81 results on '"Peter C. Gray"'
Search Results
2. CRIPTO antagonist ALK4L75A-Fc inhibits breast cancer cell plasticity and adaptation to stress
- Author
-
Ozlen Balcioglu, Richard E. Heinz, David W. Freeman, Brooke L. Gates, Berhane M. Hagos, Evan Booker, Elnaz Mirzaei Mehrabad, Hyrum T. Diesen, Kishan Bhakta, Supraja Ranganathan, Masami Kachi, Mathias Leblanc, Peter C. Gray, and Benjamin T. Spike
- Subjects
Breast cancer ,Plasticity ,CRIPTO ,Stress adaptation ,Cancer stem cells ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background CRIPTO is a multi-functional signaling protein that promotes stemness and oncogenesis. We previously developed a CRIPTO antagonist, ALK4L75A-Fc, and showed that it causes loss of the stem cell phenotype in normal mammary epithelia suggesting it may similarly inhibit CRIPTO-dependent plasticity in breast cancer cells. Methods We focused on two triple negative breast cancer cell lines (MDA-MB-231 and MDA-MB-468) to measure the effects of ALK4L75A-Fc on cancer cell behavior under nutrient deprivation and endoplasmic reticulum stress. We characterized the proliferation and migration of these cells in vitro using time-lapse microscopy and characterized stress-dependent changes in the levels and distribution of CRIPTO signaling mediators and cancer stem cell markers. We also assessed the effects of ALK4L75A-Fc on proliferation, EMT, and stem cell markers in vivo as well as on tumor growth and metastasis using inducible lentiviral delivery or systemic administration of purified ALK4L75A-Fc, which represents a candidate therapeutic approach. Results ALK4L75A-Fc inhibited adaptive responses of breast cancer cells under conditions of nutrient and ER stress and reduced their proliferation, migration, clonogenicity, and expression of EMT and cancer stem cell markers. ALK4L75A-Fc also inhibited proliferation of human breast cancer cells in stressed tumor microenvironments in xenografts and reduced both primary tumor size and metastatic burden. Conclusions Cancer cell adaptation to stresses such as nutrient deprivation, hypoxia, and chemotherapy can critically contribute to dormancy, metastasis, therapy resistance, and recurrence. Identifying mechanisms that govern cellular adaptation, plasticity, and the emergence of stem-like cancer cells may be key to effective anticancer therapies. Results presented here indicate that targeting CRIPTO with ALK4L75A-Fc may have potential as such a therapy since it inhibits breast cancer cell adaptation to microenvironmental challenges and associated stem-like and EMT phenotypes.
- Published
- 2020
- Full Text
- View/download PDF
3. Preoperative plasma fatty acid metabolites inform risk of prostate cancer progression and may be used for personalized patient stratification
- Author
-
Eugenio Zoni, Martina Minoli, Cédric Bovet, Anne Wehrhan, Salvatore Piscuoglio, Charlotte K. Y. Ng, Peter C. Gray, Martin Spahn, George N. Thalmann, and Marianna Kruithof-de Julio
- Subjects
Acylcarnitines ,Prostate cancer ,Metabolomics ,Disease progression ,Fatty acid metabolism ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Little is known about the relationship between the metabolite profile of plasma from pre-operative prostate cancer (PCa) patients and the risk of PCa progression. In this study we investigated the association between pre-operative plasma metabolites and risk of biochemical-, local- and metastatic-recurrence, with the aim of improving patient stratification. Methods We conducted a case-control study within a cohort of PCa patients recruited between 1996 and 2015. The age-matched primary cases (n = 33) were stratified in low risk, high risk without progression and high risk with progression as defined by the National Comprehensive Cancer Network. These samples were compared to metastatic (n = 9) and healthy controls (n = 10). The pre-operative plasma from primary cases and the plasma from metastatic patients and controls were assessed with untargeted metabolomics by LC-MS. The association between risk of progression and metabolite abundance was calculated using multivariate Cox proportional-hazard regression and the relationship between metabolites and outcome was calculated using median cut-off normalized values of metabolite abundance by Log-Rank test using the Kaplan Meier method. Results Medium-chain acylcarnitines (C6-C12) were positively associated with the risk of PSA progression (p = 0.036, median cut-off) while long-chain acylcarnitines (C14-C16) were inversely associated with local (p = 0.034) and bone progression (p = 0.0033). In primary cases, medium-chain acylcarnitines were positively associated with suberic acid, which also correlated with the risk of PSA progression (p = 0.032, Log-Rank test). In the metastatic samples, this effect was consistent for hexanoylcarnitine, L.octanoylcarnitine and decanoylcarnitine. Medium-chain acylcarnitines and suberic acid displayed the same inverse association with tryptophan, while indoleacetic acid, a breakdown product of tryptophan metabolism was strongly associated with PSA (p = 0.0081, Log-Rank test) and lymph node progression (p = 0.025, Log-Rank test). These data were consistent with the increased expression of indoleamine 2,3 dioxygenase (IDO1) in metastatic versus primary samples (p = 0.014). Finally, functional experiments revealed a synergistic effect of long chain fatty acids in combination with dihydrotestosterone administration on the transcription of androgen responsive genes. Conclusions This study strengthens the emerging link between fatty acid metabolism and PCa progression and suggests that measuring levels of medium- and long-chain acylcarnitines in pre-operative patient plasma may provide a basis for improving patient stratification.
- Published
- 2019
- Full Text
- View/download PDF
4. Whence CRIPTO: The Reemergence of an Oncofetal Factor in ‘Wounds’ That Fail to Heal
- Author
-
David W. Freeman, Elisa Rodrigues Sousa, Sofia Karkampouna, Eugenio Zoni, Peter C. Gray, David S. Salomon, Marianna Kruithof-de Julio, and Benjamin T. Spike
- Subjects
CRIPTO ,stem cells ,EMT ,cancer ,metastasis ,fibrosis ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
There exists a set of factors termed oncofetal proteins that play key roles in ontogeny before they decline or disappear as the organism’s tissues achieve homeostasis, only to then re-emerge in cancer. Although the unique therapeutic potential presented by such factors has been recognized for more than a century, their clinical utility has yet to be fully realized1. This review highlights the small signaling protein CRIPTO encoded by the tumor derived growth factor 1 (TDGF1/Tdgf1) gene, an oft cited oncofetal protein whose presence in the cancer literature as a tumor promoter, diagnostic marker and viable therapeutic target continues to grow. We touch lightly on features well established and well-reviewed since its discovery more than 30 years ago, including CRIPTO’s early developmental roles and modulation of SMAD2/3 activation by a selected set of transforming growth factor β (TGF-β) family ligands. We predominantly focus instead on more recent and less well understood additions to the CRIPTO signaling repertoire, on its potential upstream regulators and on new conceptual ground for understanding its mode of action in the multicellular and often stressful contexts of neoplastic transformation and progression. We ask whence it re-emerges in cancer and where it ‘hides’ between the time of its fetal activity and its oncogenic reemergence. In this regard, we examine CRIPTO’s restriction to rare cells in the adult, its potential for paracrine crosstalk, and its emerging role in inflammation and tissue regeneration—roles it may reprise in tumorigenesis, acting on subsets of tumor cells to foster cancer initiation and progression. We also consider critical gaps in knowledge and resources that stand between the recent, exciting momentum in the CRIPTO field and highly actionable CRIPTO manipulation for cancer therapy and beyond.
- Published
- 2021
- Full Text
- View/download PDF
5. The Activin Receptor, Activin-Like Kinase 4, Mediates Toxoplasma Gondii Activation of Hypoxia Inducible Factor-1
- Author
-
Agnieszka Lis, Mandi Wiley, Joan Vaughan, Peter C. Gray, and Ira J. Blader
- Subjects
toxoplasma and toxoplasmosis ,hypoxia ,transcripional regulation ,parasite - host interactions ,tgf-beta signaling ,Microbiology ,QR1-502 - Abstract
To grow and cause disease, intracellular pathogens modulate host cell processes. Identifying these processes as well as the mechanisms used by the pathogens to manipulate them is important for the development of more effective therapeutics. As an example, the intracellular parasite Toxoplasma gondii induces a wide variety of changes to its host cell, including altered membrane trafficking, cytoskeletal reorganization, and differential gene expression. Although several parasite molecules and their host targets have been identified that mediate- these changes, few are known to be required for parasite replication. One exception is the host cell transcription factor, hypoxia-inducible factor-1 (HIF-1), which is required for parasite replication in an oxygen-dependent manner. Toxoplasma activates HIF-1 by stabilizing the HIF-1α subunit, and this is dependent on the signaling from the Activin-Like Kinase (ALK) receptor superfamily. Here, we demonstrate that specific overexpression of the ALK family member, ALK4, increased HIF-1 activity in Toxoplasma-infected cells, and this increase required ALK4 kinase activity. Moreover, Toxoplasma stimulated ALK4 to dimerize with its co-receptor, ActRII, and also increased ALK4 kinase activity, thereby demonstrating that Toxoplasma activates the ALK4 receptor. ALK4 activation of HIF-1 was independent of canonical SMAD signaling but rather was dependent on the non-canonical Rho GTPase and JNK MAP kinase signaling pathways. Finally, Toxoplasma increased rates of ALK4 ubiquitination and turnover. These data provide the first evidence indicating that ALK4 signaling is a target for a microbial pathogen to manipulate its host cell.
- Published
- 2019
- Full Text
- View/download PDF
6. ALK1Fc Suppresses the Human Prostate Cancer Growth in in Vitro and in Vivo Preclinical Models
- Author
-
Letizia Astrologo, Eugenio Zoni, Sofia Karkampouna, Peter C. Gray, Irena Klima, Joël Grosjean, Marie J. Goumans, Lukas J. A. C. Hawinkels, Gabri van der Pluijm, Martin Spahn, George N. Thalmann, Peter ten Dijke, and Marianna Kruithof-de Julio
- Subjects
BMP9 ,ALK1 ,ALK2 ,ALK1Fc ,NOTCH ,prostate cancer ,Biology (General) ,QH301-705.5 - Abstract
Prostate cancer is the second most common cancer in men and lethality is normally associated with the consequences of metastasis rather than the primary tumor. Therefore, targeting the molecular pathways that underlie dissemination of primary tumor cells and the formation of metastases has a great clinical value. Bone morphogenetic proteins (BMPs) play a critical role in tumor progression and this study focuses on the role of BMP9- Activin receptor-Like Kinase 1 and 2 (ALK1 and ALK2) axis in prostate cancer. In order to study the effect of BMP9 in vitro and in vivo on cancer cells and tumor growth, we used a soluble chimeric protein consisting of the ALK1 extracellular domain (ECD) fused to human Fc (ALK1Fc) that prevents binding of BMP9 to its cell surface receptors and thereby blocks its ability to activate downstream signaling. ALK1Fc sequesters BMP9 and the closely related BMP10 while preserving the activation of ALK1 and ALK2 through other ligands. We show that ALK1Fc acts in vitro to decrease BMP9-mediated signaling and proliferation of prostate cancer cells with tumor initiating and metastatic potential. In line with these observations, we demonstrate that ALK1Fc also reduces tumor cell proliferation and tumor growth in vivo in an orthotopic transplantation model, as well as in the human patient derived xenograft BM18. Furthermore, we also provide evidence for crosstalk between BMP9 and NOTCH and find that ALK1Fc inhibits NOTCH signaling in human prostate cancer cells and blocks the induction of the NOTCH target Aldehyde dehydrogenase member ALDH1A1, which is a clinically relevant marker associated with poor survival and advanced-stage prostate cancer. Our study provides the first demonstration that ALK1Fc inhibits prostate cancer progression, identifying BMP9 as a putative therapeutic target and ALK1Fc as a potential therapy. Altogether, these findings support the validity of ongoing clinical development of drugs blocking ALK1 and ALK2 receptor activity.
- Published
- 2017
- Full Text
- View/download PDF
7. CRIPTO/GRP78 Signaling Maintains Fetal and Adult Mammary Stem Cells Ex Vivo
- Author
-
Benjamin T. Spike, Jonathan A. Kelber, Evan Booker, Madhuri Kalathur, Rose Rodewald, Julia Lipianskaya, Justin La, Marielle He, Tracy Wright, Richard Klemke, Geoffrey M. Wahl, and Peter C. Gray
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Little is known about the extracellular signaling factors that govern mammary stem cell behavior. Here, we identify CRIPTO and its cell-surface receptor GRP78 as regulators of stem cell behavior in isolated fetal and adult mammary epithelial cells. We develop a CRIPTO antagonist that promotes differentiation and reduces self-renewal of mammary stem cell-enriched populations cultured ex vivo. By contrast, CRIPTO treatment maintains the stem cell phenotype in these cultures and yields colonies with enhanced mammary gland reconstitution capacity. Surface expression of GRP78 marks CRIPTO-responsive, stem cell-enriched fetal and adult mammary epithelial cells, and deletion of GRP78 from adult mammary epithelial cells blocks their mammary gland reconstitution potential. Together, these findings identify the CRIPTO/GRP78 pathway as a developmentally conserved regulator of fetal and adult mammary stem cell behavior ex vivo, with implications for the stem-like cells found in many cancers.
- Published
- 2014
- Full Text
- View/download PDF
8. A class of anti-inflammatory lipids decrease with aging in the central nervous system
- Author
-
Dan Tan, Srihari Konduri, Meric Erikci Ertunc, Pan Zhang, Justin Wang, Tina Chang, Antonio F. M. Pinto, Andrea Rocha, Cynthia J. Donaldson, Joan M. Vaughan, Raissa G. Ludwig, Elizabeth Willey, Manasi Iyer, Peter C. Gray, Pamela Maher, Nicola J. Allen, J. Bradley Zuchero, Andrew Dillin, Marcelo A. Mori, Steven G. Kohama, Dionicio Siegel, and Alan Saghatelian
- Subjects
Cell Biology ,Molecular Biology - Abstract
Lipids contribute to the structure, development, and function of healthy brains. Dysregulated lipid metabolism is linked to aging and diseased brains. However, our understanding of lipid metabolism in aging brains remains limited. Here we examined the brain lipidome of mice across their lifespan using untargeted lipidomics. Co-expression network analysis highlighted a progressive decrease in 3-sulfogalactosyl diacylglycerols (SGDGs) and SGDG pathway members, including the potential degradation products lyso-SGDGs. SGDGs show an age-related decline specifically in the central nervous system and are associated with myelination. We also found that an SGDG dramatically suppresses LPS-induced gene expression and release of pro-inflammatory cytokines from macrophages and microglia by acting on the NF-κB pathway. The detection of SGDGs in human and macaque brains establishes their evolutionary conservation. This work enhances interest in SGDGs regarding their roles in aging and inflammatory diseases and highlights the complexity of the brain lipidome and potential biological functions in aging.
- Published
- 2022
9. 18F-FDG-PET/MR imaging to monitor disease activity in large vessel vasculitis
- Author
-
Dan Pugh, Dilip Patel, Gillian Macnaught, Alicja Czopek, Lorraine Bruce, James Donachie, Peter J. Gallacher, Sovira Tan, Mark Ahlman, Peter C. Grayson, Neil Basu, and Neeraj Dhaun
- Subjects
Science - Abstract
Abstract Disease-monitoring in large vessel vasculitis (LVV) is challenging. Simultaneous 18F-fluorodeoxyglucose positron emission tomography with magnetic resonance imaging (PET/MRI) provides functional assessment of vascular inflammation alongside high-definition structural imaging with a relatively low burden of radiation exposure. Here, we investigate the ability of PET/MRI to monitor LVV disease activity longitudinally in a prospective cohort of patients with active LVV. We demonstrate that both the PET and MRI components of the scan can distinguish active from inactive disease using established quantification methods. Using logistic-regression modelling of PET/MRI metrics, we devise a novel PET/MRI-specific Vasculitis Activity using MR PET (VAMP) score which is able to distinguish active from inactive disease with more accuracy than established methods and detects changes in disease activity longitudinally. These findings are evaluated in an independent validation cohort. Finally, PET/MRI improves clinicians’ assessment of LVV disease activity and confidence in disease management, as assessed via clinician survey. In summary, PET/MRI may be useful in tracking disease activity and assessing treatment-response in LVV. Based on our findings, larger, prospective studies assessing PET/MRI in LVV are now warranted.
- Published
- 2024
- Full Text
- View/download PDF
10. Cell surface GRP78 promotes stemness in normal and neoplastic cells
- Author
-
Siyuan Zhang, Amanda E. Yamasaki, Sergio Ruiz, Tyson W. Lager, Tomoaki Hishida, Yuriko Hishida, Juan Carlos Izpisua Belmonte, Ian H. Guldner, Athanasia D. Panopoulos, Clay Conner, Robert C. Gilson, Min-Zu Wu, Jonathan A. Kelber, and Peter C. Gray
- Subjects
Lung Neoplasms ,Induced Pluripotent Stem Cells ,Transplantation, Heterologous ,lcsh:Medicine ,Breast Neoplasms ,medicine.disease_cause ,Article ,Mice ,03 medical and health sciences ,Breast cancer ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Cell Self Renewal ,RNA, Small Interfering ,Induced pluripotent stem cell ,lcsh:Science ,Endoplasmic Reticulum Chaperone BiP ,Heat-Shock Proteins ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,Multidisciplinary ,biology ,Cancer stem cells ,CD44 ,lcsh:R ,Cancer ,Cell Differentiation ,Cellular Reprogramming ,medicine.disease ,Embryonic stem cell ,3. Good health ,Cell Transformation, Neoplastic ,HEK293 Cells ,030220 oncology & carcinogenesis ,Cancer cell ,Neoplastic Stem Cells ,biology.protein ,Cancer research ,Female ,RNA Interference ,lcsh:Q ,Stem cell ,Carcinogenesis ,Reprogramming ,Transcription Factors - Abstract
Reliable approaches to identify stem cell mechanisms that mediate aggressive cancer could have great therapeutic value, based on the growing evidence of embryonic signatures in metastatic cancers. However, how to best identify and target stem-like mechanisms aberrantly acquired by cancer cells has been challenging. We harnessed the power of reprogramming to examine GRP78, a chaperone protein generally restricted to the endoplasmic reticulum in normal tissues, but which is expressed on the cell surface of human embryonic stem cells and many cancer types. We have discovered that (1) cell surface GRP78 (sGRP78) is expressed on iPSCs and is important in reprogramming, (2) sGRP78 promotes cellular functions in both pluripotent and breast cancer cells (3) overexpression of GRP78 in breast cancer cells leads to an induction of a CD24−/CD44+ tumor initiating cell (TIC) population (4) sGRP78+ breast cancer cells are enriched for stemness genes and appear to be a subset of TICs (5) sGRP78+ breast cancer cells show an enhanced ability to seed metastatic organ sites in vivo. These collective findings show that GRP78 has important functions in regulating both pluripotency and oncogenesis, and suggest that sGRP78 marks a stem-like population in breast cancer cells that has increased metastatic potential in vivo.
- Published
- 2020
11. Whence CRIPTO: The Reemergence of an Oncofetal Factor in ‘Wounds’ That Fail to Heal
- Author
-
Eugenio Zoni, Benjamin T Spike, Peter C. Gray, David S. Salomon, Elisa Rodrigues Sousa, Sofia Karkampouna, Marianna Kruithof-de Julio, and David W. Freeman
- Subjects
QH301-705.5 ,610 Medicine & health ,Review ,Biology ,CRIPTO ,Cripto ,medicine.disease_cause ,Catalysis ,Metastasis ,Inorganic Chemistry ,Paracrine signalling ,stem cells ,Transforming Growth Factor beta ,medicine ,cancer ,metastasis ,Animals ,Humans ,Neoplastic transformation ,Physical and Theoretical Chemistry ,Biology (General) ,Molecular Biology ,QD1-999 ,Spectroscopy ,Organic Chemistry ,fibrosis ,EMT ,Cancer ,therapeutic target ,General Medicine ,Tumor-Derived ,medicine.disease ,Computer Science Applications ,Chemistry ,Carcinogenesis ,Neuroscience ,Transforming growth factor ,Signal Transduction - Abstract
There exists a set of factors termed oncofetal proteins that play key roles in ontogeny before they decline or disappear as the organism’s tissues achieve homeostasis, only to then re-emerge in cancer. Although the unique therapeutic potential presented by such factors has been recognized for more than a century, their clinical utility has yet to be fully realized1. This review highlights the small signaling protein CRIPTO encoded by the tumor derived growth factor 1 (TDGF1/Tdgf1) gene, an oft cited oncofetal protein whose presence in the cancer literature as a tumor promoter, diagnostic marker and viable therapeutic target continues to grow. We touch lightly on features well established and well-reviewed since its discovery more than 30 years ago, including CRIPTO’s early developmental roles and modulation of SMAD2/3 activation by a selected set of transforming growth factor β (TGF-β) family ligands. We predominantly focus instead on more recent and less well understood additions to the CRIPTO signaling repertoire, on its potential upstream regulators and on new conceptual ground for understanding its mode of action in the multicellular and often stressful contexts of neoplastic transformation and progression. We ask whence it re-emerges in cancer and where it ‘hides’ between the time of its fetal activity and its oncogenic reemergence. In this regard, we examine CRIPTO’s restriction to rare cells in the adult, its potential for paracrine crosstalk, and its emerging role in inflammation and tissue regeneration—roles it may reprise in tumorigenesis, acting on subsets of tumor cells to foster cancer initiation and progression. We also consider critical gaps in knowledge and resources that stand between the recent, exciting momentum in the CRIPTO field and highly actionable CRIPTO manipulation for cancer therapy and beyond.
- Published
- 2021
12. Design-augmented (DA) biologics: BMP chimeras for bone and cartilage regeneration
- Author
-
Senyon Choe and Peter C. Gray
- Subjects
Biological Products ,Bone Regeneration ,Bone Morphogenetic Protein 6 ,Bone Morphogenetic Protein 7 ,Recombinant Fusion Proteins ,Regeneration (biology) ,Cartilage ,Biomedical Engineering ,Bone Morphogenetic Protein 2 ,Biology ,Activins ,Cell biology ,medicine.anatomical_structure ,Rheumatology ,Drug Design ,Bone Morphogenetic Proteins ,medicine ,Humans ,Regeneration ,Orthopedics and Sports Medicine - Published
- 2020
13. CRIPTO antagonist ALK4L75A-Fc inhibits breast cancer cell plasticity and adaptation to stress
- Author
-
Berhane M. Hagos, Brooke L. Gates, David W. Freeman, Masami Kachi, Benjamin T. Spike, Kishan Bhakta, Elnaz Mirzaei Mehrabad, Hyrum T Diesen, Supraja Ranganathan, Mathias Leblanc, Ozlen Balcioglu, Evan Booker, Peter C. Gray, and Richard E. Heinz
- Subjects
0301 basic medicine ,Plasticity ,Cancer stem cells ,CRIPTO ,Biology ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Stem cell marker ,Cripto ,medicine.disease_cause ,lcsh:RC254-282 ,Metastasis ,03 medical and health sciences ,Breast cancer ,030104 developmental biology ,0302 clinical medicine ,Cancer stem cell ,030220 oncology & carcinogenesis ,Cancer cell ,medicine ,Cancer research ,Stress adaptation ,Carcinogenesis ,Triple-negative breast cancer - Abstract
Background CRIPTO is a multi-functional signaling protein that promotes stemness and oncogenesis. We previously developed a CRIPTO antagonist, ALK4L75A-Fc, and showed that it causes loss of the stem cell phenotype in normal mammary epithelia suggesting it may similarly inhibit CRIPTO-dependent plasticity in breast cancer cells. Methods We focused on two triple negative breast cancer cell lines (MDA-MB-231 and MDA-MB-468) to measure the effects of ALK4L75A-Fc on cancer cell behavior under nutrient deprivation and endoplasmic reticulum stress. We characterized the proliferation and migration of these cells in vitro using time-lapse microscopy and characterized stress-dependent changes in the levels and distribution of CRIPTO signaling mediators and cancer stem cell markers. We also assessed the effects of ALK4L75A-Fc on proliferation, EMT, and stem cell markers in vivo as well as on tumor growth and metastasis using inducible lentiviral delivery or systemic administration of purified ALK4L75A-Fc, which represents a candidate therapeutic approach. Results ALK4L75A-Fc inhibited adaptive responses of breast cancer cells under conditions of nutrient and ER stress and reduced their proliferation, migration, clonogenicity, and expression of EMT and cancer stem cell markers. ALK4L75A-Fc also inhibited proliferation of human breast cancer cells in stressed tumor microenvironments in xenografts and reduced both primary tumor size and metastatic burden. Conclusions Cancer cell adaptation to stresses such as nutrient deprivation, hypoxia, and chemotherapy can critically contribute to dormancy, metastasis, therapy resistance, and recurrence. Identifying mechanisms that govern cellular adaptation, plasticity, and the emergence of stem-like cancer cells may be key to effective anticancer therapies. Results presented here indicate that targeting CRIPTO with ALK4L75A-Fc may have potential as such a therapy since it inhibits breast cancer cell adaptation to microenvironmental challenges and associated stem-like and EMT phenotypes.
- Published
- 2020
14. A Multidisciplinary Review of the Roles of Cripto in the Scientific Literature Through a Bibliometric Analysis of its Biological Roles
- Author
-
Marta De Menna, Federico La Manna, Sofia Karkampouna, Eugenio Zoni, Elisa Rodrigues Sousa, Peter C. Gray, and Marianna Kruithof-de Julio
- Subjects
biochemistry and molecular biology ,0301 basic medicine ,MAPK/ERK pathway ,Cancer Research ,Cellular differentiation ,610 Medicine & health ,Review ,CRIPTO ,Biology ,Cripto ,lcsh:RC254-282 ,GDF1 ,03 medical and health sciences ,bibliometric analysis ,0302 clinical medicine ,experimental medical research ,cancer ,development ,Protein kinase B ,PI3K/AKT/mTOR pathway ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Cell biology ,TDGF-1 ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,NODAL ,hormones, hormone substitutes, and hormone antagonists ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Cripto is a small glycosylphosphatidylinisitol (GPI)-anchored and secreted oncofetal protein that plays important roles in regulating normal physiological processes, including stem cell differentiation, embryonal development, and tissue growth and remodeling, as well as pathological processes such as tumor initiation and progression. Cripto functions as a co-receptor for TGF-β ligands such as Nodal, GDF1, and GDF3. Soluble and secreted forms of Cripto also exhibit growth factor-like activity and activate SRC/MAPK/PI3K/AKT pathways. Glucose-Regulated Protein 78 kDa (GRP78) binds Cripto at the cell surface and has been shown to be required for Cripto signaling via both TGF-β and SRC/MAPK/PI3K/AKT pathways. To provide a comprehensive overview of the scientific literature related to Cripto, we performed, for the first time, a bibliometric analysis of the biological roles of Cripto as reported in the scientific literature covering the last 10 years. We present different fields of knowledge in comprehensive areas of research on Cripto, ranging from basic to translational research, using a keyword-driven approach. Our ultimate aim is to aid the scientific community in conducting targeted research by identifying areas where research has been conducted so far and, perhaps more importantly, where critical knowledge is still missing.
- Published
- 2020
15. CRIPTO antagonist ALK4
- Author
-
Ozlen, Balcioglu, Richard E, Heinz, David W, Freeman, Brooke L, Gates, Berhane M, Hagos, Evan, Booker, Elnaz, Mirzaei Mehrabad, Hyrum T, Diesen, Kishan, Bhakta, Supraja, Ranganathan, Masami, Kachi, Mathias, Leblanc, Peter C, Gray, and Benjamin T, Spike
- Subjects
Plasticity ,Recombinant Fusion Proteins ,Cell Plasticity ,Triple Negative Breast Neoplasms ,CRIPTO ,GPI-Linked Proteins ,Mice ,Breast cancer ,Protein Domains ,Cell Line, Tumor ,Tumor Microenvironment ,Animals ,Humans ,Point Mutation ,Stress adaptation ,Cancer stem cells ,Endoplasmic Reticulum Stress ,Xenograft Model Antitumor Assays ,Immunoglobulin Fc Fragments ,Neoplasm Proteins ,Neoplastic Stem Cells ,Intercellular Signaling Peptides and Proteins ,Tumor Hypoxia ,Female ,Neoplasm Recurrence, Local ,Activin Receptors, Type I ,Protein Binding ,Research Article - Abstract
Background CRIPTO is a multi-functional signaling protein that promotes stemness and oncogenesis. We previously developed a CRIPTO antagonist, ALK4L75A-Fc, and showed that it causes loss of the stem cell phenotype in normal mammary epithelia suggesting it may similarly inhibit CRIPTO-dependent plasticity in breast cancer cells. Methods We focused on two triple negative breast cancer cell lines (MDA-MB-231 and MDA-MB-468) to measure the effects of ALK4L75A-Fc on cancer cell behavior under nutrient deprivation and endoplasmic reticulum stress. We characterized the proliferation and migration of these cells in vitro using time-lapse microscopy and characterized stress-dependent changes in the levels and distribution of CRIPTO signaling mediators and cancer stem cell markers. We also assessed the effects of ALK4L75A-Fc on proliferation, EMT, and stem cell markers in vivo as well as on tumor growth and metastasis using inducible lentiviral delivery or systemic administration of purified ALK4L75A-Fc, which represents a candidate therapeutic approach. Results ALK4L75A-Fc inhibited adaptive responses of breast cancer cells under conditions of nutrient and ER stress and reduced their proliferation, migration, clonogenicity, and expression of EMT and cancer stem cell markers. ALK4L75A-Fc also inhibited proliferation of human breast cancer cells in stressed tumor microenvironments in xenografts and reduced both primary tumor size and metastatic burden. Conclusions Cancer cell adaptation to stresses such as nutrient deprivation, hypoxia, and chemotherapy can critically contribute to dormancy, metastasis, therapy resistance, and recurrence. Identifying mechanisms that govern cellular adaptation, plasticity, and the emergence of stem-like cancer cells may be key to effective anticancer therapies. Results presented here indicate that targeting CRIPTO with ALK4L75A-Fc may have potential as such a therapy since it inhibits breast cancer cell adaptation to microenvironmental challenges and associated stem-like and EMT phenotypes.
- Published
- 2020
16. Patient-derived xenografts and organoids model therapy response in prostate cancer
- Author
-
Mirjam Kiener, Marianna Kruithof-de Julio, George N. Thalmann, Salvatore Piscuoglio, Marta De Menna, Maria R. De Filippo, Federico La Manna, Peter C. Gray, Andrea Sboner, Eugenio Zoni, Charlotte K.Y. Ng, Jo eumll Grosjean, David Keller, Christian U. Stirnimann, Sofia Karkampouna, Marco Bolis, Tijmen H. Booij, Martin Spahn, Irena Klima, Mark A. Rubin, Kenneth Eng, Andrea Garofoli, Jean-Philippe Theurillat, and Vera Genitsch
- Subjects
Sorafenib ,Sunitinib ,Ponatinib ,Microsatellite instability ,SPOP ,Biology ,medicine.disease ,Primary tumor ,chemistry.chemical_compound ,Prostate cancer ,chemistry ,medicine ,Organoid ,Cancer research ,medicine.drug - Abstract
Therapy resistance and metastatic processes in prostate cancer (PCa) remain undefined, due to lack of experimental models that mimic different disease stages. We describe a novel androgen-dependent PCa patient-derived xenograft (PDX) model from treatment-naïve, soft tissue metastasis (PNPCa). RNA and whole-exome sequencing of the PDX tissue and organoids confirmed transcriptomic and genomic similarity to primary tumor. PNPCa harboursBRCA2 and CHD1somatic mutations, shows anSPOP/FOXA1-like transcriptomic signature and microsatellite instability, which occurs in 3% of advanced PCa and has never been modelledin vivo. Comparison of the treatment-naïve PNPCa with additional metastatic PDXs (BM18, LAPC9), in a medium-throughput organoid screen of FDA-approved compounds, revealed differential drug sensitivities. Multikinase inhibitors (ponatinib, sunitinib, sorafenib) were broadly effective on all PDX- and patient-derived organoids from advanced cases with acquired resistance to standard-of-care compounds. This proof-of-principle study may provide a preclinical tool to screen drug responses to standard-of-care and newly identified, repurposed compounds.
- Published
- 2020
17. Patient-derived xenografts and organoids model therapy response in prostate cancer
- Author
-
Marco Bolis, Arianna Vallerga, Salvatore Piscuoglio, Andrej Benjak, Andrea Sboner, Federico La Manna, Mirjam Kiener, Joel Grosjean, Tijmen H. Booij, David Keller, Charlotte K.Y. Ng, Eugenio Zoni, Martin Spahn, Vera Genitsch, Sofia Karkampouna, Irena Klima, Mark A. Rubin, Kenneth Eng, Marianna Kruithof-de Julio, Christian U. Stirnimann, Jean-Philippe Theurillat, Andrea Garofoli, Maria R. De Filippo, Marta De Menna, George N. Thalmann, and Peter C. Gray
- Subjects
0301 basic medicine ,Sorafenib ,Male ,Science ,Medizin ,General Physics and Astronomy ,610 Medicine & health ,Antineoplastic Agents ,SPOP ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cancer screening ,03 medical and health sciences ,chemistry.chemical_compound ,Prostate cancer ,0302 clinical medicine ,medicine ,Organoid ,Humans ,Neoplasm Metastasis ,Cancer models ,Multidisciplinary ,Sunitinib ,business.industry ,Genome, Human ,Cancer stem cells ,Ponatinib ,Microsatellite instability ,Prostatic Neoplasms ,General Chemistry ,medicine.disease ,Primary tumor ,Xenograft Model Antitumor Assays ,3. Good health ,Organoids ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Androgens ,570 Life sciences ,biology ,business ,Transcriptome ,medicine.drug - Abstract
Therapy resistance and metastatic processes in prostate cancer (PCa) remain undefined, due to lack of experimental models that mimic different disease stages. We describe an androgen-dependent PCa patient-derived xenograft (PDX) model from treatment-naïve, soft tissue metastasis (PNPCa). RNA and whole-exome sequencing of the PDX tissue and organoids confirmed transcriptomic and genomic similarity to primary tumor. PNPCa harbors BRCA2 and CHD1 somatic mutations, shows an SPOP/FOXA1-like transcriptomic signature and microsatellite instability, which occurs in 3% of advanced PCa and has never been modeled in vivo. Comparison of the treatment-naïve PNPCa with additional metastatic PDXs (BM18, LAPC9), in a medium-throughput organoid screen of FDA-approved compounds, revealed differential drug sensitivities. Multikinase inhibitors (ponatinib, sunitinib, sorafenib) were broadly effective on all PDX- and patient-derived organoids from advanced cases with acquired resistance to standard-of-care compounds. This proof-of-principle study may provide a preclinical tool to screen drug responses to standard-of-care and newly identified, repurposed compounds., To date, patients still succumb to cancer, due to tumors not responding to therapy or ultimately acquiring resistance. Here the authors show that by exploiting patient derived organoids and a treatment-naïve patient derived xenograft, patient therapy can be personalized.
- Published
- 2020
18. CRIPTO promotes an aggressive tumour phenotype and resistance to treatment in hepatocellular carcinoma
- Author
-
Marianna Kruithof-de Julio, Deborah Stroka, Arantza Farina-Sarasqueta, Peter C. Gray, Hein W. Verspaget, Bart van Hoek, Alexander F. Schaapherder, Danny van der Helm, Luigi Terracciano, Sofia Karkampouna, Ewa Snaar-Jagalska, Joel Grosjean, Mark C. Burgmans, George N. Thalmann, Minneke J. Coenraad, Lanpeng Chen, Irena Klima, and Susan Osanto
- Subjects
0301 basic medicine ,Sorafenib ,Cell growth ,business.industry ,Cripto ,medicine.disease ,Phenotype ,digestive system diseases ,Pathology and Forensic Medicine ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Cancer stem cell ,030220 oncology & carcinogenesis ,Hepatocellular carcinoma ,medicine ,Cancer research ,Doxorubicin ,business ,hormones, hormone substitutes, and hormone antagonists ,Ex vivo ,medicine.drug - Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related death worldwide. Despite increasing treatment options for this disease, prognosis remains poor. CRIPTO (TDGF1) protein is expressed at high levels in several human tumours and promotes oncogenic phenotype. Its expression has been correlated to poor prognosis in HCC. In this study, we aimed to elucidate the basis for the effects of CRIPTO in HCC. We investigated CRIPTO expression levels in three cohorts of clinical cirrhotic and HCC specimens. We addressed the role of CRIPTO in hepatic tumourigenesis using Cre-loxP-controlled lentiviral vectors expressing CRIPTO in cell line-derived xenografts. Responses to standard treatments (sorafenib, doxorubicin) were assessed directly on xenograft-derived ex vivo tumour slices. CRIPTO-overexpressing patient-derived xenografts were established and used for ex vivo drug response assays. The effects of sorafenib and doxorubicin treatment in combination with a CRIPTO pathway inhibitor were tested in ex vivo cultures of xenograft models and 3D cultures. CRIPTO protein was found highly expressed in human cirrhosis and hepatocellular carcinoma specimens but not in those of healthy participants. Stable overexpression of CRIPTO in human HepG2 cells caused epithelial-to-mesenchymal transition, increased expression of cancer stem cell markers, and enhanced cell proliferation and migration. HepG2-CRIPTO cells formed tumours when injected into immune-compromised mice, whereas HepG2 cells lacking stable CRIPTO overexpression did not. High-level CRIPTO expression in xenograft models was associated with resistance to sorafenib, which could be modulated using a CRIPTO pathway inhibitor in ex vivo tumour slices. Our data suggest that a subgroup of CRIPTO-expressing HCC patients may benefit from a combinatorial treatment scheme and that sorafenib resistance may be circumvented by inhibition of the CRIPTO pathway. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
- Published
- 2018
19. Tumor Microenvironment Heterogeneity: Challenges and Opportunities
- Author
-
Jonathan A. Kelber, Peter C. Gray, P. Shisgal, Kayla Meade, Farhana Runa, and Sarkis Hamalian
- Subjects
0301 basic medicine ,Tumor microenvironment ,business.industry ,Mesenchymal stem cell ,General Engineering ,medicine.disease ,Article ,Metastasis ,Extracellular matrix ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,Tumor progression ,030220 oncology & carcinogenesis ,Cancer research ,Medicine ,Cancer-Associated Fibroblasts ,sense organs ,Stem cell ,business - Abstract
The tumor microenvironment (TME) has been recognized as an integral component of malignancies in breast and prostate tissues, contributing in confounding ways to tumor progression, metastasis, therapy resistance, and disease recurrence. Major components of the TME are immune cells, fibroblasts, pericytes, endothelial cells, mesenchymal stroma/stem cells (MSCs), and extracellular matrix (ECM) components. Herein, we discuss the molecular and cellular heterogeneity within the TME and how this presents unique challenges and opportunities for treating breast and prostate cancers.
- Published
- 2017
20. GRP78 promotes stemness in normal and neoplastic cells
- Author
-
Juan Carlos Izpisua Belmonte, Ian H. Guldner, Tyson W. Lager, Tomoaki Hishida, Clay Conner, Jonathan A. Kelber, Peter C. Gray, Athanasia D. Panopoulos, Sergio Ruiz, Amanda E. Yamasaki, Siyuan Zhang, Robert C. Gilson, Min-Zu Wu, and Yuriko Hishida
- Subjects
0303 health sciences ,CD44 ,Cancer ,Biology ,medicine.disease ,medicine.disease_cause ,Embryonic stem cell ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,medicine ,biology.protein ,Stem cell ,Induced pluripotent stem cell ,Carcinogenesis ,Reprogramming ,030304 developmental biology - Abstract
Reliable approaches to identify stem cell mechanisms that mediate aggressive cancer could have great therapeutic value, based on the growing evidence of embryonic signatures in metastatic cancers. However, how to best identify and target stem-like mechanisms aberrantly acquired by cancer cells has been challenging. We harnessed the power of reprogramming to examine GRP78, a chaperone protein generally restricted to the endoplasmic reticulum in normal tissues, but which is expressed on the cell surface of human embryonic stem cells and many cancer types. We have discovered that (1) cell surface GRP78 (sGRP78) is expressed on iPSCs and is important in reprogramming, (2) sGRP78 promotes cellular functions in both pluripotent and breast cancer cells (3) overexpression of GRP78 in breast cancer cells leads to an induction of a CD24-/CD44+ tumor initiating cell (TIC) population (4) sGRP78+ breast cancer cells are enriched for stemness genes and appear to be a subset of TICs (5) sGRP78+ breast cancer cells show an enhanced ability to seed metastatic organ sites in vivo. These collective findings show that GRP78 has important functions in regulating both pluripotency and oncogenesis, and suggest that sGRP78 marks a stem-like population in breast cancer cells that has increased metastatic potential in vivo.
- Published
- 2019
21. The Activin Receptor, Activin-Like Kinase 4, Mediates Toxoplasma Gondii Activation of Hypoxia Inducible Factor-1
- Author
-
Joan Vaughan, Peter C. Gray, Agnieszka Lis, Mandi Wiley, and Ira J. Blader
- Subjects
0301 basic medicine ,Microbiology (medical) ,parasite - host interactions ,030106 microbiology ,Immunology ,lcsh:QR1-502 ,tgf-beta signaling ,SMAD ,Microbiology ,lcsh:Microbiology ,03 medical and health sciences ,Mice ,Cellular and Infection Microbiology ,TGF beta signaling pathway ,Animals ,Humans ,Kinase activity ,Transcription factor ,Cells, Cultured ,Original Research ,biology ,Kinase ,hypoxia ,Intracellular parasite ,Toxoplasma gondii ,Activin receptor ,biology.organism_classification ,Cell biology ,030104 developmental biology ,Infectious Diseases ,Host-Pathogen Interactions ,transcripional regulation ,Hypoxia-Inducible Factor 1 ,toxoplasma and toxoplasmosis ,Activin Receptors, Type I ,Toxoplasma - Abstract
To grow and cause disease, intracellular pathogens modulate host cell processes. Identifying these processes as well as the mechanisms used by the pathogens to manipulate them is important for the development of more effective therapeutics. As an example, the intracellular parasite Toxoplasma gondii induces a wide variety of changes to its host cell, including altered membrane trafficking, cytoskeletal reorganization, and differential gene expression. Although several parasite molecules and their host targets have been identified that mediate- these changes, few are known to be required for parasite replication. One exception is the host cell transcription factor, hypoxia-inducible factor-1 (HIF-1), which is required for parasite replication in an oxygen-dependent manner. Toxoplasma activates HIF-1 by stabilizing the HIF-1α subunit, and this is dependent on the signaling from the Activin-Like Kinase (ALK) receptor superfamily. Here, we demonstrate that specific overexpression of the ALK family member, ALK4, increased HIF-1 activity in Toxoplasma-infected cells, and this increase required ALK4 kinase activity. Moreover, Toxoplasma stimulated ALK4 to dimerize with its co-receptor, ActRII, and also increased ALK4 kinase activity, thereby demonstrating that Toxoplasma activates the ALK4 receptor. ALK4 activation of HIF-1 was independent of canonical SMAD signaling but rather was dependent on the non-canonical Rho GTPase and JNK MAP kinase signaling pathways. Finally, Toxoplasma increased rates of ALK4 ubiquitination and turnover. These data provide the first evidence indicating that ALK4 signaling is a target for a microbial pathogen to manipulate its host cell.
- Published
- 2019
22. Preoperative plasma fatty acid metabolites inform risk of prostate cancer progression and may be used for personalized patient stratification
- Author
-
Martin Spahn, Martina Minoli, Peter C. Gray, Salvatore Piscuoglio, Anne Wehrhan, Cédric Bovet, Charlotte K.Y. Ng, Eugenio Zoni, Marianna Kruithof-de Julio, and George N. Thalmann
- Subjects
0301 basic medicine ,Oncology ,Male ,Cancer Research ,Metabolite ,Medizin ,Mass Spectrometry ,chemistry.chemical_compound ,Prostate cancer ,0302 clinical medicine ,Surgical oncology ,Lymph node ,chemistry.chemical_classification ,Fatty Acids ,Middle Aged ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Prognosis ,3. Good health ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Dihydrotestosterone ,medicine.drug ,Research Article ,Acylcarnitines ,medicine.medical_specialty ,medicine.drug_class ,610 Medicine & health ,lcsh:RC254-282 ,White People ,03 medical and health sciences ,Internal medicine ,Carnitine ,Genetics ,medicine ,Metabolomics ,Humans ,Aged ,Disease progression ,Fatty acid metabolism ,business.industry ,Fatty acid ,Prostatic Neoplasms ,Androgen ,medicine.disease ,030104 developmental biology ,chemistry ,Case-Control Studies ,business ,Chromatography, Liquid - Abstract
Background Little is known about the relationship between the metabolite profile of plasma from pre-operative prostate cancer (PCa) patients and the risk of PCa progression. In this study we investigated the association between pre-operative plasma metabolites and risk of biochemical-, local- and metastatic-recurrence, with the aim of improving patient stratification. Methods We conducted a case-control study within a cohort of PCa patients recruited between 1996 and 2015. The age-matched primary cases (n = 33) were stratified in low risk, high risk without progression and high risk with progression as defined by the National Comprehensive Cancer Network. These samples were compared to metastatic (n = 9) and healthy controls (n = 10). The pre-operative plasma from primary cases and the plasma from metastatic patients and controls were assessed with untargeted metabolomics by LC-MS. The association between risk of progression and metabolite abundance was calculated using multivariate Cox proportional-hazard regression and the relationship between metabolites and outcome was calculated using median cut-off normalized values of metabolite abundance by Log-Rank test using the Kaplan Meier method. Results Medium-chain acylcarnitines (C6-C12) were positively associated with the risk of PSA progression (p = 0.036, median cut-off) while long-chain acylcarnitines (C14-C16) were inversely associated with local (p = 0.034) and bone progression (p = 0.0033). In primary cases, medium-chain acylcarnitines were positively associated with suberic acid, which also correlated with the risk of PSA progression (p = 0.032, Log-Rank test). In the metastatic samples, this effect was consistent for hexanoylcarnitine, L.octanoylcarnitine and decanoylcarnitine. Medium-chain acylcarnitines and suberic acid displayed the same inverse association with tryptophan, while indoleacetic acid, a breakdown product of tryptophan metabolism was strongly associated with PSA (p = 0.0081, Log-Rank test) and lymph node progression (p = 0.025, Log-Rank test). These data were consistent with the increased expression of indoleamine 2,3 dioxygenase (IDO1) in metastatic versus primary samples (p = 0.014). Finally, functional experiments revealed a synergistic effect of long chain fatty acids in combination with dihydrotestosterone administration on the transcription of androgen responsive genes. Conclusions This study strengthens the emerging link between fatty acid metabolism and PCa progression and suggests that measuring levels of medium- and long-chain acylcarnitines in pre-operative patient plasma may provide a basis for improving patient stratification.
- Published
- 2019
23. CRIPTO promotes an aggressive tumour phenotype and resistance to treatment in hepatocellular carcinoma
- Author
-
Sofia, Karkampouna, Danny, van der Helm, Peter C, Gray, Lanpeng, Chen, Irena, Klima, Joël, Grosjean, Mark C, Burgmans, Arantza, Farina-Sarasqueta, Ewa B, Snaar-Jagalska, Deborah M, Stroka, Luigi, Terracciano, Bart, van Hoek, Alexander F, Schaapherder, Susan, Osanto, George N, Thalmann, Hein W, Verspaget, Minneke J, Coenraad, and Marianna, Kruithof-de Julio
- Subjects
Male ,Carcinoma, Hepatocellular ,Epithelial-Mesenchymal Transition ,Antineoplastic Agents ,GPI-Linked Proteins ,Tissue Culture Techniques ,Cell Movement ,Mice, Inbred NOD ,Animals ,Humans ,Endoplasmic Reticulum Chaperone BiP ,Protein Kinase Inhibitors ,Zebrafish ,Aged ,Cell Proliferation ,Aged, 80 and over ,Antibiotics, Antineoplastic ,Liver Neoplasms ,Hep G2 Cells ,Middle Aged ,Sorafenib ,Xenograft Model Antitumor Assays ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,Phenotype ,Doxorubicin ,Drug Resistance, Neoplasm ,Neoplastic Stem Cells ,Intercellular Signaling Peptides and Proteins ,Female ,Peptides ,Signal Transduction - Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related death worldwide. Despite increasing treatment options for this disease, prognosis remains poor. CRIPTO (TDGF1) protein is expressed at high levels in several human tumours and promotes oncogenic phenotype. Its expression has been correlated to poor prognosis in HCC. In this study, we aimed to elucidate the basis for the effects of CRIPTO in HCC. We investigated CRIPTO expression levels in three cohorts of clinical cirrhotic and HCC specimens. We addressed the role of CRIPTO in hepatic tumourigenesis using Cre-loxP-controlled lentiviral vectors expressing CRIPTO in cell line-derived xenografts. Responses to standard treatments (sorafenib, doxorubicin) were assessed directly on xenograft-derived ex vivo tumour slices. CRIPTO-overexpressing patient-derived xenografts were established and used for ex vivo drug response assays. The effects of sorafenib and doxorubicin treatment in combination with a CRIPTO pathway inhibitor were tested in ex vivo cultures of xenograft models and 3D cultures. CRIPTO protein was found highly expressed in human cirrhosis and hepatocellular carcinoma specimens but not in those of healthy participants. Stable overexpression of CRIPTO in human HepG2 cells caused epithelial-to-mesenchymal transition, increased expression of cancer stem cell markers, and enhanced cell proliferation and migration. HepG2-CRIPTO cells formed tumours when injected into immune-compromised mice, whereas HepG2 cells lacking stable CRIPTO overexpression did not. High-level CRIPTO expression in xenograft models was associated with resistance to sorafenib, which could be modulated using a CRIPTO pathway inhibitor in ex vivo tumour slices. Our data suggest that a subgroup of CRIPTO-expressing HCC patients may benefit from a combinatorial treatment scheme and that sorafenib resistance may be circumvented by inhibition of the CRIPTO pathway. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John WileySons, Ltd.
- Published
- 2017
24. CRIPTO and its signaling partner GRP78 drive the metastatic phenotype in human osteotropic prostate cancer
- Author
-
Peter Kloen, Sofia Karkampouna, Rob C.M. Pelger, G J L H van Leenders, Lijkele Beimers, Zoraide Granchi, E Snaar-Jagalska, F. La Manna, Lanpeng Chen, M D Henry, Eugenio Zoni, Esther I. Verhoef, Peter C. Gray, M. Kruithof-De Julio, G. van der Pluijm, Jonathan A. Kelber, Pathology, Other departments, APH - Personalized Medicine, APH - Quality of Care, Orthopedic Surgery and Sports Medicine, AMS - Restoration & Development, AMS - Ageing & Morbidty, and Other Research
- Subjects
0301 basic medicine ,Male ,Cancer Research ,Mice, Nude ,Bone Neoplasms ,Kaplan-Meier Estimate ,Biology ,Cripto ,GPI-Linked Proteins ,Molecular oncology ,Metastasis ,03 medical and health sciences ,Prostate cancer ,Mice ,Growth factor receptor ,SDG 3 - Good Health and Well-being ,Cell Movement ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,Endoplasmic Reticulum Chaperone BiP ,Heat-Shock Proteins ,Cell Proliferation ,Cancer ,Bone metastasis ,Prostatic Neoplasms ,medicine.disease ,Neoplasm Proteins ,030104 developmental biology ,Gene Knockdown Techniques ,Immunology ,Cancer cell ,Cancer research ,Intercellular Signaling Peptides and Proteins ,Original Article ,hormones, hormone substitutes, and hormone antagonists ,Neoplasm Transplantation - Abstract
CRIPTO (CR-1, TDGF1) is a cell surface/secreted oncoprotein actively involved in development and cancer. Here, we report that high expression of CRIPTO correlates with poor survival in stratified risk groups of prostate cancer (PCa) patients. CRIPTO and its signaling partner glucose-regulated protein 78 (GRP78) are highly expressed in PCa metastases and display higher levels in the metastatic ALDH(high) sub-population of PC-3M-Pro4Luc2 PCa cells compared with non-metastatic ALDH(low). Coculture of the osteotropic PC-3M-Pro4Luc2 PCa cells with differentiated primary human osteoblasts induced CRIPTO and GRP78 expression in cancer cells and increases the size of the ALDH(high) sub-population. Additionally, CRIPTO or GRP78 knockdown decreases proliferation, migration, clonogenicity and the size of the metastasis-initiating ALDH(high) sub-population. CRIPTO knockdown reduces the invasion of PC-3M-Pro4Luc2 cells in zebrafish and inhibits bone metastasis in a preclinical mouse model. These results highlight a functional role for CRIPTO and GRP78 in PCa metastasis and suggest that targeting CRIPTO/GRP78 signaling may have significant therapeutic potential.
- Published
- 2017
25. An Activin A/BMP2 Chimera, AB204, Displays Bone-Healing Properties Superior to Those of BMP2
- Author
-
Sang Kyu Ye, Senyon Choe, Chihoon Ahn, Byung Hak Yoon, Witek Kwiatkowski, Luis Esquivies, and Peter C. Gray
- Subjects
medicine.medical_specialty ,animal structures ,Chemistry ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,fungi ,Antagonist ,Bone healing ,Bone morphogenetic protein 2 ,Cell biology ,Chimera (genetics) ,Endocrinology ,Cytokine ,Internal medicine ,embryonic structures ,medicine ,Potency ,Orthopedics and Sports Medicine ,Noggin ,Receptor - Abstract
Recombinant bone morphogenetic protein 2 (rhBMP2) has been used clinically to treat bone fractures in human patients. However, the high doses of rhBMP2 required for a therapeutic response can cause undesirable side effects. Here, we demonstrate that a novel Activin A/BMP2 (AB2) chimera, AB204, promotes osteogenesis and bone healing much more potently and effectively than rhBMP2. Remarkably, 1 month of AB204 treatment completely heals tibial and calvarial defects of critical size in mice at a concentration 10-fold lower than a dose of rhBMP2 that only partially heals the defect. We determine the structure of AB204 to 2.3 A that reveals a distinct BMP2-like fold in which the Activin A sequence segments confer insensitivity to the BMP2 antagonist Noggin and an affinity for the Activin/BMP type II receptor ActRII that is 100-fold greater than that of BMP2. The structure also led to our identification of a single Activin A-derived amino acid residue, which, when mutated to the corresponding BMP2 residue, resulted in a significant increase in the affinity of AB204 for its type I receptor BMPRIa and a further enhancement in AB204's osteogenic potency. Together, these findings demonstrate that rationally designed AB2 chimeras can provide BMP2 substitutes with enhanced potency for treating non-union bone fractures.
- Published
- 2014
26. Role of Activin-A and Myostatin and Their Signaling Pathway in Human Myometrial and Leiomyoma Cell Function
- Author
-
Milijana Janjusevic, Mario Castellucci, Stefano Raffaele Giannubilo, William H. Catherino, Pasquapina Ciarmela, Andrea Ciavattini, Andrea L. Tranquilli, Soriful Islam, Pasquale Lamanna, Felice Petraglia, Peter C. Gray, and Olga Protic
- Subjects
Adult ,medicine.medical_specialty ,Cell signaling ,Endocrinology, Diabetes and Metabolism ,Clinical Biochemistry ,Immunocytochemistry ,Context (language use) ,Smad2 Protein ,Myostatin ,Biochemistry ,Collagen Type I ,Cell Line ,Endocrinology ,Internal medicine ,medicine ,Humans ,Smad3 Protein ,Uterine Neoplasm ,Cell Proliferation ,Tumor ,Cultured ,Uterine leiomyoma ,Leiomyoma ,biology ,Biochemistry (medical) ,Myometrium ,Middle Aged ,musculoskeletal system ,medicine.disease ,Activins ,Fibronectins ,Tumor Cells ,Cell Line, Tumor ,Female ,Signal Transduction ,Tumor Cells, Cultured ,Uterine Neoplasms ,biology.protein - Abstract
Context: Uterine leiomyomas are highly prevalent benign tumors of premenopausal women and the most common indication for hysterectomy. However, the exact etiology of this tumor is not fully understood. Objective: The objective of the study was to evaluate the role of activin-A and myostatin and their signaling pathways in human myometrial and leiomyoma cells. Design: This was a laboratory study. Setting: Myometrial and leiomyoma cells (primary and cell lines) were cultured in vitro. Patients: The study included premenopausal women who were admitted to the hospital for myomectomy or hysterectomy. Interventions: Primary myometrial and leiomyoma cells and/or cell lines were treated with activin-A (4 nM) and myostatin (4 nM) for different days of interval (to measure proliferation rate) or 30 minutes (to measure signaling molecules) or 48 hours to measure proliferating markers, extracellular matrix mRNA, and/or protein expression by real-time PCR, Western blot, and/or immunocytochemistry. Results: We found th...
- Published
- 2014
27. CRIPTO/GRP78 Signaling Maintains Fetal and Adult Mammary Stem Cells Ex Vivo
- Author
-
Jonathan A. Kelber, Justin La, Rose Rodewald, Evan Booker, Geoffrey M. Wahl, Julia Lipianskaya, Madhuri Kalathur, Benjamin T. Spike, Marielle He, Tracy Wright, Richard L. Klemke, and Peter C. Gray
- Subjects
Cellular differentiation ,Mammary gland ,Gene Expression ,Biology ,GPI-Linked Proteins ,Cripto ,Biochemistry ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Report ,Genetics ,medicine ,Humans ,Regeneration ,Mammary Glands, Human ,Endoplasmic Reticulum Chaperone BiP ,lcsh:QH301-705.5 ,Cells, Cultured ,Heat-Shock Proteins ,030304 developmental biology ,lcsh:R5-920 ,0303 health sciences ,Stem Cells ,Regeneration (biology) ,Cell Membrane ,Cell Differentiation ,Epithelial Cells ,Cell Biology ,Neoplasm Proteins ,Cell biology ,Adult Stem Cells ,medicine.anatomical_structure ,lcsh:Biology (General) ,Cell culture ,030220 oncology & carcinogenesis ,Mutation ,Intercellular Signaling Peptides and Proteins ,Signal transduction ,Stem cell ,lcsh:Medicine (General) ,Activin Receptors, Type I ,Biomarkers ,hormones, hormone substitutes, and hormone antagonists ,Protein Binding ,Signal Transduction ,Developmental Biology ,Adult stem cell - Abstract
Summary Little is known about the extracellular signaling factors that govern mammary stem cell behavior. Here, we identify CRIPTO and its cell-surface receptor GRP78 as regulators of stem cell behavior in isolated fetal and adult mammary epithelial cells. We develop a CRIPTO antagonist that promotes differentiation and reduces self-renewal of mammary stem cell-enriched populations cultured ex vivo. By contrast, CRIPTO treatment maintains the stem cell phenotype in these cultures and yields colonies with enhanced mammary gland reconstitution capacity. Surface expression of GRP78 marks CRIPTO-responsive, stem cell-enriched fetal and adult mammary epithelial cells, and deletion of GRP78 from adult mammary epithelial cells blocks their mammary gland reconstitution potential. Together, these findings identify the CRIPTO/GRP78 pathway as a developmentally conserved regulator of fetal and adult mammary stem cell behavior ex vivo, with implications for the stem-like cells found in many cancers., Graphical Abstract, Highlights • CRIPTO/GRP78 signaling activates PI3K/AKT in fetal mammary epithelial cells ex vivo • Cell-surface GRP78 marks a CRIPTO-responsive adult mammary stem cell population • An antagonist, ALK4L75A-Fc, blocks soluble CRIPTO growth-factor-like activity • CRIPTO promotes and ALK4L75A-Fc inhibits mammary stem cell maintenance ex vivo, Little is known about the niche factors governing mammary stem cell function and if they are conserved throughout mammary gland development and homeostasis. Here, Gray and colleagues identify CRIPTO as a stem cell maintenance factor in fetal and adult mammary stem/progenitor cells and show that its cell-surface receptor, GRP78, is a mammary stem cell marker and determinant of CRIPTO responsiveness.
- Published
- 2014
28. Designer Nodal/BMP2 Chimeras Mimic Nodal Signaling, Promote Chondrogenesis, and Reveal a BMP2-like Structure
- Author
-
Chihoon Ahn, Peter C. Gray, Macarena Perán, Concepción Rodríguez-Esteban, Evan Booker, Juan Carlos Izpisua Belmonte, Alissa Blackler, Witek Kwiatkowski, Luis Esquivies, and Senyon Choe
- Subjects
Adult ,Nodal Protein ,Recombinant Fusion Proteins ,Bone Morphogenetic Protein 2 ,Nodal signaling ,Smad2 Protein ,Biology ,GPI-Linked Proteins ,Cripto ,Biochemistry ,Bone morphogenetic protein 2 ,Cell Line ,parasitic diseases ,Humans ,Heart looping ,Molecular Biology ,Stem Cells ,food and beverages ,Lefty ,Cell Biology ,Chondrogenesis ,Neoplasm Proteins ,Cell biology ,carbohydrates (lipids) ,Cartilage ,Adipose Tissue ,Intercellular Signaling Peptides and Proteins ,Female ,Signal transduction ,NODAL ,Signal Transduction - Abstract
Nodal, a member of the TGF-β superfamily, plays an important role in vertebrate and invertebrate early development. The biochemical study of Nodal and its signaling pathway has been a challenge, mainly because of difficulties in producing the protein in sufficient quantities. We have developed a library of stable, chemically refoldable Nodal/BMP2 chimeric ligands (NB2 library). Three chimeras, named NB250, NB260, and NB264, show Nodal-like signaling properties including dependence on the co-receptor Cripto and activation of the Smad2 pathway. NB250, like Nodal, alters heart looping during the establishment of embryonic left-right asymmetry, and both NB250 and NB260, as well as Nodal, induce chondrogenic differentiation of human adipose-derived stem cells. This Nodal-induced differentiation is shown to be more efficient than BPM2-induced differentiation. Interestingly, the crystal structure of NB250 shows a backbone scaffold similar to that of BMP2. Our results show that these chimeric ligands may have therapeutic implications in cartilage injuries.
- Published
- 2014
29. Cripto/Grp78 drive the metastatic phenotype in human osteotropic prostate cancer
- Author
-
Peter C. Gray, Rob C.M. Pelger, Lanpeng Chen, Julio Marianna Kruithof-de, Zoraide Granchi, Manna Federico La, Leenders Geert van, Eugenio Zoni, Ewa Snaar-Jagalska, Peter Kloen, der Pluijm Gabri van, Sofia Karkampouna, Ester Verhoef, and Lijkele Beimers
- Subjects
Oncology ,Prostate cancer ,medicine.medical_specialty ,business.industry ,Internal medicine ,Metastatic phenotype ,medicine ,General Medicine ,medicine.disease ,business ,Cripto - Published
- 2016
30. Cripto/GRP78 modulation of the TGF-β pathway in development and oncogenesis
- Author
-
Wylie Vale and Peter C. Gray
- Subjects
TGF-β ,GRP78 ,MAPK/ERK pathway ,Plasticity ,Biophysics ,GPI-Linked Proteins ,Cripto ,Models, Biological ,Biochemistry ,Article ,Teratocarcinoma-derived growth factor 1 ,Mice ,Transforming Growth Factor beta ,Structural Biology ,Neoplasms ,Genetics ,Animals ,Humans ,Endoplasmic Reticulum Chaperone BiP ,Molecular Biology ,Protein kinase B ,Heat-Shock Proteins ,PI3K/AKT/mTOR pathway ,Cancer ,Stem cell ,biology ,Stem Cells ,Cell Membrane ,Gene Expression Regulation, Developmental ,Cell Biology ,Transforming growth factor beta ,Neoplasm Proteins ,Protein Structure, Tertiary ,Cell biology ,Gene Expression Regulation, Neoplastic ,biology.protein ,Intercellular Signaling Peptides and Proteins ,Signal transduction ,hormones, hormone substitutes, and hormone antagonists ,Signal Transduction ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Cripto is a small, GPI-anchored signaling protein that regulates cellular survival, proliferation, differentiation and migration during normal developmental processes and tumorigenesis. Cripto functions as an obligatory co-receptor for the TGF-β ligands Nodal, GDF1 and GDF3 but attenuates signaling of others such as activin-A, activin-B and TGF-β1. Soluble, secreted forms of Cripto also activate Src, ras/raf/MAPK and PI3K/Akt pathways via a mechanism that remains largely obscure. This review describes the biological roles and signaling mechanisms of Cripto, highlighting our identification of the 78 kDa glucose regulated protein (GRP78) as a cell surface receptor/co-factor required for Cripto signaling via both TGF-β and Src/MAPK/PI3K pathways. We discuss emerging evidence indicating that Cripto/GRP78 signaling regulates normal somatic stem cells and their tumorigenic counterparts.
- Published
- 2012
31. Possible role of RKIP in cytotrophoblast migration: immunohistochemical and in vitro studies
- Author
-
Luigi Terracciano, Teresa Lorenzi, Pasquapina Ciarmela, Tullia Todros, Felice Petraglia, Md. Soriful Islam, Daniela Marzioni, Mario Castellucci, and Peter C. Gray
- Subjects
MAPK/ERK pathway ,medicine.medical_specialty ,G-Protein-Coupled Receptor Kinase 2 ,Physiology ,Placenta ,Clinical Biochemistry ,Cell ,Phosphatidylethanolamine Binding Protein ,RKIP ,cytotrophoblast migration ,Biology ,Cell Line ,Syncytiotrophoblast ,Pre-Eclampsia ,Cell Movement ,Pregnancy ,Internal medicine ,Plasminogen Activator Inhibitor 1 ,medicine ,Humans ,Oxazolidinones ,reproductive and urinary physiology ,Cytotrophoblast ,Kinase ,NF-kappa B ,Cell migration ,Cell Biology ,Trophoblasts ,Cell biology ,medicine.anatomical_structure ,Endocrinology ,Cell culture ,embryonic structures ,Female ,Mitogen-Activated Protein Kinases ,Immunostaining ,Signal Transduction - Abstract
Raf kinase inhibitor protein (RKIP) regulates growth and differentiation signaling of mitogen-activated protein kinases (MAPK), GRK2 and NF-kappaB pathways each of which regulates cytotrophoblast differentiation and normal placental development. We show here that RKIP is expressed in human normal and preeclampic placentas as detected by immunostaining. RKIP was detected in villous cytotrophoblast in normal placenta and switched to syncytiotrophoblast in pre-eclampsia (PE)-complicated pregnancies. RKIP was also localized in extravillous cytotrophoblast of cell islands and cell columns both in normal and in PE placentas, although staining was less uniform in the latter specimens. In order to test RKIP involvement in cytotrophoblast function, we performed in vitro studies on HTR-8/SVneo cells, a first trimester cytotrophoblast cell line. We show that the RKIP inhibitor locostatin reduces ERK phosphorylation and impairs HTR-8/SV neo cells motility in wound closure experiments. We also document the presence of GRK2 mRNA, the reduction of phosphorylated RKIP expression by locostatin and the induction of PAI mRNA expression in HTR-8/SV neo cells, suggesting the involvement of GRK2 and NF-kappaB pathways in these cells. In conclusion, our work provides evidence that RKIP is a novel factor expressed in cytotrophoblast cells where it likely regulates cell migration.
- Published
- 2012
32. Growth factors and myometrium: biological effects in uterine fibroid and possible clinical implications
- Author
-
Enrrico Bloise, Fernando M. Reis, Peter C. Gray, Mario Castellucci, Md. Soriful Islam, Felice Petraglia, Wylie Vale, and Pasquapina Ciarmela
- Subjects
medicine.medical_specialty ,Platelet-derived growth factor ,medicine.medical_treatment ,Basic fibroblast growth factor ,Reviews ,Biology ,Models, Biological ,Paracrine signalling ,chemistry.chemical_compound ,Growth factors ,Leiomyoma ,Myometrium ,Steroid hormones ,Pregnancy ,Epidermal growth factor ,Internal medicine ,medicine ,Humans ,Gonadal Steroid Hormones ,Growth Substances ,Growth factor ,Obstetrics and Gynecology ,Vascular endothelial growth factor A ,Endocrinology ,Reproductive Medicine ,chemistry ,Uterine Neoplasms ,biology.protein ,Female ,Platelet-derived growth factor receptor ,Signal Transduction ,Transforming growth factor - Abstract
Background Growth factors are proteins secreted by a number of cell types that are capable of modulating cellular growth, proliferation and cellular differentiation. It is well accepted that uterine cellular events such as proliferation and differentiation are regulated by sex steroids and their actions in target tissues are mediated by local production of growth factors acting through paracrine and/or autocrine mechanisms. Myometrial mass is ultimately modified in pregnancy as well as in tumour conditions such as leiomyoma and leiomyosarcoma. Leiomyomas, also known as fibroids, are benign tumours of the uterus, considered to be one of the most frequent causes of infertility in reproductive years in women. Methods For this review, we searched the database MEDLINE and Google Scholar for articles with content related to growth factors acting on myometrium; the findings are hereby reviewed and discussed. Results Different growth factors such as epidermal growth factor (EGF), transforming growth factor-α (TGF-α), heparin-binding EGF (HB-EGF), acidic fibroblast growth factor (aFGF), basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF), insulin-like growth factor (IGF), platelet-derived growth factor (PDGF) and TGF-β perform actions in myometrium and in leiomyomas. In addition to these growth factors, activin and myostatin have been recently identified in myometrium and leiomyoma. Conclusions Growth factors play an important role in the mechanisms involved in myometrial patho-physiology.
- Published
- 2011
33. Abstract B060: Molecular characterization of cancer stem cells from patient-derived xenografts of advanced prostate cancer
- Author
-
Sofia Karkampouna, Markus Germann, Marta De Menna, Peter C. Gray, Joel Grosjean, Marianna Kruithof-de Julio, and George N. Thalmann
- Subjects
Cancer Research ,biology ,CD44 ,Cancer ,urologic and male genital diseases ,medicine.disease ,Stem cell marker ,Prostate cancer ,Oncology ,Cancer stem cell ,Cancer cell ,biology.protein ,medicine ,Cancer research ,Stem cell ,Progenitor cell - Abstract
Introduction: Androgen-deprivation therapy is the standard treatment for prostate cancer. Despite the initial response, a fraction of cases manifest progression to castration-resistant prostate cancer. The paradigm of PCa recurrence is the existence of androgen-independent cancer stem cells, which differentiate into androgen-dependent cancer cells that reinitiate tumor growth. The phenotypic properties of cancer stem cells versus highly proliferative progenitor cells/facultative stem cells remain to be further characterized in terms of androgen receptor signaling, quiescence/proliferation, tumorigenicity, and therapy resistance. Methods: To address the molecular basis of CSC switch from androgen-dependency to independency we have employed two patient-derived xenograft models (LAPC-9 and BM-18) that have different androgen sensitivity properties. We have performed microarray and proteomic analysis of BM-18 tumor tissues prior to and following castration as well as androgen replacement. To characterize the cancer stem cells in these models we have isolated different subpopulations based on combination of selected markers and assessed the transcriptomic changes, proteomic profile, and in vivo self-renewal. Results: Castration induces a rapid tumor volume decrease in the BM-18 and a stabilization of tumor burden in the LAPC-9, reflecting different androgen-dependent cell states. Proteomic analysis of bulk BM-18 tumors (intact, 14 days post castration, 50 days post androgen replacement) indicates an enrichment of stem cell markers upon castration; CD44, NKX3.1, and ALDH1 isoforms. Microarray analysis has confirmed upregulation of CD44 and ALDH1A1 at castrated state, and downregulation of expression upon androgen replacement at early time points (24 hours). We have isolated CD44+/-ALDHhigh/low subpopulation by flow cytometry and analyzed their transcriptome and proteomic changes. Castration in the BM-18 model induces an increase in the subpopulations of CD44+/ALDHlow (from 0.44% to 2.3%) and CD44+/ALDHhigh (from 0.06% to 0.38%). However, the same subpopulations are decreased in the LAPC-9 model upon castration, while only the CD44-/ALDHhigh subset is enriched (from 3.5% to 7.1%). Conclusions: Different androgen-independent cancer stem cell subpopulations may be distinguished by the ALDH activity status in combination with CD44. The androgen-independent cells in the BM-18 androgen-dependent model are reflected by enrichment of CD44 expression, as well as a rare double positive population. In the androgen-independent LAPC-9 model, CD44 expression is contrastingly decreased, potentially reflecting androgen-dependent CD44+ cells, and ALDH activity increased in CD44- cells. Ongoing analysis may elucidate the molecular mechanisms controlling the different cancer stem cell fates and their androgen sensitivity. Cancer stem cell potential of the different cell populations will have to be elucidated by transplantation experiments. Citation Format: Sofia Karkampouna, Marta de Menna, Markus Germann, Jöel Grosjean, Peter C. Gray, George N. Thalmann, Marianna Kruithof-de Julio. Molecular characterization of cancer stem cells from patient-derived xenografts of advanced prostate cancer [abstract]. In: Proceedings of the AACR Special Conference: Prostate Cancer: Advances in Basic, Translational, and Clinical Research; 2017 Dec 2-5; Orlando, Florida. Philadelphia (PA): AACR; Cancer Res 2018;78(16 Suppl):Abstract nr B060.
- Published
- 2018
34. Abstract 1990: Aberrant cell surface expression of GRP78 in breast cancer cells marks a stem-like population that has increased metastatic potential in vivo
- Author
-
Ian H. Guldner, Peter C. Gray, Jonathan A. Kelber, Siyuan Zhang, Sergio Ruiz, Athanasia D. Panopoulos, Amanda E. Yamasaki, Yuriko Hishida, Henry C. Conner, Michael Z. Wu, Juan Carlos Izpisua Belmonte, Tyson W. Lager, and Tomoaki Hishida
- Subjects
Cancer Research ,CD44 ,Cancer ,Biology ,medicine.disease_cause ,medicine.disease ,Embryonic stem cell ,Oncology ,Cancer cell ,medicine ,Cancer research ,biology.protein ,Stem cell ,Induced pluripotent stem cell ,Carcinogenesis ,Reprogramming - Abstract
Reliable approaches to identify and target stem-cell mechanisms that mediate aggressive cancer could have great therapeutic value, based on the growing evidence of embryonic signatures in metastatic cancers. However, how to best identify and target stem-like mechanisms aberrantly utilized by cancer cells has been challenging. We harnessed the power of induced pluripotent stem cells (iPSCs) to identify embryonic mechanisms exploited by cancer. A screen comparing the cell surface proteome of iPSCs and breast cancer cells identified GRP78, a heat shock protein that is normally ER-restricted, but has been shown to be aberrantly expressed on the cell surface of several cancers, where it can act as a signaling molecule by poorly understood mechanisms. Although cell surface GRP78 (sGRP78) has emerged as an attractive chemotherapeutic target, understanding how sGRP78 is functioning in cancer has been complicated by the fact that GRP78 can function to regulate a variety of cellular responses, using a diverse array of reported binding partners, which can vary by cell type. Therefore, without insight into the specific GRP78-dependent mechanisms that are responsible for mediating aggressive cancer, it will be difficult to determine how to best target GRP78. We have discovered that (1) sGRP78 is expressed on iPSCs (but not their somatic parental populations) and plays an important role in reprogramming, (2) sGRP78 promotes cellular functions such as proliferation/survival and migration in both stem cells and breast cancer cells (3) overexpression of GRP78 in breast cancer cells leads to an induction of a previously established CD24-/CD44+ 'cancer stem cell' (CSC) population (4) sGRP78+ breast cancer cell populations are enriched for genes involved in stemness and appear to be a subset of previously established CSCs (5) sGRP78+ breast cancer cell populations show a significantly enhanced ability to seed metastatic organ sites in vivo (6) GRP78 interacts with Dermcidin (DCD) at the cell surface of cancer cells and iPSCs, where it is important in regulating stem cell and cancer cell migration and survival/proliferation. These collective findings suggest that sGRP78 marks a stem-like population in breast cancer cells that has increased metastatic potential in vivo, and that sGRP78 and DCD cooperate to regulate key cellular functions important in mediating tumorigenesis. Overall, this work has implications for understanding how cancer cells exploit embryonic-like mechanisms, which could provide novel strategies for chemotherapeutic targeting of aggressive breast cancer cell populations. Citation Format: Tyson W. Lager, Henry C. Conner, Ian H. Guldner, Michael Z. Wu, Yuriko Hishida, Tomoaki Hishida, Sergio Ruiz, Amanda E. Yamasaki, Juan Carlos Izpisua Belmonte, Peter C. Gray, Jonathan A. Kelber, Siyuan Zhang, Athanasia D. Panopoulos. Aberrant cell surface expression of GRP78 in breast cancer cells marks a stem-like population that has increased metastatic potential in vivo [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 1990.
- Published
- 2018
35. Neutrophil activation in patients with anti-neutrophil cytoplasmic autoantibody-associated vasculitis and large-vessel vasculitis
- Author
-
Despina Michailidou, Bhargavi Duvvuri, Runa Kuley, David Cuthbertson, Peter C. Grayson, Nader A. Khalidi, Curry L. Koening, Carol A. Langford, Carol A. McAlear, Larry W. Moreland, Christian Pagnoux, Philip Seo, Ulrich Specks, Antoine G. Sreih, Kenneth J. Warrington, Tomas Mustelin, Paul A. Monach, Peter A. Merkel, and Christian Lood
- Subjects
Anti-neutrophil cytoplasmic antibody-associated vasculitis ,Large-vessel vasculitis ,Neutrophils ,Mitochondria ,Formyl peptide receptor 1 ,Diseases of the musculoskeletal system ,RC925-935 - Abstract
Abstract Objective To assess markers of neutrophil activation such as calprotectin and N-formyl methionine (fMET) in anti-neutrophil cytoplasmic autoantibody-associated vasculitis (AAV) and large-vessel vasculitis (LVV). Methods Levels of fMET, and calprotectin, were measured in the plasma of healthy controls (n=30) and patients with AAV (granulomatosis with polyangiitis (GPA, n=123), microscopic polyangiitis (MPA, n=61)), and LVV (Takayasu’s arteritis (TAK, n=58), giant cell arteritis (GCA, n=68)), at times of remission or flare. Disease activity was assessed by physician global assessment. In vitro neutrophil activation assays were performed in the presence or absence of formyl peptide receptor 1 (FPR1) inhibitor cyclosporine H. Results Levels of calprotectin, and fMET were elevated in patients with vasculitis as compared to healthy individuals. Levels of fMET correlated with markers of systemic inflammation: C-reactive protein (r=0.82, p
- Published
- 2022
- Full Text
- View/download PDF
36. Activin A/BMP2 chimera AB235 drives efficient redifferentiation of long term cultured autologous chondrocytes
- Author
-
Esmeralda Carrillo, Gema Jiménez, Witek Kwiatkowski, Elvira Montañez, J. C. Izpisua Belmonte, Macarena Perán, Peter C. Gray, Juan A. Marchal, Senyon Choe, Francisco Arrebola, Elena López-Ruiz, [Jiménez,G, Carrillo,E, Marchal,JA] Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, Granada, Spain. Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain. Biosanitary Institute of Granada (ibs.GRANADA), University Hospitals of Granada-Univesity of Granada, Granada, Spain. [López-Ruiz,E, Perán,M] Department of Health Sciences, University of Jaén, Jaén, Spain. [Kwiatkowski,W, Choe,S] Structural Biology Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA. [Montañez,E] Department of Orthopedic Surgery and Traumatology, Virgen de la Victoria University Hospital, Málaga, Spain. [Arrebola,F] Department of Histology, Faculty of Medicine, University of Granada, Granada, Spain. [Gray,PC] Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla CA, California, USA. [Izpisua Belmonte, JC] Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla CA, California, USA. [Choe,S] Qualcomm Institute, Univ. California, San Diego, La Jolla, USA., and This work was supported by the Consejería de Economía, Innovación y Ciencia (Junta de Andalucía, excellence project number CTS-6568).
- Subjects
Cartilage, Articular ,Male ,Time Factors ,Cellular differentiation ,Cell ,Diseases::Musculoskeletal Diseases::Rheumatic Diseases::Osteoarthritis [Medical Subject Headings] ,Chemicals and Drugs::Hormones, Hormone Substitutes, and Hormone Antagonists::Hormones::Gonadal Hormones::Activins [Medical Subject Headings] ,Bone Morphogenetic Protein 2 ,Gene Expression ,Anatomy::Cells::Connective Tissue Cells::Chondrocytes [Medical Subject Headings] ,Anatomy::Tissues::Connective Tissue::Subcutaneous Tissue [Medical Subject Headings] ,Mice, SCID ,Ligands ,Organisms::Eukaryota::Animals::Chordata::Vertebrates::Mammals::Primates::Haplorhini::Catarrhini::Hominidae::Humans [Medical Subject Headings] ,Extracellular matrix ,Mice ,Mice, Inbred NOD ,Organisms::Eukaryota::Animals [Medical Subject Headings] ,Activinas ,Autologous chondrocyte implantation ,Tejido subcutáneo ,Anatomy::Cells::Cellular Structures::Extracellular Space::Extracellular Matrix [Medical Subject Headings] ,Multidisciplinary ,Reverse Transcriptase Polymerase Chain Reaction ,Cell Differentiation ,Middle Aged ,Immunohistochemistry ,Humanos ,Analytical, Diagnostic and Therapeutic Techniques and Equipment::Investigative Techniques::Genetic Techniques::Nucleic Acid Amplification Techniques::Polymerase Chain Reaction::Reverse Transcriptase Polymerase Chain Reaction [Medical Subject Headings] ,Activins ,Extracellular Matrix ,Cell biology ,Chemicals and Drugs::Carbohydrates::Polysaccharides::Proteoglycans [Medical Subject Headings] ,medicine.anatomical_structure ,Anatomy::Musculoskeletal System::Cartilage::Hyaline Cartilage::Cartilage, Articular [Medical Subject Headings] ,Female ,Proteoglycans ,Matriz extracelular ,Collagen ,Cartílago articular ,Proteoglicanos ,Transplantation, Heterologous ,Biology ,Transplantation, Autologous ,Bone morphogenetic protein 2 ,Article ,Chondrocytes ,In vivo ,Osteoarthritis ,Analytical, Diagnostic and Therapeutic Techniques and Equipment::Diagnosis::Diagnostic Techniques and Procedures::Clinical Laboratory Techniques::Cytological Techniques::Cytodiagnosis::Biopsy [Medical Subject Headings] ,medicine ,Animals ,Humans ,Aged ,Organisms::Eukaryota::Animals::Chordata::Vertebrates::Mammals::Rodentia::Muridae::Murinae::Mice [Medical Subject Headings] ,Reacción en cadena de la polimerasa de transcriptasa inversa ,Chemicals and Drugs::Macromolecular Substances::Polymers::Biopolymers::Collagen [Medical Subject Headings] ,Chondrogenesis ,Transplantation ,Biopsia ,Condrocitos ,Osteoartritis ,Animales ,Immunology ,Colágeno - Abstract
Autologous chondrocyte implantation (ACI) depends on the quality and quantity of implanted cells and is hindered by the fact that chondrocytes cultured for long periods of time undergo dedifferentiation. Here we have developed a reproducible and efficient chondrogenic protocol to redifferentiate chondrocytes isolated from osteoarthritis (OA) patients. We used morphological, histological and immunological analysis together with a RT-PCR detection of collagen I and collagen II gene expression to show that chondrocytes isolated from articular cartilage biopsies of patients and subjected to long-term culture undergo dedifferentiation and that these cells can be redifferentiated following treatment with the chimeric Activin A/BMP2 ligand AB235. Examination of AB235-treated cell pellets in both in vitro and in vivo experiments revealed that redifferentiated chondrocytes synthesized a cartilage-specific extracellular matrix (ECM), primarily consisting of vertically-orientated collagen fibres and cartilage-specific proteoglycans. AB235-treated cell pellets also integrated into the surrounding subcutaneous tissue following transplantation in mice as demonstrated by their dramatic increase in size while non-treated control pellets disintegrated upon transplantation. Thus, our findings describe an effective protocol for the promotion of redifferentiation of autologous chondrocytes obtained from OA patients and the formation of a cartilage-like ECM that can integrate into the surrounding tissue in vivo.
- Published
- 2015
37. Cripto-1: an extracellular protein - connecting the sequestered biological dots
- Author
-
Frank Cuttitta, Luigi Strizzi, Malgorzata Klauzinska, Menotti Ruvo, Maria Cristina Rangel, Annalia Focà, Mary J.C. Hendrix, Monica Gonzales, David S. Salomon, Annamaria Sandomenico, Peter C. Gray, Daniel Bertolette, Meg Duroux, Christian Wechselberger, Sudhamsh Tippireddy, and Nadia P. Castro
- Subjects
Epithelial-Mesenchymal Transition ,Biology ,Cripto ,GPI-Linked Proteins ,Biochemistry ,Rheumatology ,Transforming Growth Factor beta ,Humans ,Orthopedics and Sports Medicine ,Epithelial–mesenchymal transition ,Molecular Biology ,Protein kinase B ,Autoantibodies ,Epidermal Growth Factor ,Drug discovery ,Cell growth ,Cell Biology ,Transforming growth factor beta ,Cell biology ,Neoplasm Proteins ,biology.protein ,Intercellular Signaling Peptides and Proteins ,Signal transduction ,Extracellular Space ,Proto-oncogene tyrosine-protein kinase Src ,Signal Transduction - Abstract
Cripto-1 (CR-1) is a multifunctional embryonic protein that is re-expressed during inflammation, wound repair, and malignant transformation. CR-1 can function either as a tethered co-receptor or shed as a free ligand underpinning its flexible role in cell physiology. CR-1 has been shown to mediate cell growth, migration, invasion, and induce epithelial to mesenchymal transition (EMT). The main signaling pathways mediating CR-1 effects include Nodal-dependent (Smad2/3) and Nodal-independent (Src/p44/42/Akt) signaling transduction pathways. In addition, there are several naturally occurring binding partner proteins (BPPs) for CR-1 that can either agonize or antagonize its bioactivity. We will review the collective role of CR-1 as an extracellular protein, discuss caveats to consider in developing a quantitation assay, define possible mechanistic avenues applicable for drug discovery, and report on our experimental approaches to overcome these problematic issues.
- Published
- 2015
38. Identification of Distinct Inhibin and Transforming Growth Factor β-binding Sites on Betaglycan
- Author
-
Wylie Vale, Kathy A. Lewis, Peter C. Gray, Ezra Wiater, and Craig A. Harrison
- Subjects
Genetics ,chemistry.chemical_classification ,endocrine system ,Inhibin binding ,Co-receptor ,biology ,Chemistry ,Mutant ,Cell Biology ,Transforming growth factor beta ,Biochemistry ,Cell biology ,Amino acid ,Extracellular ,biology.protein ,Binding site ,Molecular Biology ,hormones, hormone substitutes, and hormone antagonists ,Transforming growth factor - Abstract
Betaglycan is a co-receptor that mediates signaling by transforming growth factor beta (TGFbeta) superfamily members, including the distinct and often opposed actions of TGFbetas and inhibins. Loss of betaglycan expression, or abrogation of betaglycan function, is implicated in several human and animal diseases, although both betaglycan actions and the ligands involved in these disease states remain unclear. Here we identify a domain spanning amino acids 591-700 of the betaglycan extracellular domain as the only inhibin-binding region in betaglycan. This binding site is within the betaglycan ZP domain, but inhibin binding is not integral to the ZP motif of other proteins. We show that the inhibin and TGFbeta-binding residues of this domain overlap and identify individual amino acids essential for binding of each ligand. Mutation of Val614 to Tyr abolishes both inhibin and TGFbeta binding to this domain. Full-length betaglycan V614Y, and other mutations, retain TGFbeta binding activity via a distinct site, but are unable to bind inhibin-A. These betaglycan mutants fail to mediate inhibin antagonism of activin signaling but can present TGFbeta to TbetaRII. Separating the co-receptor actions of betaglycan toward inhibin and TGFbeta will allow the clarification of the role of betaglycan in disease states such as renal cell carcinoma and endometrial adenocarcinoma.
- Published
- 2006
39. Antagonists of activin signaling: mechanisms and potential biological applications
- Author
-
Peter C. Gray, Craig A. Harrison, Wylie Vale, and David Robertson
- Subjects
endocrine system ,medicine.medical_specialty ,Cachexia ,animal structures ,Endocrinology, Diabetes and Metabolism ,Biology ,ACVR1 ,Cripto ,Hormone Antagonists ,Endocrinology ,Neoplasms ,Internal medicine ,TGF beta signaling pathway ,medicine ,Animals ,Humans ,Activin type 2 receptors ,Wound Healing ,Activins ,Cell biology ,embryonic structures ,biology.protein ,BAMBI ,Signal transduction ,hormones, hormone substitutes, and hormone antagonists ,ACVR2B ,Signal Transduction ,Follistatin - Abstract
Activins are members of the transforming growth factor-beta (TGF-beta) superfamily that control many physiological processes such as cell proliferation and differentiation, immune responses, wound repair and various endocrine activities. Activins elicit these diverse biological responses by signaling via type I and type II receptor serine kinases. Recent studies have revealed details of the roles of inhibin, betaglycan, follistatin and its related protein follistatin-related gene (FLRG), Cripto and BAMBI in antagonizing activin action, and exogenous antagonists against the activin type I (SB-431542 and SB-505124) and type II (activin-M108A) receptors have been developed. Understanding how activin signaling is controlled extracellularly is the first step in providing treatment for wound healing and for disorders such as cachexia and cancer, which result from a deregulated activin pathway.
- Published
- 2005
40. Identification of a Functional Binding Site for Activin on the Type I Receptor ALK4
- Author
-
Steven C. Koerber, Wolfgang H. Fischer, Peter C. Gray, Craig A. Harrison, and Wylie Vale
- Subjects
Molecular Sequence Data ,Receptor, Transforming Growth Factor-beta Type I ,Activin binding ,Gene Expression ,Protein Serine-Threonine Kinases ,Biology ,ACVR1 ,Kidney ,Biochemistry ,Serine ,Structure-Activity Relationship ,TGF beta signaling pathway ,Animals ,Humans ,Receptors, Growth Factor ,Amino Acids ,Binding site ,Lung ,Molecular Biology ,Bone Morphogenetic Protein Receptors, Type I ,Cells, Cultured ,Activin type 2 receptors ,Binding Sites ,Sequence Homology, Amino Acid ,Proteins ,Epithelial Cells ,Cell Biology ,Transmembrane protein ,Activins ,Protein Structure, Tertiary ,Mink ,Mutagenesis ,Activin Receptors, Type I ,Receptors, Transforming Growth Factor beta ,ACVR2B - Abstract
Activins, like other members of the transforming growth factor-beta (TGF-beta) superfamily, initiate signaling by assembling a complex of two types of transmembrane serine/threonine receptor kinases classified as type II (ActRII or ActRIIB) and type I (ALK4). A kinase-deleted version of ALK4 can form an inactive complex with activin and ActRII/IIB and thereby acts in a dominant negative manner to block activin signaling. Using the complex structure of bone morphogenetic protein-2 bound to its type I receptor (ALK3) as a guide, we introduced extracellular domain mutations in the context of the truncated ALK4 (ALK4-trunc) construct and assessed the ability of the mutants to inhibit activin function. We have identified five hydrophobic amino acid residues on the ALK4 extracellular domain (Leu40, Ile70, Val73, Leu75, and Pro77) that, when mutated to alanine, have substantial effects on ALK4-trunc dominant negative activity. In addition, eleven mutants partially affected activin binding to ALK4. Together, these residues likely constitute the binding surface for activin on ALK4. Cross-linking studies measuring binding of 125I-activin-A to the ALK4-trunc mutants in the presence of ActRII implicated the same residues. Our results indicate that there is only a partial overlap of the binding sites on ALK4 and ALK3 for activin-A and bone morphogenetic protein-2, respectively. In addition three of the residues required for activin binding to ALK4 are conserved on the type I TGF-beta receptor ALK5, suggesting the corresponding region on ALK5 may be important for TGF-beta binding.
- Published
- 2003
41. The BMP7/ActRII Extracellular Domain Complex Provides New Insights into the Cooperative Nature of Receptor Assembly
- Author
-
Jay C. Groppe, Peter C. Gray, Wylie Vale, Senyon Choe, Witek Kwiatkowski, Ezra Wiater, and Jason Greenwald
- Subjects
Models, Molecular ,medicine.medical_specialty ,Cytoplasm ,animal structures ,genetic structures ,Protein Conformation ,Activin Receptors, Type II ,Bone Morphogenetic Protein 7 ,Molecular Sequence Data ,Biology ,Bone morphogenetic protein ,Crystallography, X-Ray ,Ligands ,Protein Structure, Secondary ,Cell Line ,Epitopes ,Mice ,Protein structure ,Transforming Growth Factor beta ,Internal medicine ,TGF beta signaling pathway ,medicine ,Escherichia coli ,Animals ,Humans ,Amino Acid Sequence ,Phosphorylation ,Receptor ,Luciferases ,Molecular Biology ,Sequence Homology, Amino Acid ,Activin receptor ,Transforming growth factor beta ,Cell Biology ,Surface Plasmon Resonance ,BMPR2 ,Cell biology ,Extracellular Matrix ,Protein Structure, Tertiary ,Endocrinology ,Cross-Linking Reagents ,embryonic structures ,Bone Morphogenetic Proteins ,biology.protein ,ACVR2B ,Protein Binding - Abstract
Activins and bone morphogenetic proteins (BMPs) elicit diverse biological responses by signaling through two pairs of structurally related type I and type II receptors. Here we report the crystal structure of BMP7 in complex with the extracellular domain (ECD) of the activin type II receptor. Our structure produces a compelling four-receptor model, revealing that the types I and II receptor ECDs make no direct contacts. Nevertheless, we find that truncated receptors lacking their cytoplasmic domain retain the ability to cooperatively assemble in the cell membrane. Also, the affinity of BMP7 for its low-affinity type I receptor ECD increases 5-fold in the presence of its type II receptor ECD. Taken together, our results provide a view of the ligand-mediated cooperative assembly of BMP and activin receptors that does not rely on receptor-receptor contacts.
- Published
- 2003
- Full Text
- View/download PDF
42. Erratum to 'Antagonism of activin by inhibin and inhibin receptors: a functional role for betaglycan-glycan'
- Author
-
Louise M. Bilezikjian, Wylie Vale, and Peter C. Gray
- Subjects
endocrine system ,endocrine system diseases ,Cell growth ,Activin and inhibin ,Cell ,Biology ,Biochemistry ,Cell biology ,Endocrinology ,medicine.anatomical_structure ,medicine ,Signal transduction ,Mode of action ,Receptor ,Molecular Biology ,hormones, hormone substitutes, and hormone antagonists ,Function (biology) ,Hormone - Abstract
Activin and inhibin research has provided important insight into reproductive physiology as well as many areas involving regulation of cell growth, differentiation and function. Progress in understanding the roles of these hormones in various cell and tissue types has been complimented by novel discoveries at the molecular level that have shed light on ligand/receptor interactions, signaling mechanisms and regulation. While the receptors and signaling pathway for activin are now well characterized, the molecular basis for inhibin action has remained relatively unclear. Here we summarize recent advances in understanding inhibin’s mode of action focusing on our recent identification of betaglycan as an inhibin co-receptor capable of mediating inhibin action. © 2001 Elsevier Science Ireland Ltd. All rights reserved.
- Published
- 2002
43. Abstract 4344: Cd146 modulates the malignant phenotype in human prostate cancer
- Author
-
Eugenio Zoni, Gabri van der Pluijm, Peter C. Gray, Marco G. Cecchini, Irena Klima, Janine E. Melsen, Joel Grosjean, Letizia Astrologo, Sofia Karkampouna, Marianna Kruithof-de Julio, and George N. Thalmann
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Internal medicine ,medicine ,Cancer ,CD146 ,medicine.disease ,business ,Human prostate ,Malignant phenotype - Abstract
Prostate Cancer (PCa) is the most frequent cancer in males and the second leading cause of death from cancer in men. When PCa progress from androgen-responsive to castration resistance, the formation of incurable metastases, mainly in the bone, is almost inevitable. Therefore, understanding the factors that regulate homing and survival of metastatic cancer cells in the bone is important for the identification of new therapeutic targets. High CD146 expression has been measured in the stroma of lytic and blastic lesions in preclinical models of PCa bone metastasis. The objective of this study is to characterize the role of CD146 in the maintenance of the aggressive and invasive phenotype in human PCa. We used shRNAs to knockdown the expression of CD146 in the lytic PC-3M-Pro4Luc2dTomato and in the blastic C4-2BdTomato PCa cell lines. We validated the knockdown at protein level and tested the effect with functional assays such as migration, proliferation. We used RT-qPCR to test CD146 knockdown on EMT markers. We measured the effect of the knockdown on the maintenance of cancer stem/progenitor-like cells by ALDEFLUOR assay. CD146 knockdown reduced proliferation in PC-3M-Pro4Luc2dTomato PCa cells and resulted in increased E-Cadherin expression. Conversely, no effect on proliferation was measured on C4-2BdTomato cells. It has been described that metastatic human PCa cells target the hematopoietic stem cell (HSC) niche in the bone marrow at the level of an “endosteal/osteoblast” niche and a “vascular/perivascular” niche. We optimized an in vitro model of “osteoblast niche” to study the behavior of prostate cancer cells upon co-culture with osteoblasts and to measure the resulting effects on cancer stem/progenitor-like markers. Our results showed that CD146 is required for the osteoblast-mediated induction of ALDH activity on PCa cells and CD146 knockdown prevented the increase in the size of the ALDHhigh subpopulation in the tumor cells, mediated by human osteoblasts. Additionally, CD146 knockdown in PCa cells co-cultured with osteoblast, reduced the amount of CD146 expressed by osteoblasts compared to non-targeted control. Finally, we showed that CD146 is significantly increased in the highly metastatic ALDHhigh cells and identified a new subset of ALDHhigh / CD146high cells which could be depleted upon CD146 knockdown. In Conclusion, we detected a novel subset of ALDHhigh/CD146high cells and found that CD146 influences the maintenance of an aggressive-mesenchymal phenotype in human PCa. Therefore, CD146 represents a promising molecule to modulate the behavior of aggressive PCa cells. Note: This abstract was not presented at the meeting. Citation Format: Eugenio Zoni, Letizia Astrologo, Janine Melsen, Sofia Karkampouna, Irena Klima, Joël Grosjean, Peter C. Gray, Gabri van der Pluijm, Marco G. Cecchini, Marianna Kruithof-de Julio, George N. Thalmann. Cd146 modulates the malignant phenotype in human prostate cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 4344. doi:10.1158/1538-7445.AM2017-4344
- Published
- 2017
44. Effects of ALK1Fc treatment on prostate cancer cells interacting with bone and bone cells in bone metastasis models
- Author
-
Martin Spahn, Peter ten Dijke, Joel Grosjean, Marianna Kruithof-de Julio, George N. Thalmann, Irena Klima, Lukas J. A. C. Hawinkels, Marie-José Goumans, Letizia Astrologo, Sofia Karkampouna, Eugenio Zoni, Peter C. Gray, and Gabri van der Pluijm
- Subjects
Cancer Research ,Prostate cancer ,Oncology ,business.industry ,Bone cell ,medicine ,Cancer research ,Bone metastasis ,Cancer ,medicine.disease ,business ,Primary tumor ,Metastasis - Abstract
e16576 Background: Prostate cancer is the second most common cancer in men worldwide. Lethality is normally associated with the consequences of metastasis rather than the primary tumor. In particular, bone is the most frequent site of metastasis and once prostate tumor cells are engrafted in the skeleton, curative therapy is no longer possible. Bone morphogenetic proteins (BMPs) play a critical role in bone physiology and pathology. However, little is known about the role of BMP9 and its signaling receptors, ALK1 and ALK2, in prostate cancer and bone metastasis. In this context, we investigate the impact of BMP9 on primary prostate cancer and derived bone metastasis. Methods: The human ALK1 extracellular domain (ECD) binds BMP9 and BMP10 with high affinity. In order to study the effect of BMP9 in vitro and in vivo we use a soluble chimeric protein, consisting of ALK1 ECD fused to human Fc (ALK1Fc), for preventing the activation of endogenous signaling. ALK1Fc sequesters BMP9 and BMP10, preserving the activation of ALK1 through other ligands. Results: We show that ALK1Fc reduces BMP9-mediated signaling and decreases proliferation of highly metastatic and tumor initiating human prostate cancer cells in vitro. In line with these observations, we demonstrate that ALK1Fc reduces tumor growth in vivo in an orthotopic transplantation model. The propensity of the primary prostate cancer to metastasize to the bone is also investigated. In particular, we report how the ALK1Fc influences the prostate cancer cells in vitro and in vivo when these are probed in different bone settings (co-culture with bone cells and intraosseous transplantation in mice). Conclusions: Our study provides the first demonstration that ALK1Fc inhibits prostate cancer cells growth identifying BMP9 as a putative therapeutic target and ALK1Fc as a potential therapy. All together, these findings justify the ongoing clinical development of drugs blocking ALK1 and ALK2 receptor activity.
- Published
- 2017
45. Engineering TGF-β superfamily ligands for clinical applications
- Author
-
Peter C. Gray, Witek Kwiatkowski, and Senyon Choe
- Subjects
Models, Molecular ,Computational biology ,Toxicology ,medicine.disease_cause ,Crystallography, X-Ray ,Ligands ,Protein Engineering ,Regenerative medicine ,Epitope ,Protein Structure, Secondary ,Structure-Activity Relationship ,Transforming Growth Factor beta ,medicine ,Structure–activity relationship ,Animals ,Humans ,Pharmacology ,Mutation ,biology ,Ligand ,Chemistry ,Protein engineering ,Transforming growth factor beta ,Immunology ,biology.protein ,Tgf β superfamily - Abstract
TGF-β superfamily ligands govern normal tissue development and homeostasis, and their dysfunction is a hallmark of many diseases. These ligands are also well defined both structurally and functionally. This review focuses on TGF-β superfamily ligand engineering for therapeutic purposes, in particular for regenerative medicine and musculoskeletal disorders. We describe the key discovery that structure-guided mutation of receptor-binding epitopes, especially swapping of these epitopes between ligands, results in new ligands with unique functional properties that can be harnessed clinically. Given the promising results with prototypical engineered TGF-β superfamily ligands, and the vast number of such molecules that remain to be produced and tested, this strategy is likely to hold great promise for the development of new biologics.
- Published
- 2014
46. Regulation of FSHβ induction in LβT2 cells by BMP2 and an Activin A/BMP2 chimera, AB215
- Author
-
Jae Woo Jung, Sun Young Shim, Witek Kwiatkowski, Chihoon Ahn, Senyon Choe, and Peter C. Gray
- Subjects
Transcriptional Activation ,endocrine system ,medicine.medical_specialty ,animal structures ,Endocrinology, Diabetes and Metabolism ,Recombinant Fusion Proteins ,Blotting, Western ,Bone Morphogenetic Protein 2 ,Gonadotrophs ,Smad2 Protein ,ACVR1 ,Bone morphogenetic protein ,Bone morphogenetic protein 2 ,Cell Line ,Mice ,Endocrinology ,Internal medicine ,TGF beta signaling pathway ,medicine ,Animals ,Humans ,Smad3 Protein ,Phosphorylation ,Receptor ,Promoter Regions, Genetic ,Activin type 2 receptors ,Chemistry ,Reverse Transcriptase Polymerase Chain Reaction ,fungi ,Activins ,HEK293 Cells ,embryonic structures ,Follicle Stimulating Hormone, beta Subunit ,Signal transduction ,hormones, hormone substitutes, and hormone antagonists ,ACVR2B ,Signal Transduction - Abstract
Activins and bone morphogenetic proteins (BMPs) share activin type 2 signaling receptors but utilize different type 1 receptors and Smads. We designed AB215, a potent BMP2-like Activin A/BMP2 chimera incorporating the high-affinity type 2 receptor-binding epitope of Activin A. In this study, we compare the signaling properties of AB215 and BMP2 in HEK293T cells and gonadotroph LβT2 cells in which Activin A and BMP2 synergistically induce FSHβ. In HEK293T cells, AB215 is more potent than BMP2 and competitively blocks Activin A signaling, while BMP2 has a partial blocking activity. Activin A signaling is insensitive to BMP pathway antagonism in HEK293T cells but is strongly inhibited by constitutively active (CA) BMP type 1 receptors. By contrast, the potencies of AB215 and BMP2 are indistinguishable in LβT2 cells and although AB215 blocks Activin A signaling, BMP2 has no inhibitory effect. Unlike HEK293T, Activin A signaling is strongly inhibited by BMP pathway antagonism in LβT2 cells but is largely unaffected by CA BMP type 1 receptors. BMP2 increases phospho-Smad3 levels in LβT2 cells, in both the absence and the presence of Activin A treatment, and augments Activin A-induced FSHβ. AB215 has the opposite effect and sharply decreases basal phospho-Smad3 levels and blocks Smad2 phosphorylation and FSHβ induction resulting from Activin A treatment. These findings together demonstrate that while AB215 activates the BMP pathway, it has opposing effects to those of BMP2 on FSHβ induction in LβT2 cells apparently due to its ability to block Activin A signaling.
- Published
- 2014
47. Differential requirement of GRP94 and GRP78 in mammary gland development
- Author
-
Jieli Shen, Si-Yi Chen, Benjamin T. Spike, Miao Wang, Sung-Hyung Lee, Amy S. Lee, Peter C. Gray, and Genyuan Zhu
- Subjects
Pathology ,medicine.medical_specialty ,Glucose-regulated protein ,Morphogenesis ,Article ,Green fluorescent protein ,Mice ,Transduction (genetics) ,Mammary Glands, Animal ,Conditional gene knockout ,medicine ,Animals ,Progenitor cell ,Endoplasmic Reticulum Chaperone BiP ,Heat-Shock Proteins ,Mice, Knockout ,Regulation of gene expression ,Membrane Glycoproteins ,Multidisciplinary ,biology ,Gene Expression Regulation, Developmental ,Phenotype ,Cell biology ,biology.protein ,Molecular Chaperones - Abstract
Glucose Regulated Protein (GRP) 94 and GRP78 are critical molecular chaperones and regulators of signaling. Conditional knockout mouse models have revealed tissue specific requirements for GRP94 and GRP78, including selection for allele retention in specific cell types. Here we report the consequences of mammary-targeted knockout of these GRPs. Our studies revealed that MMTV-Cre, Grp94(f/f) mammary glands, despite GRP94 deficiency, exhibited normal proliferation and ductal morphogenesis. Interestingly, MMTV-Cre, Grp78(f/f) mammary glands displayed only slightly reduced GRP78 protein levels, associating with the retention of the non-recombined Grp78 floxed alleles in isolated mammary epithelial cells and displayed phenotypes comparable to wild-type glands. In contrast, transduction of isolated Grp78(f/f) mammary epithelial stem/progenitor cells with adenovirus expressing GFP and Cre-recombinase was successful in GRP78 ablation, and the GFP sorted cells failed to give rise to repopulated mammary glands in de-epithelialized recipient mice. These studies imply GRP78, but not GRP94, is required for mammary gland development.
- Published
- 2014
- Full Text
- View/download PDF
48. Noninfective mitral valve vegetations identified by transesophageal echocardiography as a cause of stroke
- Author
-
Frank D. Tice, Andrew P. Slivka, Elizabeth T. Walz, Anthony C. Pearson, Peter C. Gray, and David A. Orsinelli
- Subjects
medicine.medical_specialty ,medicine.diagnostic_test ,Stroke patient ,business.industry ,Rehabilitation ,medicine.disease ,medicine.anatomical_structure ,Mitral valve vegetations ,Embolism ,Internal medicine ,Mitral valve ,cardiovascular system ,medicine ,Cardiology ,Infectious etiology ,Surgery ,In patient ,cardiovascular diseases ,Neurology (clinical) ,Radiology ,Cardiology and Cardiovascular Medicine ,business ,Stroke ,Cerebral angiography - Abstract
Backround : Transesophageal echocardiography (TEE) is a useful procedure to evaluate selected stroke patients for cardiac sources of embolism. To date, noninfective valvular vegetations have not been described in large studies using transesophageal echocardiography to detect cardiac sources of embolism. We sought to investigate the frequency of noninfective valvular vegetations in patients with unexplained stroke referred for TEE and to determine the relationship of these vegetations to unrecognized thrombophilic disorders. Methods : We evaluated 641 consecutive patients referred for TEE as a result of unexplained stroke or transient ischemic attack for the presence of valvular vegetations. Of those with vegetations identified, serial blood cultures were obtained to evaluate for an infectious etiology. Patients also had serum testing for thrombophilic disorders and selected patients underwent cerebral angiography. Results : Thirteen patients (2%) who underwent TEE evaluation for unexplained stroke or transient ischemic attack were found to have noninfective valvular vegetations, all involving the mitral valve; none were identified by transthoracic echocardiography. Antiphospholipid antibodies were identified in 8 of these 13 patients (62%) and a protein C deficiency in 1 patient (8%). Conclusions : Noninfective valvular vegetations are a potential cardiac source of embolism in patients with unexplained stroke that can be better identified using transesophageal echocardiography. A large percentage of these individuals have a previously unrecognized thrombophilic disorder.
- Published
- 1998
49. Regulation of ion channels by cAMP-dependent protein kinase and A-kinase anchoring proteins
- Author
-
John D. Scott, Peter C. Gray, and William A. Catterall
- Subjects
A-kinase-anchoring protein ,endocrine system ,Calcium Channels, L-Type ,Voltage-dependent calcium channel ,Kinase ,Chemistry ,General Neuroscience ,A Kinase Anchor Proteins ,Membrane Proteins ,Cyclic AMP-Dependent Protein Kinases ,Cell biology ,Regulator of G protein signaling ,Receptors, Glutamate ,Biochemistry ,Cyclic AMP ,Phosphorylation ,Calcium Channels ,Carrier Proteins ,Protein kinase A ,Ion Channel Gating ,cGMP-dependent protein kinase ,Neuroscience ,Ion channel ,Adaptor Proteins, Signal Transducing ,Signal Transduction - Abstract
Subcellular targeting of the cAMP-dependent protein kinase is achieved, in part, through association with A-kinase anchoring proteins (AKAPs). Recent evidence suggests that specific AKAPs direct the kinase to submembrane sites to facilitate phosphorylation and modulation of a variety of ion channels. A new membrane-anchored AKAP targets cAMP-dependent protein kinase to calcium channels and enhances their regulation in multiple cell types.
- Published
- 1998
50. Primary Structure and Function of an A Kinase Anchoring Protein Associated with Calcium Channels
- Author
-
Peter C. Gray, Lara G. Hays, William A. Catterall, Todd Scheuer, Barry D. Johnson, John R. Yates, Ruth E. Westenbroek, and Brian J. Murphy
- Subjects
Calcium Channels, L-Type ,Neuroscience(all) ,Molecular Sequence Data ,A Kinase Anchor Proteins ,Muscle Proteins ,N-type calcium channel ,Biology ,Kidney ,Gene Expression Regulation, Enzymologic ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Microsomes ,Consensus Sequence ,Animals ,Humans ,ASK1 ,Amino Acid Sequence ,RNA, Messenger ,Muscle, Skeletal ,Protein kinase A ,Adaptor Proteins, Signal Transducing ,030304 developmental biology ,0303 health sciences ,Voltage-dependent calcium channel ,General Neuroscience ,Calcium channel ,Membrane Proteins ,Acetylation ,Blotting, Northern ,Cyclic AMP-Dependent Protein Kinases ,Precipitin Tests ,Rats ,Cell biology ,Calcium ATPase ,R-type calcium channel ,Biochemistry ,Mutagenesis ,Calcium Channels ,Rabbits ,Carrier Proteins ,Ion Channel Gating ,030217 neurology & neurosurgery ,Muscle Contraction ,Protein Binding - Abstract
Rapid, voltage-dependent potentiation of skeletal muscle L-type calcium channels requires phosphorylation by cAMP-dependent protein kinase (PKA) anchored via an A kinase anchoring protein (AKAP). Here we report the isolation, primary sequence determination, and functional characterization of AKAP15, a lipid-anchored protein of 81 amino acid residues with a single amphipathic helix that binds PKA. AKAP15 colocalizes with L-type calcium channels in transverse tubules and is associated with L-type calcium channels in transfected cells. A peptide fragment of AKAP15 encompassing the RII-binding domain blocks voltage-dependent potentiation. These results indicate that AKAP15 targets PKA to the calcium channel and plays a critical role in voltage-dependent potentiation and regulation of skeletal muscle contraction. The expression of AKAP15 in the brain and heart suggests that it may mediate rapid PKA regulation of L-type calcium channels in neurons and cardiac myocytes.
- Published
- 1998
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.