numerous cancer malignancy events exerted through EV release. Metastatic oral cancer-derived EVs (MEV) were enriched with HSP90α HSP90β and cancer-initiating cell marker CD326/EpCAM. Depletion of these chaperones individually induced compensatory increases in the other chaperones, whereas triple siRNA targeting of these molecules markedly diminished the levels of the chaperone trio and attenuated EMT. MEV were potent agents in initiating EMT in normal epithelial cells, a process that was attenuated by the triple chaperone depletion. The migration, invasion, and in vitro tumour initiation of oral cancer cells were significantly promoted by MEV, while triple depletion of CDC37/HSP90α/β reversed these MEV-driven malignancy events. In metastatic oral cancer patient-derived tumours, HSP90β was significantly accumulated in infiltrating tumour-associated macrophages (TAM) as compared to lower grade oral cancer cases. HSP90-enriched MEV-induced TAM polarization to an M2 phenotype, a transition known to support cancer progression, whereas the triple chaperone depletion attenuated this effect. Mechanistically, the triple chaperone depletion in metastatic oral cancer cells effectively reduced MEV transmission into macrophages. Hence, siRNA-mediated knockdown of the chaperone trio (CDC37/HSP90α/HSP90β) could potentially be a novel therapeutic strategy to attenuate several EV-driven malignancy events in the tumour microenvironment. Abbreviations CDC37: cell division control 37; EMT: epithelial-mesenchymal transmission; EV: extracellular vesicles; HNSCC: head and neck squamous cell carcinoma; HSP90: heat shock protein 90; TAM: tumour-associated macrophage
- Published
- 2020
39. HSF1: Primary Factor in Molecular Chaperone Expression and a Major Contributor to Cancer Morbidity
- Author
-
Stuart K. Calderwood, Martin E. Guerrero-Gimenez, Thomas Prince, Andrew Ackerman, Benjamin J. Lang, and Juan Manuel Fernandez-Muñoz
- Subjects
HEAT SHOCK PROTEINS ,Review ,medicine.disease_cause ,HSF1 expression in cancer ,purl.org/becyt/ford/1 [https] ,HSF1 IN CANCER ,Heat Shock Transcription Factors ,Neoplasms ,Molecular Targeted Therapy ,HSR ,HSF1 ,lcsh:QH301-705.5 ,Regulation of gene expression ,HSPS ,molecular chaperones ,CANCER THERAPY ,General Medicine ,CANCER ,Cell Transformation, Neoplastic ,heat shock factor 1 ,MOLECULAR CHAPERONES ,HSF1 in cancer ,HSF1 EXPRESSION IN CANCER ,cancer therapy ,HEAT SHOCK RESPONSE ,HSPs ,Biology ,heat shock response ,Heat shock protein ,TUMORIGENESIS ,medicine ,Animals ,Humans ,metastasis ,cancer ,Heat shock ,purl.org/becyt/ford/1.6 [https] ,fungi ,Cancer ,medicine.disease ,HEAT SHOCK FACTOR 1 ,tumorigenesis ,Proteostasis ,lcsh:Biology (General) ,METASTASIS ,heat shock proteins ,Cancer cell ,Cancer research ,Morbidity ,Carcinogenesis - Abstract
Heat shock factor 1 (HSF1) is the primary component for initiation of the powerful heat shock response (HSR) in eukaryotes. The HSR is an evolutionarily conserved mechanism for responding to proteotoxic stress and involves the rapid expression of heat shock protein (HSP) molecular chaperones that promote cell viability by facilitating proteostasis. HSF1 activity is amplified in many tumor contexts in a manner that resembles a chronic state of stress, characterized by high levels of HSP gene expression as well as HSF1-mediated non-HSP gene regulation. HSF1 and its gene targets are essential for tumorigenesis across several experimental tumor models, and facilitate metastatic and resistant properties within cancer cells. Recent studies have suggested the significant potential of HSF1 as a therapeutic target and have motivated research efforts to understand the mechanisms of HSF1 regulation and develop methods for pharmacological intervention. We review what is currently known regarding the contribution of HSF1 activity to cancer pathology, its regulation and expression across human cancers, and strategies to target HSF1 for cancer therapy. Fil: Prince, Thomas L.. Geisinger Clinic. Department of Molecular Functional Genomics; Estados Unidos Fil: Lang, Benjamin J.. Harvard Medical School; Estados Unidos Fil: Guerrero Gimenez, Martin Eduardo. Consejo Nacional de Investigaciones Científicas y Técnicas. Centro Científico Tecnológico Conicet - Mendoza. Instituto de Medicina y Biología Experimental de Cuyo; Argentina Fil: Fernandez Muñoz, Juan Manuel. Consejo Nacional de Investigaciones Científicas y Técnicas. Centro Científico Tecnológico Conicet - Mendoza. Instituto de Medicina y Biología Experimental de Cuyo; Argentina Fil: Ackerman, Andrew. Geisinger Clinic. Department of Molecular Functional Genomics; Estados Unidos Fil: Calderwood, Stuart K.. Harvard Medical School; Estados Unidos
- Published
- 2020
40. Carcinogenic epithelial-mesenchymal transition initiated by oral cancer exosomes is inhibited by anti-EGFR antibody cetuximab
- Author
-
Jun Ichi Asaumi, Toshifumi Fujiwara, Stuart K. Calderwood, Kisho Ono, Ken ichi Kozaki, Takanori Eguchi, Kuniaki Okamoto, Jun Murakami, Soichiro Ibaragi, and Chiharu Sogawa
- Subjects
0301 basic medicine ,Cancer Research ,Epithelial-Mesenchymal Transition ,Cetuximab ,Vimentin ,Exosomes ,Exosome ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Humans ,Epithelial–mesenchymal transition ,Epidermal growth factor receptor ,biology ,Epidermal Growth Factor ,Chemistry ,Squamous Cell Carcinoma of Head and Neck ,Head and neck cancer ,Mesenchymal stem cell ,Epithelial Cells ,Extracellular vesicle ,medicine.disease ,ErbB Receptors ,stomatognathic diseases ,030104 developmental biology ,Cell Transformation, Neoplastic ,Oncology ,Cancer research ,biology.protein ,Mouth Neoplasms ,Oral Surgery ,medicine.drug - Abstract
Overexpression and increased signaling from the epidermal growth factor receptor (EGFR) often changes oral squamous cell carcinoma (OSCC) and thus EGFR is frequently targeted molecularly by the therapeutic antibody cetuximab. We assessed the roles of OSCC-derived extracellular vesicles (EVs), including exosomes in the trafficking of cetuximab and in epithelial-mesenchymal transition (EMT) of epithelial cells. OSCC cells abundantly expressed EGFR, which was secreted from cells with OSCC-EVs upon EGF stimulations. The OSCC-EGFR-EVs were then able to enter into and transform epithelial cells leading to increased mesenchymal traits with increased vimentin and spindle-like shapes. EGF priming of OSCC cells further increased this EMT-initiating effect of the OSCC-EVs. The internalization and pro-EMT effects of the OSCC-EVs were largely blocked by cetuximab. Thus, OSCC-derived EVs transform normal epithelial cells into a mesenchymal phenotype and anti-EGFR therapeutic antibody cetuximab inhibits such a carcinogenic effect of the OSCC-EVs.
- Published
- 2018
41. Regulatory Roles for Hsp70 in Cancer Incidence and Tumor Progression
- Author
-
Taka Eguchi, Benjamin J. Lang, Ayesha Murshid, Thomas Prince, Jianlin Gong, and Stuart K Calderwood
- Published
- 2018
42. Emerging roles for scavenger receptor SREC-I in immunity
- Author
-
Stuart K. Calderwood, Ayesha Murshid, and Thiago J. Borges
- Subjects
Cell signaling ,T-Lymphocytes ,Immunology ,Apoptosis ,chemical and pharmacologic phenomena ,Biochemistry ,Article ,Cross-Priming ,Immune system ,Phagocytosis ,MHC class I ,Humans ,Immunology and Allergy ,HSP70 Heat-Shock Proteins ,Scavenger receptor ,Antigen-presenting cell ,Molecular Biology ,Antigen Presentation ,Innate immune system ,biology ,Macrophages ,Toll-Like Receptors ,Cross-presentation ,Dendritic Cells ,Hematology ,Acquired immune system ,Endocytosis ,Scavenger Receptors, Class F ,Cell biology ,biology.protein ,Signal Transduction - Abstract
SREC-I is a class F scavenger receptor with key role in the immune response, particularly in antigen presenting cell (APC) such as macrophages and dendritic cells (DC). This receptor is able to mediate engulfment of dead cells as well as endocytosis of heat shock protein (HSP)-antigen complexes. SREC-I could thus potentially mediate the tolerizing influence of apoptotic cells or the immunostimulatory effects of HSP-peptide complexes, depending on context. This receptor was able to mediate presentation of external antigens, bound to HSPs through both the class II pathway as well as cross presentation via MHC class I complexes. In addition to its recently established role in adaptive immunity, emerging studies are indicating a broad role in innate immunity and regulation of cell signaling through Toll Like Receptors (TLR). SREC-I may thus play a key role in APC function by coordinating immune responses to internal and external antigens in APC.
- Published
- 2015
43. Role and Regulation of Myeloid Zinc Finger Protein 1 in Cancer
- Author
-
Stuart K. Calderwood, Thomas Prince, Taka Eguchi, and Barbara Wegiel
- Subjects
Myeloid ,Colorectal cancer ,SUMO protein ,Myeloid zinc finger 1 ,Cancer ,Cell Biology ,Biology ,medicine.disease ,Biochemistry ,Leukemia ,medicine.anatomical_structure ,Immunology ,medicine ,Cancer research ,Phosphorylation ,Molecular Biology ,Transcription factor - Abstract
Myeloid zinc finger 1 (MZF1) belongs to the SCAN-Zinc Finger (SCAN-ZF) transcription factor family that has recently been implicated in a number of types of cancer. Although the initial studies concentrated on the role of MZF1 in myeloid differentiation and leukemia, the factor now appears to be involved in the etiology of major solid tumors such as lung, cervical, breast, and colorectal cancer. Here we discuss the regulation of MZF1 that mediated its recruitment and activation in cancer, concentrating on posttranslational modification by phosphorylation, and sumoylation, formation of promyelocytic leukemia nuclear bodies and its association with co-activators and co-repressors.
- Published
- 2015
44. Targeting the hsp70 gene delays mammary tumor initiation and inhibits tumor cell metastasis
- Author
-
Michael Y. Sherman, Ayesha Murshid, Stuart K. Calderwood, Takanori Eguchi, Desheng Weng, Jianlin Gong, and Baizheng Song
- Subjects
Cancer Research ,Down-Regulation ,Antineoplastic Agents ,HSP72 Heat-Shock Proteins ,Tumor initiation ,medicine.disease_cause ,Article ,Metastasis ,Mice ,Cell Movement ,Genetics ,medicine ,Animals ,Neoplasm Metastasis ,Molecular Biology ,Mice, Knockout ,Mammary tumor ,biology ,Oncogene ,Mouse mammary tumor virus ,CD44 ,Mammary Neoplasms, Experimental ,Oncogenes ,Proto-Oncogene Proteins c-met ,Cell cycle ,biology.organism_classification ,medicine.disease ,Mice, Inbred C57BL ,Cell Transformation, Neoplastic ,biology.protein ,Cancer research ,Female ,Carcinogenesis - Abstract
Elevated levels of the inducible heat shock protein 70 (Hsp72) have been implicated in mammary tumorigenesis in histological investigations of human breast cancer. We therefore examined the role of Hsp72 in mice, using animals in which the hsp70 gene was inactivated. We used a spontaneous tumor system with mice expressing the polyomavirus middle T (PyMT) oncogene under control of the mouse mammary tumor virus (MMTV) long terminal repeat (MMT mice). These mice developed spontaneous, metastatic mammary cancer. We then showed Hsp72 to be upregulated in a fraction of mammary cancer initiating cells (CIC) within the MMT tumor cell population. These cells were characterized by elevated surface levels of stem cell markers CD44 and Sca1 and by rapid metastasis. Inactivation of the hsp70 gene delayed the initiation of mammary tumors. This delay in tumor initiation imposed by loss of hsp70 was correlated with a decreased pool of CIC. Interestingly, hsp70 knockout significantly reduced invasion and metastasis by mammary tumor cells and implicated its product Hsp72 in cell migration and formation of secondary neoplasms. Impaired tumorigenesis and metastasis in hsp70 knockout MMT mice was associated with down-regulation of the met gene and reduced activition of the oncogenic c-Met protein. These experiments therefore showed Hsp72 to be involved in the growth and progression of mammary carcinoma and highlighted this protein as a potential target for anti-cancer drug development.
- Published
- 2015
45. Prenatal protein malnutrition decreases KCNJ3 and 2DG activity in rat prefrontal cortex
- Author
-
David J. Mokler, Janina R. Galler, Ana C. Amaral, Jill McGaughy, Mira Jakovcevski, R. J. Rushmore, Stuart K. Calderwood, Douglas L. Rosene, and Schahram Akbarian
- Subjects
Male ,medicine.medical_specialty ,Brain activity and meditation ,Down-Regulation ,Gene Expression ,Prefrontal Cortex ,Hippocampus ,Deoxyglucose ,Article ,Western blot ,Pregnancy ,Internal medicine ,Gene expression ,medicine ,Animals ,Rats, Long-Evans ,G protein-coupled inwardly-rectifying potassium channel ,Prefrontal cortex ,Prenatal Nutritional Physiological Phenomena ,medicine.diagnostic_test ,General Neuroscience ,Malnutrition ,Rats ,Endocrinology ,Real-time polymerase chain reaction ,G Protein-Coupled Inwardly-Rectifying Potassium Channels ,nervous system ,Female ,Brainstem ,Psychology - Abstract
Prenatal protein malnutrition (PPM) in rats causes enduring changes in brain and behavior including increased cognitive rigidity and decreased inhibitory control. A preliminary gene microarray screen of PPM rat prefrontal cortex (PFC) identified alterations in KCNJ3 (GIRK1/Kir3.1), a gene important for regulating neuronal excitability. Follow-up with polymerase chain reaction and Western blot showed decreased KCNJ3 expression in the PFC, but not hippocampus or brainstem. To verify localization of the effect to the PFC, baseline regional brain activity was assessed with 14C-2-deoxyglucose. Results showed decreased activation in the PFC but not hippocampus. Together these findings point to the unique vulnerability of the PFC to the nutritional insult during early brain development, with enduring effects in adulthood on KCNJ3 expression and baseline metabolic activity.
- Published
- 2015
46. Report on the VIIth International Symposium on Heat Shock Proteins in Biology & Medicine
- Author
-
Lawrence E. Hightower and Stuart K. Calderwood
- Subjects
Inflammation ,Aging ,Cell Biology ,Biology ,Meeting Review ,Biochemistry ,Hsp70 ,DNA-Binding Proteins ,Heat shock factor ,Graduate students ,Neoplasms ,Shock (circulatory) ,Heat shock protein ,Immunology ,medicine ,Humans ,medicine.symptom ,Heat shock ,Protein Processing, Post-Translational ,Heat-Shock Proteins ,Molecular Chaperones - Abstract
This seventh symposium in a series on heat shock proteins in biology and medicine was held November 1-5, 2014, at the Hilton Hotel in Old Town Alexandria, Virginia. Approximately 70 participants including principal investigators, postdoctoral fellows, and graduate students were in attendance. The major themes were: new properties of heat shock proteins (HSPs) and heat shock factor (HSF) and role in the etiology of cancer, molecular chaperones in aging, extracellular HSPs in inflammation and immunity, role of heat shock and the heat shock response in immunity and cancer, protein aggregation disorders and HSP expression, and Hsp70 in blood cell differentiation. The next meeting is planned for the fall of 2016 in the same venue.
- Published
- 2014
47. A Novel Heat Shock Protein 70-based Vaccine Prepared from DC-Tumor Fusion Cells
- Author
-
Desheng, Weng, Stuart K, Calderwood, and Jianlin, Gong
- Subjects
Cell Fusion ,Ovarian Neoplasms ,Humans ,Breast Neoplasms ,Female ,HSP70 Heat-Shock Proteins ,Dendritic Cells ,Immunotherapy ,Cancer Vaccines ,Neoplasm Proteins - Abstract
We have developed an enhanced molecular chaperone-based vaccine through rapid isolation of Hsp70 peptide complexes after the fusion of tumor and dendritic cells (Hsp70.PC-F). In this approach, the tumor antigens are introduced into the antigen processing machinery of dendritic cells through the cell fusion process and thus we can obtain antigenic tumor peptides or their intermediates that have been processed by dendritic cells. Our results show that Hsp70.PC-F has increased immunogenicity compared to preparations from tumor cells alone and therefore constitutes an improved formulation of chaperone protein-based tumor vaccine.
- Published
- 2017
48. Molecular Chaperone Receptors
- Author
-
Stuart K. Calderwood, Jimmy R. Theriault, Ayesha Murshid, and Jianlin Gong
- Subjects
0301 basic medicine ,Cell type ,CHO Cells ,Spodoptera ,Article ,Mice ,03 medical and health sciences ,Cricetulus ,Cell Line, Tumor ,Heat shock protein ,Sf9 Cells ,Extracellular ,Animals ,Humans ,HSP70 Heat-Shock Proteins ,Lectins, C-Type ,HSP90 Heat-Shock Proteins ,Cloning, Molecular ,Scavenger receptor ,Receptor ,Mice, Knockout ,Receptors, Scavenger ,Chemistry ,Chromatography, Ion Exchange ,Prefoldin ,Hsp70 ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,CDC37 - Abstract
Extracellular heat shock proteins (HSP) play important roles in cell signaling and immunity. Many of these effects are mediated by surface receptors expressed on a wide range of cell types. We have investigated the nature of such proteins by cloning candidate receptors into cells (CHO-K1) with the rare property of being null for HSP binding. Using this approach we have discovered that Hsp70 binds avidly to at least two classes of receptors including: (1) c-type lectin receptors (CLR) and (2) scavenger receptors (SR). However, the structural nature of the receptor-ligand interactions is not clear at this time. Hsp70 can bind to LOX-1 (a member of both the CLR and SR), with the c-type lectin binding domain (CTLD) as well as the SR family members SREC-I and FEEL-1/CLEVER-1/STABILIN-1, which by contrast have arrays of EGF-like repeats in their extracellular domains. In this chapter we will discuss: (1) methods for discovery of HSP receptors, (2) approaches to the study of individual receptors in cells that contain multiple such receptors, and (3) methods for investigating HSP receptor function in vivo.
- Published
- 2017
49. Role of Heat Shock Factors in Stress-Induced Transcription
- Author
-
Ayesha, Murshid, Thomas L, Prince, Ben, Lang, and Stuart K, Calderwood
- Subjects
0301 basic medicine ,Chromatin Immunoprecipitation ,Electrophoretic Mobility Shift Assay ,DNA ,Article ,Mice ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Heat Shock Transcription Factors ,Stress, Physiological ,NIH 3T3 Cells ,Animals ,Humans ,Promoter Regions, Genetic ,Molecular Biology ,Heat-Shock Proteins ,Heat-Shock Response ,030217 neurology & neurosurgery ,HeLa Cells ,Transcription Factors - Abstract
Heat shock proteins (HSP) are rapidly induced after stresses such as heat shock and accumulate at high concentrations in cells. HSP induction involves primarily a family of heat shock transcription factors (HSF) that bind the heat shock elements of the HSP genes and mediate transcription in trans. We discuss methods for the study of HSP binding to HSP promoters and the consequent increases in HSP gene expression in vitro and in vivo.
- Published
- 2017
50. A Novel Heat Shock Protein 70-based Vaccine Prepared from DC-Tumor Fusion Cells
- Author
-
Desheng Weng, Stuart K. Calderwood, and Jianlin Gong
- Subjects
0301 basic medicine ,chemistry.chemical_classification ,Fusion ,Cell fusion ,Antigen processing ,medicine.medical_treatment ,Immunogenicity ,Peptide ,Immunotherapy ,Hsp70 ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Antigen ,chemistry ,030220 oncology & carcinogenesis ,medicine - Abstract
We have developed an enhanced molecular chaperone-based vaccine through rapid isolation of Hsp70 peptide complexes after the fusion of tumor and dendritic cells (Hsp70.PC-F). In this approach, the tumor antigens are introduced into the antigen processing machinery of dendritic cells through the cell fusion process and thus we can obtain antigenic tumor peptides or their intermediates that have been processed by dendritic cells. Our results show that Hsp70.PC-F has increased immunogenicity compared to preparations from tumor cells alone and therefore constitutes an improved formulation of chaperone protein-based tumor vaccine.
- Published
- 2017
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.