46 results on '"Iglesias-Bartolome R"'
Search Results
2. 733 G-protein-coupled receptors (GPCRs) in the regulation of keratinocyte proliferation and differentiation
- Author
-
Pedro, P., primary, Lund, K., additional, and Iglesias-Bartolome, R., additional
- Published
- 2022
- Full Text
- View/download PDF
3. 349 Regulation of epithelial stem cell proliferation and differentiation by the Gαi heterotrimeric G protein
- Author
-
Pedro, P., primary, Salinas Parra, N., additional, and Iglesias-Bartolome, R., additional
- Published
- 2019
- Full Text
- View/download PDF
4. 1395 Unique transcriptional signature primes oral mucosa for rapid wound healing in humans
- Author
-
Iglesias-Bartolome, R., primary, Uchiyama, A., additional, Graf, R., additional, Molinolo, A.A., additional, Abusleme, L., additional, Brooks, S.R., additional, Callejas-Valera, J., additional, Edwards, D., additional, Doci, C., additional, Asselin-Labat, M., additional, Onaitis, M.W., additional, Moutsopoulos, N., additional, Gutkind, J., additional, and Morasso, M.I., additional
- Published
- 2018
- Full Text
- View/download PDF
5. 144 Regulation of keratinocyte differentiation and proliferation by heterotrimeric G proteins
- Author
-
Pedro, P., primary, Salinas Parra, N., additional, and Iglesias-Bartolome, R., additional
- Published
- 2018
- Full Text
- View/download PDF
6. 893 Transcriptomic changes during oral and cutaneous wound healing reveal differential regulation of wound-activated networks
- Author
-
Iglesias-Bartolome, R., primary, Uchiyama, A., additional, Edwards, D., additional, Molinolo, A., additional, Abusleme, L., additional, Brooks, S., additional, Doci, C., additional, Moutsopoulos, N., additional, Gutkind, S., additional, and Morasso, M.I., additional
- Published
- 2017
- Full Text
- View/download PDF
7. 407 Epigenetic control of epithelial stem cell differentiation by Galpha-s and protein kinase A signaling
- Author
-
Park, J., primary and Iglesias-Bartolome, R., additional
- Published
- 2017
- Full Text
- View/download PDF
8. 711 The molecular anatomy of human oral and cutaneous wound healing
- Author
-
Iglesias-Bartolome, R., primary, Edwards, D., additional, Molinolo, A.A., additional, Brooks, S., additional, Doci, C., additional, Abusleme, L., additional, Moutsopoulos, N., additional, Gutkind, J.S., additional, and Morasso, M.I., additional
- Published
- 2016
- Full Text
- View/download PDF
9. Targeting YAP/TAZ-TEAD signaling as a therapeutic approach in head and neck squamous cell carcinoma.
- Author
-
Sato K, Faraji F, Cervantes-Villagrana RD, Wu X, Koshizuka K, Ishikawa T, Iglesias-Bartolome R, Chen L, Miliani de Marval PL, Gwaltney SL, Adler B, and Gutkind JS
- Abstract
Genetic alterations in Hippo pathway and the consequent activation of YAP/TAZ-TEAD are frequently observed in HPV-negative head and neck squamous cell carcinoma (HNSCC) patients. These include loss-of-function mutation and/or copy number loss of FAT1, and amplification of YAP1 and WWTR1 (encoding TAZ), thus raising the possibility that HNSCC cells may be dependent on YAP/TAZ-TEAD-mediated transcriptional programs. In this regard, the recent development of small molecule TEAD inhibitors (smTEADi) provides an opportunity to therapeutically target Hippo pathway dysregulation in human malignancies. This prompted us to explore the potential benefit of pharmacologically targeting the YAP/TAZ-TEAD axis in this disease. Here, we provide the pre-clinical evidence for the antitumor activity of novel smTEADi, SW-682 in HPV-negative HNSCC. By the use of multiple complementary experimental approaches, including siRNA knockdown, expression of a genetically encoded TEAD inhibitor peptide (pTEADi), and SW-682, we revealed that disruption of YAP/TAZ-TEAD interaction suppresses YAP/TAZ-TEAD-dependent target gene transcription and growth of HNSCC tumors. HNSCC cells with genetic alterations in FAT1 were more sensitive to TEADi compared to FAT1-wild type cells. Mechanistically, TEADi suppressed cell cycle progression and promoted the expression of terminal differentiation gene programs, resulting in tumor growth inhibition. A HNSCC-specific TEADi target gene set was defined from RNA-seq data, which is highly expressed in HNSCC tissues and predicts poor prognosis of HPV-negative HNSCC patients. Our results underscore that YAP/TAZ-TEAD-mediated growth-promoting programs represent a vulnerability in HPV-negative HNSCC, thus providing a pre-clinical rationale for the future evaluation of YAP/TAZ-TEAD targeting strategies as a therapeutic approach for HPV-negative HNSCC patients., Competing Interests: Declaration of competing interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: J. Silvio Gutkind reports consulting fees from Radionetics Oncology, BTB Therapeutics, Pangea Therapeutics, and io9 and is the founder of Kadima Pharmaceuticals, all unrelated to the current study. Lei Chen, Paula L. Miliani de Marval, Stephen L. Gwaltney and Benjamin Adler are employees of SpringWorks Therapeutics., (Copyright © 2025. Published by Elsevier B.V.)
- Published
- 2025
- Full Text
- View/download PDF
10. Control of Epithelial Tissue Organization by mRNA Localization.
- Author
-
Mason DE, Madsen TD, Gasparski AN, Jiwnani N, Lechler T, Weigert R, Iglesias-Bartolome R, and Mili S
- Abstract
mRNA localization to specific subcellular regions is common in mammalian cells but poorly understood in terms of its physiological roles
1-6,7 . This study demonstrates the functional importance of Net1 mRNA, which we find prominently localized at the dermal-epidermal junction (DEJ) in stratified squamous epithelia. Net1 mRNA accumulates at DEJ protrusion-like structures that interact with the basement membrane and connect to a mechanosensitive network of microfibrils. Disrupting Net1 mRNA localization in mouse epithelium alters DEJ morphology and keratinocyte-matrix connections, affecting tissue homeostasis. mRNA localization dictates Net1 protein distribution and its function as a RhoA GTPase exchange factor (GEF). Altered RhoA activity is in turn sufficient to alter the ultrastructure of the DEJ. This study provides a high-resolution in vivo view of mRNA targeting in a physiological context. It further demonstrates how the subcellular localization of a single mRNA can significantly influence mammalian epithelial tissue organization, thus revealing an unappreciated level of post-transcriptional regulation that controls tissue physiology., Competing Interests: Declaration of Interests The authors declare no competing interests.- Published
- 2024
- Full Text
- View/download PDF
11. An improved TEAD dominant-negative protein inhibitor to study Hippo YAP1/TAZ-dependent transcription.
- Author
-
Branch B, Yuan Y, Cascone M, Raimondi F, and Iglesias-Bartolome R
- Abstract
Hippo signaling is one of the top pathways altered in human cancer, and intensive focus has been devoted to developing therapies targeting Hippo-dependent transcription mediated by YAP1 and TAZ interaction with TEAD proteins. However, a significant challenge in evaluating the efficacy of these approaches is the lack of models that can precisely characterize the consequences of TEAD inhibition. To address this gap, our laboratory developed a strategy that utilizes a fluorescently traceable, dominant-negative protein named TEADi. TEADi specifically blocks the nuclear interactions of TEAD with YAP1 and TAZ, enabling precise dissection of Hippo TEAD-dependent and independent effects on cell fate. In this study, we aimed to enhance TEADi effectiveness by altering post-transcriptional modification sites within its TEAD-binding domains (TBDs). We demonstrate that a D93E mutation in the YAP1 TBD significantly increases TEADi inhibitory capacity. Additionally, we find that TBDs derived from VGLL4 and YAP1 are insufficient to block TAZ-induced TEAD activity, revealing crucial differences in YAP1 and TAZ displacement mechanisms by dominant-negative TBDs. Structural differences in YAP1 and TAZ TBDs were also identified, which may contribute to the distinct binding of these proteins to TEAD. Our findings expand our understanding of TEAD regulation and highlight the potential of an optimized TEADi as a more potent, specific, and versatile tool for studying TEAD-transcriptional activity., Competing Interests: CONFLICT OF INTEREST Ramiro Iglesias-Bartolome holds an NCI Employee Invention Report (EIR) for commercial licensing of Peptide inhibitors of YAP1/TAZ-TEAD, E-108–2019-0. The other authors do not have any conflict of interest to declare.
- Published
- 2024
- Full Text
- View/download PDF
12. GPCR Screening Reveals that the Metabolite Receptor HCAR3 Regulates Epithelial Proliferation, Migration, and Cellular Respiration.
- Author
-
Pedro MP, Lund K, Kang SWS, Chen T, Stuelten CH, Porat-Shliom N, and Iglesias-Bartolome R
- Subjects
- Humans, Signal Transduction, Cell Differentiation, Cells, Cultured, Receptors, Leukotriene B4 metabolism, Receptors, Leukotriene B4 genetics, Epithelial Cells metabolism, Receptors, Nicotinic, Cell Movement, Keratinocytes metabolism, Keratinocytes cytology, Receptors, G-Protein-Coupled metabolism, Receptors, G-Protein-Coupled genetics, Cell Proliferation, Cell Respiration physiology
- Abstract
Epithelial cells in the skin and other tissues rely on signals from their environment to maintain homeostasis and respond to injury, and GPCRs play a critical role in this communication. A better understanding of the GPCRs expressed in epithelial cells will contribute to understanding the relationship between cells and their niche and could lead to developing new therapies to modulate cell fate. This study used human primary keratinocytes as a model to investigate the specific GPCRs regulating epithelial cell proliferation and differentiation. We identified 3 key receptors-HCAR3, LTB4R, and GPR137-and found that knockdown of these receptors led to changes in numerous gene networks that are important for maintaining cell identity and promoting proliferation while inhibiting differentiation. Our study also revealed that the metabolite receptor HCAR3 regulates keratinocyte migration and cellular metabolism. Knockdown of HCAR3 led to reduced keratinocyte migration and respiration, which could be attributed to altered metabolite use and aberrant mitochondrial morphology caused by the absence of the receptor. This study contributes to understanding the complex interplay between GPCR signaling and epithelial cell fate decisions., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
13. Epithelial-derived interleukin-23 promotes oral mucosal immunopathology.
- Author
-
Kim TS, Ikeuchi T, Theofilou VI, Williams DW, Greenwell-Wild T, June A, Adade EE, Li L, Abusleme L, Dutzan N, Yuan Y, Brenchley L, Bouladoux N, Sakamachi Y, Palmer RJ Jr, Iglesias-Bartolome R, Trinchieri G, Garantziotis S, Belkaid Y, Valm AM, Diaz PI, Holland SM, and Moutsopoulos NM
- Subjects
- Humans, Epithelial Cells, Inflammation, Toll-Like Receptor 5 metabolism, Interleukin-23, Periodontitis
- Abstract
At mucosal surfaces, epithelial cells provide a structural barrier and an immune defense system. However, dysregulated epithelial responses can contribute to disease states. Here, we demonstrated that epithelial cell-intrinsic production of interleukin-23 (IL-23) triggers an inflammatory loop in the prevalent oral disease periodontitis. Epithelial IL-23 expression localized to areas proximal to the disease-associated microbiome and was evident in experimental models and patients with common and genetic forms of disease. Mechanistically, flagellated microbial species of the periodontitis microbiome triggered epithelial IL-23 induction in a TLR5 receptor-dependent manner. Therefore, unlike other Th17-driven diseases, non-hematopoietic-cell-derived IL-23 served as an initiator of pathogenic inflammation in periodontitis. Beyond periodontitis, analysis of publicly available datasets revealed the expression of epithelial IL-23 in settings of infection, malignancy, and autoimmunity, suggesting a broader role for epithelial-intrinsic IL-23 in human disease. Collectively, this work highlights an important role for the barrier epithelium in the induction of IL-23-mediated inflammation., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
14. The GPCR-Gα s -PKA signaling axis promotes T cell dysfunction and cancer immunotherapy failure.
- Author
-
Wu VH, Yung BS, Faraji F, Saddawi-Konefka R, Wang Z, Wenzel AT, Song MJ, Pagadala MS, Clubb LM, Chiou J, Sinha S, Matic M, Raimondi F, Hoang TS, Berdeaux R, Vignali DAA, Iglesias-Bartolome R, Carter H, Ruppin E, Mesirov JP, and Gutkind JS
- Subjects
- Mice, Animals, Signal Transduction, Mice, Transgenic, Immunotherapy, Tumor Microenvironment, CD8-Positive T-Lymphocytes, Neoplasms
- Abstract
Immune checkpoint blockade (ICB) targeting PD-1 and CTLA-4 has revolutionized cancer treatment. However, many cancers do not respond to ICB, prompting the search for additional strategies to achieve durable responses. G-protein-coupled receptors (GPCRs) are the most intensively studied drug targets but are underexplored in immuno-oncology. Here, we cross-integrated large singe-cell RNA-sequencing datasets from CD8
+ T cells covering 19 distinct cancer types and identified an enrichment of Gαs -coupled GPCRs on exhausted CD8+ T cells. These include EP2 , EP4 , A2A R, β1 AR and β2 AR, all of which promote T cell dysfunction. We also developed transgenic mice expressing a chemogenetic CD8-restricted Gαs -DREADD to activate CD8-restricted Gαs signaling and show that a Gαs -PKA signaling axis promotes CD8+ T cell dysfunction and immunotherapy failure. These data indicate that Gαs -GPCRs are druggable immune checkpoints that might be targeted to enhance the response to ICB immunotherapies., (© 2023. The Author(s), under exclusive licence to Springer Nature America, Inc.)- Published
- 2023
- Full Text
- View/download PDF
15. Regulation of Photosensitivity by the Hippo Pathway in Lupus Skin.
- Author
-
Hile GA, Coit P, Xu B, Victory AM, Gharaee-Kermani M, Estadt SN, Maz MP, Martens JWS, Wasikowski R, Dobry C, Tsoi LC, Iglesias-Bartolome R, Berthier CC, Billi AC, Gudjonsson JE, Sawalha AH, and Kahlenberg JM
- Subjects
- Humans, Keratinocytes metabolism, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, RNA, Small Interfering, Intracellular Signaling Peptides and Proteins genetics, Intracellular Signaling Peptides and Proteins metabolism, Hippo Signaling Pathway, Lupus Erythematosus, Systemic genetics
- Abstract
Objective: Photosensitivity is one of the most common manifestations of systemic lupus erythematosus (SLE), yet its pathogenesis is not well understood. The normal-appearing epidermis of patients with SLE exhibits increased ultraviolet B (UVB)-driven cell death that persists in cell culture. Here, we investigated the role of epigenetic modification and Hippo signaling in enhanced UVB-induced apoptosis seen in SLE keratinocytes., Methods: We analyzed DNA methylation in cultured keratinocytes from SLE patients compared to keratinocytes from healthy controls (n = 6/group). Protein expression was validated in cultured keratinocytes using immunoblotting and immunofluorescence. An immortalized keratinocyte line overexpressing WWC1 was generated via lentiviral vector. WWC1-driven changes were inhibited using a large tumor suppressor kinase 1/2 (LATS1/2) inhibitor (TRULI) and small interfering RNA (siRNA). The interaction between the Yes-associated protein (YAP) and the transcriptional enhancer associate domain (TEAD) was inhibited by overexpression of an N/TERT cell line expressing a tetracycline-inducible green fluorescent protein-tagged protein that inhibits YAP-TEAD binding (TEADi). Apoptosis was assessed using cleaved caspase 3/7 and TUNEL staining., Results: Hippo signaling was the top differentially methylated pathway in SLE versus control keratinocytes. SLE keratinocytes (n = 6) showed significant hypomethylation (Δβ = -0.153) and thus overexpression of the Hippo regulator WWC1 (P = 0.002). WWC1 overexpression increased LATS1/2 kinase activation, leading to YAP cytoplasmic retention and altered proapoptotic transcription in SLE keratinocytes. Accordingly, UVB-mediated apoptosis in keratinocytes could be enhanced by WWC1 overexpression or YAP-TEAD inhibition, mimicking SLE keratinocytes. Importantly, inhibition of LATS1/2 with either the chemical inhibitor TRULI or siRNA effectively eliminated enhanced UVB-apoptosis in SLE keratinocytes., Conclusion: Our work unravels a novel driver of photosensitivity in SLE: overactive Hippo signaling in SLE keratinocytes restricts YAP transcriptional activity, leading to shifts that promote UVB apoptosis., (© 2023 American College of Rheumatology.)
- Published
- 2023
- Full Text
- View/download PDF
16. A GPCR screening in human keratinocytes identifies that the metabolite receptor HCAR3 controls epithelial proliferation, migration, and cellular respiration.
- Author
-
Pedro MP, Lund K, Kang SWS, Chen T, Stuelten CH, Porat-Shliom N, and Iglesias-Bartolome R
- Abstract
Epithelial cells in the skin and other tissues rely on signals from their environment to maintain homeostasis and respond to injury, and G protein-coupled receptors (GPCRs) play a critical role in this communication. A better understanding of the GPCRs expressed in epithelial cells will contribute to understanding the relationship between cells and their niche and could lead to developing new therapies to modulate cell fate. This study used human primary keratinocytes as a model to investigate the specific GPCRs regulating epithelial cell proliferation and differentiation. We identified three key receptors, hydroxycarboxylic acid-receptor 3 (HCAR3), leukotriene B4-receptor 1 (LTB4R), and G Protein-Coupled Receptor 137 (GPR137) and found that knockdown of these receptors led to changes in numerous gene networks that are important for maintaining cell identity and promoting proliferation while inhibiting differentiation. Our study also revealed that the metabolite receptor HCAR3 regulates keratinocyte migration and cellular metabolism. Knockdown of HCAR3 led to reduced keratinocyte migration and respiration, which could be attributed to altered metabolite use and aberrant mitochondrial morphology caused by the absence of the receptor. This study contributes to understanding the complex interplay between GPCR signaling and epithelial cell fate decisions., Competing Interests: Competing Interest Statement: No competing interests to declare.
- Published
- 2023
- Full Text
- View/download PDF
17. Oncogenic Hedgehog-Smoothened Signaling Depends on YAP1‒TAZ/TEAD Transcription to Restrain Differentiation in Basal Cell Carcinoma.
- Author
-
Yuan Y, Salinas Parra N, Chen Q, and Iglesias-Bartolome R
- Subjects
- Animals, Carcinogenesis, Carcinoma, Basal Cell genetics, Cell Differentiation, Cell Line, Tumor, Disease Models, Animal, Gene Expression Regulation, Neoplastic, Gene Knockdown Techniques, Hippo Signaling Pathway, Humans, Kruppel-Like Factor 4 metabolism, Mice, NF-kappa B metabolism, STAT3 Transcription Factor metabolism, Snail Family Transcription Factors metabolism, Transcriptional Coactivator with PDZ-Binding Motif Proteins genetics, YAP-Signaling Proteins genetics, Carcinoma, Basal Cell metabolism, Hedgehogs metabolism, TEA Domain Transcription Factors metabolism, Transcriptional Coactivator with PDZ-Binding Motif Proteins metabolism, YAP-Signaling Proteins metabolism
- Abstract
Disruption of the transcriptional activity of the Hippo pathway members YAP1 and TAZ has become a major target for cancer treatment. However, detailed analysis of the effectiveness and networks affected by YAP1/TAZ transcriptional targeting is limited. In this study, we utilize TEAD inhibitor, an inhibitor of the binding of YAP1 and TAZ with their main transcriptional target TEAD in a mouse model of basal cell carcinoma, to unveil the consequences of YAP1/TAZ transcriptional blockage in cancer cells. Both TEAD inhibitor and YAP1/TAZ knockdown lead to reduced proliferation and increased differentiation of mouse basal cell carcinoma driven by oncogenic hedgehog-smoothened (SmoM2) activity. Although TEAD-transcriptional networks were essential to inactivate differentiation, this inactivation was found to be indirect and potentially mediated through the repression of KLF4 by SNAI2. By comparing the transcriptional effects of TEAD inhibition with those caused by YAP1/TAZ depletion, we determined YAP1/TAZ‒TEAD‒independent effects in cancer cells that impact STAT3 and NF-κB. Our results reveal the gene networks affected by targeting YAP1/TAZ‒TEAD in basal cell carcinoma tumors and expose the potential pitfalls for targeting TEAD transcription in cancer., (Published by Elsevier Inc.)
- Published
- 2022
- Full Text
- View/download PDF
18. Kallikrein 5 Inhibition by the Lympho-Epithelial Kazal-Type Related Inhibitor Hinders Matriptase-Dependent Carcinogenesis.
- Author
-
da Silva EZM, Fraga-Silva TFC, Yuan Y, Alves MG, Publio GA, da Fonseca CK, Kodama MH, Vieira GV, Candido MF, Innocentini LMAR, Miranda MG, da Silva AR, Alves-Filho JC, Bonato VLD, Iglesias-Bartolome R, and Sales KU
- Abstract
Head and neck squamous cell carcinoma remains challenging to treat with no improvement in survival rates over the past 50 years. Thus, there is an urgent need to discover more reliable therapeutic targets and biomarkers for HNSCC. Matriptase, a type-II transmembrane serine protease, induces malignant transformation in epithelial stem cells through proteolytic activation of pro-HGF and PAR-2, triggering PI3K-AKT-mTOR and NFKB signaling. The serine protease inhibitor lympho-epithelial Kazal-type-related inhibitor (LEKTI) inhibits the matriptase-driven proteolytic pathway, directly blocking kallikreins in epithelial differentiation. Hence, we hypothesized LEKTI could inhibit matriptase-dependent squamous cell carcinogenesis, thus implicating kallikreins in this process. Double-transgenic mice with simultaneous expression of matriptase and LEKTI under the keratin-5 promoter showed a prominent rescue of K5-Matriptase
+/0 premalignant phenotype. Notably, in DMBA-induced SCC, heterotopic co-expression of LEKTI and matriptase delayed matriptase-driven tumor incidence and progression. Co-expression of LEKTI reverted altered Kallikrein-5 expression observed in the skin of K5-Matriptase+/0 mice, indicating that matriptase-dependent proteolytic pathway inhibition by LEKTI occurs through kallikreins. Moreover, we showed that Kallikrein-5 is necessary for PAR-2-mediated IL-8 release, YAP1-TAZ/TEAD activation, and matriptase-mediated oral squamous cell carcinoma migration. Collectively, our data identify a third signaling pathway for matriptase-dependent carcinogenesis in vivo. These findings are critical for the identification of more reliable biomarkers and effective therapeutic targets in Head and Neck cancer.- Published
- 2021
- Full Text
- View/download PDF
19. A biomechanical switch regulates the transition towards homeostasis in oesophageal epithelium.
- Author
-
McGinn J, Hallou A, Han S, Krizic K, Ulyanchenko S, Iglesias-Bartolome R, England FJ, Verstreken C, Chalut KJ, Jensen KB, Simons BD, and Alcolea MP
- Subjects
- Animals, Epithelium metabolism, Esophageal Mucosa metabolism, Humans, Kruppel-Like Factor 4, Mice, Stem Cells metabolism, Cell Differentiation physiology, Cell Proliferation physiology, Epithelial Cells metabolism, Homeostasis physiology
- Abstract
Epithelial cells rapidly adapt their behaviour in response to increasing tissue demands. However, the processes that finely control these cell decisions remain largely unknown. The postnatal period covering the transition between early tissue expansion and the establishment of adult homeostasis provides a convenient model with which to explore this question. Here, we demonstrate that the onset of homeostasis in the epithelium of the mouse oesophagus is guided by the progressive build-up of mechanical strain at the organ level. Single-cell RNA sequencing and whole-organ stretching experiments revealed that the mechanical stress experienced by the growing oesophagus triggers the emergence of a bright Krüppel-like factor 4 (KLF4) committed basal population, which balances cell proliferation and marks the transition towards homeostasis in a yes-associated protein (YAP)-dependent manner. Our results point to a simple mechanism whereby mechanical changes experienced at the whole-tissue level are integrated with those sensed at the cellular level to control epithelial cell fate.
- Published
- 2021
- Full Text
- View/download PDF
20. Insights into epithelial cell senescence from transcriptome and secretome analysis of human oral keratinocytes.
- Author
-
Schwartz RE, Shokhirev MN, Andrade LR, Gutkind JS, Iglesias-Bartolome R, and Shadel GS
- Subjects
- Extracellular Vesicles, Humans, Sequence Analysis, RNA, Signal Transduction, Cellular Senescence physiology, Epithelial Cells physiology, Gene Expression Profiling, Keratinocytes metabolism, Mouth Mucosa
- Abstract
Senescent cells produce chronic inflammation that contributes to the diseases and debilities of aging. How this process is orchestrated in epithelial cells, the origin of human carcinomas, is poorly understood. We used human normal oral keratinocytes (NOKs) to elucidate senescence programs in a prototype primary mucosal epithelial cell that senesces spontaneously. While NOKs exhibit several typical facets of senescence, they also display distinct characteristics. These include expression of p21WAF1/CIP1 at early passages, making this common marker of senescence unreliable in NOKs. Transcriptome analysis by RNA-seq revealed specific commonalities with and differences from cancer cells, explicating the tumor avoidance role of senescence. Repression of DNA repair genes that correlated with downregulation of E2F1 mRNA and protein was observed for two donors; a divergent result was seen for the third. Using proteomic profiling of soluble (non-vesicular) and extracellular vesicle (EV) associated secretions, we propose additions to the senescence associated secretory phenotype, including HSP60, which localizes to the surface of EVs. Finally, EVs from senescent NOKs activate interferon pathway signaling in THP-1 monocytes in a STING-dependent manner and associate with mitochondrial and nuclear DNA. Our results highlight senescence changes in epithelial cells and how they might contribute to chronic inflammation and age-related diseases.
- Published
- 2021
- Full Text
- View/download PDF
21. Protein kinase A inhibitor proteins (PKIs) divert GPCR-Gαs-cAMP signaling toward EPAC and ERK activation and are involved in tumor growth.
- Author
-
Hoy JJ, Salinas Parra N, Park J, Kuhn S, and Iglesias-Bartolome R
- Subjects
- Acetylcysteine metabolism, Animals, Cell Line, Tumor, Cyclic AMP metabolism, Cyclic AMP-Dependent Protein Kinases metabolism, Erythromycin metabolism, Extracellular Signal-Regulated MAP Kinases metabolism, Feedback, Physiological, Female, GTP-Binding Protein alpha Subunits metabolism, HEK293 Cells, Humans, Intracellular Signaling Peptides and Proteins genetics, Male, Mice, Mice, Nude, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology, Acetylcysteine analogs & derivatives, Erythromycin analogs & derivatives, Intracellular Signaling Peptides and Proteins metabolism, MAP Kinase Signaling System, Prostatic Neoplasms metabolism
- Abstract
The PKA-inhibitor (PKI) family members PKIα, PKIβ, and PKIγ bind with high affinity to PKA and block its kinase activity, modulating the extent, and duration of PKA-mediated signaling events. While PKA is a well-known regulator of physiological and oncogenic events, the role of PKI proteins in these pathways has remained elusive. Here, by measuring activation of the MAPK pathway downstream of GPCR-Gαs-cAMP signaling, we show that the expression levels of PKI proteins can alter the balance of activation of two major cAMP targets: PKA and EPAC. Our results indicate that PKA maintains repressive control over MAPK signaling as well as a negative feedback on cAMP concentration. Overexpression of PKI and its subsequent repression of PKA dysregulates these signaling pathways, resulting in increased intracellular cAMP, and enhanced activation of EPAC and MAPK. We also find that amplifications of PKIA are common in prostate cancer and are associated with reduced progression free survival. Depletion of PKIA in prostate cancer cells leads to reduced migration, increased sensitivity to anoikis and reduced tumor growth. By altering PKA activity PKI can act as a molecular switch, driving GPCR-Gαs-cAMP signaling toward activation of EPAC-RAP1 and MAPK, ultimately modulating tumor growth., (© Published 2020. This article is a U.S. Government work and is in the public domain in the USA.)
- Published
- 2020
- Full Text
- View/download PDF
22. The landscape of GPCR signaling in the regulation of epidermal stem cell fate and skin homeostasis.
- Author
-
Pedro MP, Lund K, and Iglesias-Bartolome R
- Abstract
Continuous integration of signals from the micro and macro-environment is necessary for somatic stem cells to adapt to changing conditions, maintain tissue homeostasis and activate repair mechanisms. G-protein coupled receptors (GPCRs) facilitate this integration by binding to numerous hormones, metabolites and inflammatory mediators, influencing a diverse network of pathways that regulate stem cell fate. This adaptive mechanism is particularly relevant for tissues that are exposed to environmental assault, like skin. The skin is maintained by a set of basal keratinocyte stem and progenitor cells located in the hair follicle and interfollicular epidermis, and several GPCRs and their signaling partners serve as makers and regulators of epidermal stem cell activity. GPCRs utilize heterotrimeric G protein dependent and independent pathways to translate extracellular signals into intracellular molecular cascades that dictate the activation of keratinocyte proliferative and differentiation networks, including Hedgehog GLI, Hippo YAP1 and WNT/β-catenin, ultimately regulating stem cell identity. Dysregulation of GPCR signaling underlines numerous skin inflammatory diseases and cancer, with smoothened-driven basal cell carcinoma being a main example of a GPCR associated cancer. In this review, we discuss the impact of GPCRs and their signaling partners in skin keratinocyte biology, particularly in the regulation of the epidermal stem cell compartment., (Published 2020. This article is a U.S. Government work and is in the public domain in the USA.)
- Published
- 2020
- Full Text
- View/download PDF
23. Unleashing Immunotherapy by Targeting Cancer Stem Cells.
- Author
-
Iglesias-Bartolome R and Gutkind JS
- Subjects
- Humans, Immunotherapy, Neoplasm Recurrence, Local, Neoplastic Stem Cells, Polycomb Repressive Complex 1, Carcinoma, Squamous Cell, Head and Neck Neoplasms therapy
- Abstract
In this issue of Cell Stem Cell, Jia et al. (2020) identify residual cancer stem cells (CSCs) as a mechanism of immunotherapy resistance in head and neck squamous cell carcinoma (HNSCC). Remarkably, targeting this population of CSCs can be exploited to potentiate immunotherapy and reduce tumor recurrence and metastasis., Competing Interests: Declaration of Interests J.S.G. is a Member of the Scientific Advisory Board of Oncoceutics, Vividion Therapeutics, and Domain Therapeutics., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
24. Activation of G-Protein Coupled Receptor-Gαi Signaling Increases Keratinocyte Proliferation and Reduces Differentiation, Leading to Epidermal Hyperplasia.
- Author
-
Pedro MP, Salinas Parra N, Gutkind JS, and Iglesias-Bartolome R
- Subjects
- Animals, Cell Differentiation, Cell Proliferation, Female, Humans, Hyperplasia pathology, Male, Mice, NIH 3T3 Cells, Signal Transduction physiology, Epidermis pathology, GTP-Binding Protein alpha Subunits, Gi-Go metabolism, Keratinocytes pathology
- Abstract
G-protein coupled receptors (GPCRs) and their associated heterotrimeric G proteins impinge on pathways that control epithelial cell self-renewal and differentiation. Although it is known that Gαs protein signaling regulates skin homeostasis in vivo, the role of GPCR-coupled Gαi proteins in the skin is unclear. Here, by using a chemogenetic approach, we demonstrate that GPCR-Gαi activation can regulate keratinocyte proliferation and differentiation and that overactivation of Gαi-signaling in the basal compartment of the mouse skin can lead to epidermal hyperplasia. Our results expand our understanding of the role of GPCR-cAMP signaling in skin homeostasis and reveal overlapping and divergent roles of the cAMP-regulating heterotrimeric Gαs and Gαi proteins in keratinocytes., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
25. YAP1/TAZ-TEAD transcriptional networks maintain skin homeostasis by regulating cell proliferation and limiting KLF4 activity.
- Author
-
Yuan Y, Park J, Feng A, Awasthi P, Wang Z, Chen Q, and Iglesias-Bartolome R
- Subjects
- Animals, Binding Sites, Cell Cycle Checkpoints genetics, Cell Differentiation genetics, Cell Proliferation genetics, HEK293 Cells, Humans, Inflammation pathology, Keratinocytes cytology, Keratinocytes metabolism, Kruppel-Like Factor 4, Mice, Models, Animal, Models, Biological, Protein Binding, Stem Cells cytology, Stem Cells metabolism, Transcription, Genetic, Transcriptional Coactivator with PDZ-Binding Motif Proteins, YAP-Signaling Proteins, Adaptor Proteins, Signal Transducing metabolism, Cell Cycle Proteins metabolism, Gene Regulatory Networks, Homeostasis genetics, Kruppel-Like Transcription Factors metabolism, Skin metabolism, Trans-Activators metabolism, Transcription Factors metabolism
- Abstract
The Hippo TEAD-transcriptional regulators YAP1 and TAZ are central for cell renewal and cancer growth; however, the specific downstream gene networks involved in their activity are not completely understood. Here we introduce TEADi, a genetically encoded inhibitor of the interaction of YAP1 and TAZ with TEAD, as a tool to characterize the transcriptional networks and biological effects regulated by TEAD transcription factors. Blockage of TEAD activity by TEADi in human keratinocytes and mouse skin leads to reduced proliferation and rapid activation of differentiation programs. Analysis of gene networks affected by TEADi and YAP1/TAZ knockdown identifies KLF4 as a central transcriptional node regulated by YAP1/TAZ-TEAD in keratinocyte differentiation. Moreover, we show that TEAD and KLF4 can regulate the activity of each other, indicating that these factors are part of a transcriptional regulatory loop. Our study establishes TEADi as a resource for studying YAP1/TAZ-TEAD dependent effects.
- Published
- 2020
- Full Text
- View/download PDF
26. Genome-wide prediction of synthetic rescue mediators of resistance to targeted and immunotherapy.
- Author
-
Sahu AD, S Lee J, Wang Z, Zhang G, Iglesias-Bartolome R, Tian T, Wei Z, Miao B, Nair NU, Ponomarova O, Friedman AA, Amzallag A, Moll T, Kasumova G, Greninger P, Egan RK, Damon LJ, Frederick DT, Jerby-Arnon L, Wagner A, Cheng K, Park SG, Robinson W, Gardner K, Boland G, Hannenhalli S, Herlyn M, Benes C, Flaherty K, Luo J, Gutkind JS, and Ruppin E
- Subjects
- Female, Gene Expression Profiling, Humans, Immunotherapy, Male, Melanoma drug therapy, Molecular Targeted Therapy, Synthetic Lethal Mutations, Computational Biology, Drug Resistance, Neoplasm genetics, Drug Synergism, Melanoma genetics
- Abstract
Most patients with advanced cancer eventually acquire resistance to targeted therapies, spurring extensive efforts to identify molecular events mediating therapy resistance. Many of these events involve synthetic rescue (SR) interac tions, where the reduction in cancer cell viability caused by targeted gene inactivation is rescued by an adaptive alteration of another gene (the rescuer ). Here, we perform a genome-wide in silico prediction of SR rescuer genes by analyzing tumor transcriptomics and survival data of 10,000 TCGA cancer patients. Predicted SR interactions are validated in new experimental screens. We show that SR interactions can successfully predict cancer patients' response and emerging resistance. Inhibiting predicted rescuer genes sensitizes resistant cancer cells to therapies synergistically, providing initial leads for developing combinatorial approaches to overcome resistance proactively. Finally, we show that the SR analysis of melanoma patients successfully identifies known mediators of resistance to immunotherapy and predicts novel rescuers., (© 2019 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2019
- Full Text
- View/download PDF
27. Transcriptional signature primes human oral mucosa for rapid wound healing.
- Author
-
Iglesias-Bartolome R, Uchiyama A, Molinolo AA, Abusleme L, Brooks SR, Callejas-Valera JL, Edwards D, Doci C, Asselin-Labat ML, Onaitis MW, Moutsopoulos NM, Gutkind JS, and Morasso MI
- Subjects
- Biopsy, Humans, Keratinocytes metabolism, Skin cytology, Skin metabolism, Transcription Factors genetics, Transcription Factors metabolism, Wound Healing genetics, Mouth Mucosa metabolism, Wound Healing physiology
- Abstract
Oral mucosal wound healing has long been regarded as an ideal system of wound resolution. However, the intrinsic characteristics that mediate optimal healing at mucosal surfaces are poorly understood, particularly in humans. We present a unique comparative analysis between human oral and cutaneous wound healing using paired and sequential biopsies during the repair process. Using molecular profiling, we determined that wound-activated transcriptional networks are present at basal state in the oral mucosa, priming the epithelium for wound repair. We show that oral mucosal wound-related networks control epithelial cell differentiation and regulate inflammatory responses, highlighting fundamental global mechanisms of repair and inflammatory responses in humans. The paired comparative analysis allowed for the identification of differentially expressed SOX2 (sex-determining region Y-box 2) and PITX1 (paired-like homeodomain 1) transcriptional regulators in oral versus skin keratinocytes, conferring a unique identity to oral keratinocytes. We show that SOX2 and PITX1 transcriptional function has the potential to reprogram skin keratinocytes to increase cell migration and improve wound resolution in vivo. Our data provide insights into therapeutic targeting of chronic and nonhealing wounds based on greater understanding of the biology of healing in human mucosal and cutaneous environments., (Copyright © 2018 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2018
- Full Text
- View/download PDF
28. Assembly and activation of the Hippo signalome by FAT1 tumor suppressor.
- Author
-
Martin D, Degese MS, Vitale-Cross L, Iglesias-Bartolome R, Valera JLC, Wang Z, Feng X, Yeerna H, Vadmal V, Moroishi T, Thorne RF, Zaida M, Siegele B, Cheong SC, Molinolo AA, Samuels Y, Tamayo P, Guan KL, Lippman SM, Lyons JG, and Gutkind JS
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing metabolism, Cadherins genetics, Cadherins metabolism, Gene Knockdown Techniques, HEK293 Cells, Head and Neck Neoplasms metabolism, Head and Neck Neoplasms pathology, Hepatocyte Growth Factor metabolism, Hippo Signaling Pathway, Humans, Phosphoproteins genetics, Phosphoproteins metabolism, Protein Serine-Threonine Kinases genetics, Proto-Oncogene Proteins metabolism, Signal Transduction, Squamous Cell Carcinoma of Head and Neck metabolism, Squamous Cell Carcinoma of Head and Neck pathology, Transcription Factors, YAP-Signaling Proteins, Adaptor Proteins, Signal Transducing physiology, Cadherins physiology, Head and Neck Neoplasms genetics, Phosphoproteins physiology, Protein Serine-Threonine Kinases metabolism, Squamous Cell Carcinoma of Head and Neck genetics
- Abstract
Dysregulation of the Hippo signaling pathway and the consequent YAP1 activation is a frequent event in human malignancies, yet the underlying molecular mechanisms are still poorly understood. A pancancer analysis of core Hippo kinases and their candidate regulating molecules revealed few alterations in the canonical Hippo pathway, but very frequent genetic alterations in the FAT family of atypical cadherins. By focusing on head and neck squamous cell carcinoma (HNSCC), which displays frequent FAT1 alterations (29.8%), we provide evidence that FAT1 functional loss results in YAP1 activation. Mechanistically, we found that FAT1 assembles a multimeric Hippo signaling complex (signalome), resulting in activation of core Hippo kinases by TAOKs and consequent YAP1 inactivation. We also show that unrestrained YAP1 acts as an oncogenic driver in HNSCC, and that targeting YAP1 may represent an attractive precision therapeutic option for cancers harboring genomic alterations in the FAT1 tumor suppressor genes.
- Published
- 2018
- Full Text
- View/download PDF
29. Expression of an active Gα s mutant in skeletal stem cells is sufficient and necessary for fibrous dysplasia initiation and maintenance.
- Author
-
Zhao X, Deng P, Iglesias-Bartolome R, Amornphimoltham P, Steffen DJ, Jin Y, Molinolo AA, de Castro LF, Ovejero D, Yuan Q, Chen Q, Han X, Bai D, Taylor SS, Yang Y, Collins MT, and Gutkind JS
- Subjects
- Animals, Anti-Bacterial Agents toxicity, Bone Development drug effects, Bone and Bones pathology, Cell Differentiation, Doxycycline toxicity, GTP-Binding Protein alpha Subunits, Gs genetics, Gene Expression Regulation, Gene Expression Regulation, Developmental drug effects, Mice, Mutation, Fibrous Dysplasia of Bone metabolism, GTP-Binding Protein alpha Subunits, Gs metabolism, Mesenchymal Stem Cells metabolism
- Abstract
Fibrous dysplasia (FD) is a disease caused by postzygotic activating mutations of GNAS (R201C and R201H) that encode the α-subunit of the G
s stimulatory protein. FD is characterized by the development of areas of abnormal fibroosseous tissue in the bones, resulting in skeletal deformities, fractures, and pain. Despite the well-defined genetic alterations underlying FD, whether GNAS activation is sufficient for FD initiation and the molecular and cellular consequences of GNAS mutations remains largely unresolved, and there are no currently available targeted therapeutic options for FD. Here, we have developed a conditional tetracycline (Tet)-inducible animal model expressing the Gαs R201C in the skeletal stem cell (SSC) lineage (Tet-Gαs R201C / Prrx1 -Cre/LSL-rtTA-IRES-GFP mice), which develops typical FD bone lesions in both embryos and adult mice in less than 2 weeks following doxycycline (Dox) administration. Conditional Gαs R201C expression promoted PKA activation and proliferation of SSCs along the osteogenic lineage but halted their differentiation to mature osteoblasts. Rather, as is seen clinically, areas of woven bone admixed with fibrous tissue were formed. Gαs R201C caused the concomitant expression of receptor activator of nuclear factor kappa-B ligand (Rankl) that led to marked osteoclastogenesis and bone resorption. Gαs R201C expression ablation by Dox withdrawal resulted in FD-like lesion regression, supporting the rationale for Gαs -targeted drugs to attempt FD cure. This model, which develops FD-like lesions that can form rapidly and revert on cessation of mutant Gαs expression, provides an opportunity to identify the molecular mechanism underlying FD initiation and progression and accelerate the development of new treatment options., Competing Interests: The authors declare no conflict of interest., (Copyright © 2018 the Author(s). Published by PNAS.)- Published
- 2018
- Full Text
- View/download PDF
30. Policing Tumorigenesis within the Skin: Good Outs Bad.
- Author
-
Iglesias-Bartolome R and Morasso MI
- Subjects
- Homeostasis, Humans, Stem Cells cytology, Carcinogenesis, Skin
- Abstract
Recently published in Nature, Brown et al. (2017) shed new light on how the skin handles the activation of oncogenic pathways in the stem cell compartment and how wild-type cells limit the proliferation of mutant cells to maintain proper tissue homeostasis., (Published by Elsevier Inc.)
- Published
- 2017
- Full Text
- View/download PDF
31. mTOR inhibition prevents rapid-onset of carcinogen-induced malignancies in a novel inducible HPV-16 E6/E7 mouse model.
- Author
-
Callejas-Valera JL, Iglesias-Bartolome R, Amornphimoltham P, Palacios-Garcia J, Martin D, Califano JA, Molinolo AA, and Gutkind JS
- Subjects
- 9,10-Dimethyl-1,2-benzanthracene toxicity, Animals, Carcinoma, Squamous Cell chemically induced, Carcinoma, Squamous Cell drug therapy, Carcinoma, Squamous Cell virology, Cell Proliferation drug effects, Gene Expression Regulation, Neoplastic, Human papillomavirus 16 genetics, Human papillomavirus 16 pathogenicity, Humans, Mice, Oncogene Proteins, Viral genetics, Oropharyngeal Neoplasms chemically induced, Oropharyngeal Neoplasms drug therapy, Oropharyngeal Neoplasms virology, Papillomavirus E7 Proteins genetics, Papillomavirus Infections pathology, Papillomavirus Infections virology, Phorbol Esters toxicity, Repressor Proteins genetics, Sirolimus administration & dosage, TOR Serine-Threonine Kinases antagonists & inhibitors, TOR Serine-Threonine Kinases genetics, Carcinogens toxicity, Carcinoma, Squamous Cell genetics, Oropharyngeal Neoplasms genetics, Papillomavirus Infections genetics, TOR Serine-Threonine Kinases biosynthesis
- Abstract
The rising incidence of human papillomavirus (HPV)-associated malignancies, especially for oropharyngeal cancers, has highlighted the urgent need to understand how the interplay between high-risk HPV oncogenes and carcinogenic exposure results in squamous cell carcinoma (SCC) development. Here, we describe an inducible mouse model expressing high risk HPV-16 E6/E7 oncoproteins in adults, bypassing the impact of these viral genes during development. HPV-16 E6/E7 genes were targeted to the basal squamous epithelia in transgenic mice using a doxycycline inducible cytokeratin 5 promoter (cK5-rtTA) system. After doxycycline induction, both E6 and E7 were highly expressed, resulting in rapid epidermal hyperplasia with a remarkable expansion of the proliferative cell compartment to the suprabasal layers. Surprisingly, in spite of the massive growth of epithelial cells and their stem cell progenitors, HPV-E6/E7 expression was not sufficient to trigger mTOR activation, a key oncogenic driver in HPV-associated malignancies, and malignant progression to SCC. However, these mice develop SCC rapidly after a single exposure to a skin carcinogen, DMBA, which was increased by the prolonged exposure to a tumor promoter, 12-O-tetradecanoylphorbol-13-acetate (TPA). Thus, only few oncogenic hits may be sufficient to induce cancer in E6/E7 expressing cells. All HPV-E6/E7 expressing SCC lesions exhibited increased mTOR activation. Remarkably, rapamycin, an mTOR inhibitor, abolished tumor development when administered to HPV-E6/E7 mice prior to DMBA exposure. Our findings revealed that mTOR inhibition protects HPV-E6/E7 expressing tissues form SCC development upon carcinogen exposure, thus supporting the potential clinical use of mTOR inhibitors as a molecular targeted approach for prevention of HPV-associated malignancies., (© The Author 2016. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2016
- Full Text
- View/download PDF
32. Prevention of irradiation-induced salivary hypofunction by rapamycin in swine parotid glands.
- Author
-
Zhu Z, Pang B, Iglesias-Bartolome R, Wu X, Hu L, Zhang C, Wang J, Gutkind JS, and Wang S
- Subjects
- Animals, Antibiotics, Antineoplastic pharmacology, Apoptosis drug effects, Apoptosis radiation effects, Cell Movement drug effects, Cell Movement radiation effects, Cell Proliferation drug effects, Cell Proliferation radiation effects, Cells, Cultured, Male, Parotid Gland pathology, Parotid Gland radiation effects, Salivary Glands pathology, Salivary Glands radiation effects, Submandibular Gland pathology, Submandibular Gland radiation effects, Swine, Parotid Gland drug effects, Salivary Glands drug effects, Sirolimus pharmacology, Submandibular Gland drug effects
- Abstract
Radiotherapy is commonly used in patients with oral cavity and pharyngeal cancers, usually resulting in irreversible salivary hypofunction. Currently management of radiation damage to salivary glands still remains a great challenge. Recent studies show that activation of mammalian target of rapamycin (mTOR) occurs in salivary gland lesions, making it possible to apply mTOR inhibitor for treatment. Our results indicate inhibition of mTOR by rapamycin significantly alleviated irradiation-induced salivary hypofunction by restoring 46% salivary flow rate and protecting histological structures in swine. Furthermore, rapamycin protected human submandibular gland cell line (HSG) from irradiation-induced cell depletion and loss of cell proliferation capacity. These findings lay the foundation for a new clinical application of rapamycin to prevent irradiation-induced salivary hypofunction.
- Published
- 2016
- Full Text
- View/download PDF
33. Inactivation of a Gα(s)-PKA tumour suppressor pathway in skin stem cells initiates basal-cell carcinogenesis.
- Author
-
Iglesias-Bartolome R, Torres D, Marone R, Feng X, Martin D, Simaan M, Chen M, Weinstein LS, Taylor SS, Molinolo AA, and Gutkind JS
- Subjects
- 3T3 Cells, Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing metabolism, Animals, Cell Cycle Proteins, Cell Differentiation genetics, Cell Line, Cell Proliferation genetics, Chromogranins, GTP-Binding Protein alpha Subunits, Gs metabolism, Gene Expression Regulation, Developmental, HEK293 Cells, Hedgehog Proteins metabolism, Hippo Signaling Pathway, Humans, Kruppel-Like Transcription Factors metabolism, Mice, Mice, Knockout, Phosphoproteins genetics, Phosphoproteins metabolism, Protein Serine-Threonine Kinases antagonists & inhibitors, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, RNA Interference, RNA, Small Interfering, Tumor Suppressor Proteins genetics, Tumor Suppressor Proteins metabolism, YAP-Signaling Proteins, Zinc Finger Protein GLI1, Cell Transformation, Neoplastic genetics, GTP-Binding Protein alpha Subunits, Gs genetics, Hair Follicle cytology, Stem Cells cytology
- Abstract
Genomic alterations in GNAS, the gene coding for the Gαs heterotrimeric G protein, are associated with a large number of human diseases. Here, we explored the role of Gαs on stem cell fate decisions by using the mouse epidermis as a model system. Conditional epidermal deletion of Gnas or repression of PKA signalling caused a remarkable expansion of the stem cell compartment, resulting in rapid basal-cell carcinoma formation. In contrast, inducible expression of active Gαs in the epidermis caused hair follicle stem cell exhaustion and hair loss. Mechanistically, we found that Gαs-PKA disruption promotes the cell autonomous Sonic Hedgehog pathway stimulation and Hippo signalling inhibition, resulting in the non-canonical activation of GLI and YAP1. Our study highlights an important tumour suppressive function of Gαs-PKA, limiting the proliferation of epithelial stem cells and maintaining proper hair follicle homeostasis. These findings could have broad implications in multiple pathophysiological conditions, including cancer.
- Published
- 2015
- Full Text
- View/download PDF
34. Fluorescent, bioactive protein nanoparticles (prodots) for rapid, improved cellular uptake.
- Author
-
Deshapriya IK, Stromer BS, Pattammattel A, Kim CS, Iglesias-Bartolome R, Gonzalez-Fajardo L, Patel V, Gutkind JS, Lu X, and Kumar CV
- Subjects
- Animals, Cattle, Cell Line, Tumor, Fluorescein-5-isothiocyanate metabolism, Fluorescent Dyes metabolism, Humans, Nanoparticles metabolism, Particle Size, Serum Albumin, Bovine metabolism, Spectrometry, Fluorescence methods, Cell Membrane metabolism, Fluorescein-5-isothiocyanate chemistry, Fluorescent Dyes chemistry, Nanoparticles chemistry, Serum Albumin, Bovine chemistry
- Abstract
A simple and effective method for synthesizing highly fluorescent, protein-based nanoparticles (Prodots) and their facile uptake into the cytoplasm of cells is described here. Prodots made from bovine serum albumin (nBSA), glucose oxidase (nGO), horseradish peroxidase (nHRP), catalase (nCatalase), and lipase (nLipase) were found to be 15-50 nm wide and have been characterized by gel electrophoresis, transmission electron microscopy (TEM), circular dichroism (CD), fluorescence spectroscopy, dynamic light scattering (DLS), and optical microscopic methods. Data showed that the secondary structure of the protein in Prodots is retained to a significant extent and specific activities of nGO, nHRP, nCatalase, and nLipase were 80%, 70%, 65%, and 50% of their respective unmodified enzyme activities. Calorimetric studies indicated that the denaturation temperatures of nGO and nBSA increased while those of other Prodots remained nearly unchanged, and accelerated storage half-lives of Prodots at 60 °C increased by 4- to 8-fold. Exposure of nGO and nBSA+ nGO to cells indicated rapid uptake within 1-3 h, accompanied by significant blebbing of the plasma membrane, but no uptake has been noted in the absence of nGO. The presence of nGO/glucose in the media facilitated the uptake, and hydrogen peroxide induced membrane permeability could be responsible for this rapid uptake of Prodots. In control studies, FITC alone did not enter the cell, BSA-FITC was not internalized even in the presence of nGO, and there has been no uptake of nBSA-FITC in the absence of nGO. These are the very first examples of very rapid cellular uptake of fluorescent nanoparticles into cells, particularly nanoparticles made from pure proteins. The current approach is a simple and efficient method for the preparation of bioactive, fluorescent protein nanoparticles of controllable size for cellular imaging, and cell uptake is under the control of two separate chemical triggers.
- Published
- 2015
- Full Text
- View/download PDF
35. mTOR co-targeting in cetuximab resistance in head and neck cancers harboring PIK3CA and RAS mutations.
- Author
-
Wang Z, Martin D, Molinolo AA, Patel V, Iglesias-Bartolome R, Degese MS, Vitale-Cross L, Chen Q, and Gutkind JS
- Subjects
- Animals, Antibiotics, Antineoplastic pharmacology, Carcinoma, Squamous Cell genetics, Cell Line, Tumor, Cell Proliferation drug effects, Cetuximab, Class I Phosphatidylinositol 3-Kinases, Drug Resistance, Neoplasm, ErbB Receptors antagonists & inhibitors, Head and Neck Neoplasms genetics, Humans, Lymphangiogenesis drug effects, Mice, Signal Transduction drug effects, Xenograft Model Antitumor Assays, Antibodies, Monoclonal, Humanized pharmacology, Antineoplastic Combined Chemotherapy Protocols pharmacology, Carcinoma, Squamous Cell drug therapy, Carcinoma, Squamous Cell metabolism, Head and Neck Neoplasms drug therapy, Head and Neck Neoplasms metabolism, Mutation, Phosphatidylinositol 3-Kinases genetics, Sirolimus pharmacology, TOR Serine-Threonine Kinases antagonists & inhibitors, ras Proteins genetics
- Abstract
Background: Cetuximab, a monoclonal blocking antibody against the epidermal growth factor receptor EGFR, has been approved for the treatment of squamous cell carcinomas of the head and neck (HNSCC). However, only few patients display long-term responses, prompting the search for cetuximab resistance mechanisms and new therapeutic options enhancing cetuximab effectiveness., Methods: Cetuximab-sensitive HNSCC cells were retro-engineered to express PIK3CA and RAS oncogenes. These cells and HNSCC cells harboring endogenous PIK3CA and RAS oncogenes were xenografted into mice (n = 10 per group) and studied for their biochemical, antitumor, antiangiogenic, and antilymphangiogenic responses to cetuximab and mTOR targeting agents. All P values are two-sided., Results: Cetuximab treatment of PIK3CA- and RAS-expressing HNSCC xenografts promoted an initial antitumor response, but all tumors relapsed within few weeks. In these tumors, cetuximab did not decrease the activity of mTOR, a downstream signaling target of EGFR, PIK3CA, and RAS. The combined administration of cetuximab and mTOR inhibitors exerted a remarkably increased antitumor activity, particularly in HNSCC cells that are resistant to cetuximab as a single agent. Indeed, cotargeting mTOR together with cetuximab caused a rapid tumor collapse of both PIK3CA- and RAS-expressing HNSCC xenografts (P < .001), concomitant with reduced proliferation (P < .001) and lymphangiogenesis (P < .001)., Conclusion: The presence of PIK3CA and RAS mutations and other alterations affecting the mTOR pathway activity in HNSCC could be exploited to predict the potential resistance to cetuximab, and to select the patients that may benefit the most from the concomitant administration of cetuximab and PI3K and/or mTOR inhibitors as a precision molecular therapeutic option for HNSCC patients., (© The Author 2014. Published by Oxford University Press.)
- Published
- 2014
- Full Text
- View/download PDF
36. Hippo-independent activation of YAP by the GNAQ uveal melanoma oncogene through a trio-regulated rho GTPase signaling circuitry.
- Author
-
Feng X, Degese MS, Iglesias-Bartolome R, Vaque JP, Molinolo AA, Rodrigues M, Zaidi MR, Ksander BR, Merlino G, Sodhi A, Chen Q, and Gutkind JS
- Subjects
- Animals, Cell Cycle Proteins, Cell Growth Processes physiology, Cell Line, Tumor, Female, GTP Phosphohydrolases genetics, GTP-Binding Protein alpha Subunits metabolism, GTP-Binding Protein alpha Subunits, Gq-G11, Gene Knockdown Techniques, HEK293 Cells, Heterografts, Hippo Signaling Pathway, Humans, Melanoma enzymology, Melanoma metabolism, Mice, Mice, Inbred NOD, Mutation, Nuclear Proteins metabolism, Protein Serine-Threonine Kinases genetics, Signal Transduction, Skin Neoplasms, Transcription Factors metabolism, Transfection, Uveal Neoplasms enzymology, Uveal Neoplasms metabolism, rho GTP-Binding Proteins genetics, rho GTP-Binding Proteins metabolism, Melanoma, Cutaneous Malignant, Uveal Melanoma, GTP Phosphohydrolases metabolism, GTP-Binding Protein alpha Subunits genetics, Melanoma genetics, Nuclear Proteins genetics, Protein Serine-Threonine Kinases metabolism, Transcription Factors genetics, Uveal Neoplasms genetics
- Abstract
Mutually exclusive activating mutations in the GNAQ and GNA11 oncogenes, encoding heterotrimeric Gαq family members, have been identified in ∼ 83% and ∼ 6% of uveal and skin melanomas, respectively. However, the molecular events underlying these GNAQ-driven malignancies are not yet defined, thus limiting the ability to develop cancer-targeted therapies. Here, we focused on the transcriptional coactivator YAP, a critical component of the Hippo signaling pathway that controls organ size. We found that Gαq stimulates YAP through a Trio-Rho/Rac signaling circuitry promoting actin polymerization, independently of phospholipase Cβ and the canonical Hippo pathway. Furthermore, we show that Gαq promotes the YAP-dependent growth of uveal melanoma cells, thereby identifying YAP as a suitable therapeutic target in uveal melanoma, a GNAQ/GNA11-initiated human malignancy., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
37. Exploiting the head and neck cancer oncogenome: widespread PI3K-mTOR pathway alterations and novel molecular targets.
- Author
-
Iglesias-Bartolome R, Martin D, and Gutkind JS
- Subjects
- Carcinoma, Squamous Cell genetics, Carcinoma, Squamous Cell pathology, Cell Cycle drug effects, Cell Cycle genetics, Elafin antagonists & inhibitors, Gene Expression Regulation, Neoplastic drug effects, Genomic Instability, Head and Neck Neoplasms genetics, Head and Neck Neoplasms pathology, Humans, Molecular Targeted Therapy, Signal Transduction drug effects, TOR Serine-Threonine Kinases antagonists & inhibitors, Carcinoma, Squamous Cell drug therapy, Elafin metabolism, Head and Neck Neoplasms drug therapy, Protein Kinase Inhibitors administration & dosage, TOR Serine-Threonine Kinases metabolism
- Abstract
Summary: Two studies published in this issue of Cancer Discovery describe the emerging mutational landscape of head and neck squamous cell carcinomas (HNSCC) and their genomic and epigenetic alterations, thus identifying novel actionable cancer drivers and predictive biomarkers for targeted therapies. Most genomic alterations in HNSCC converge in a handful of molecular pathways, resulting in cell-cycle deregulation, genomic instability, cell differentiation defects, and persistent mitogenic signaling, the latter involving aberrant phosphoinositide 3-kinase (PI3K)/mTOR pathway activation, thereby rendering HNSCC responsive to PI3K/mTOR inhibitors. Cancer Discov; 3(7); 722-5. ©2013 AACR.
- Published
- 2013
- Full Text
- View/download PDF
38. Control of the epithelial stem cell epigenome: the shaping of epithelial stem cell identity.
- Author
-
Iglesias-Bartolome R, Callejas-Valera JL, and Gutkind JS
- Subjects
- Animals, Chromatin genetics, Chromatin metabolism, Chromatin pathology, Cyclin-Dependent Kinase Inhibitor p16 metabolism, DNA Methylation, Epigenomics, Histones chemistry, Histones metabolism, Humans, Methylation, Tumor Suppressor Protein p14ARF metabolism, Cell Differentiation genetics, Epigenesis, Genetic, Epithelial Cells cytology, Epithelial Cells metabolism, Stem Cells cytology, Stem Cells metabolism
- Abstract
The squamous epithelium covering the skin and oral mucosa relies on epithelial stem cells for tissue renewal. Dynamic changes in DNA methylation, histone methylation and acetylation, and higher order chromatin structure are required to preserve their self-renewal capacity while orchestrating the timely execution of cell differentiation programs. This complex network of epigenetic modifications shapes the epithelial stem cell identity and fate. Pathological alterations can be perceived by aberrant chromatin sensors, such as the INK4/ARF locus, which initiate tumor suppressive cell senescence programs, and can often result in epithelial stem cell exhaustion. Unveiling the mechanisms controlling the epigenome in epithelial stem cells may help protect against the loss of their tissue regenerative capacity, thereby preventing premature aging without increasing cancer risk., (Copyright © 2013. Published by Elsevier Ltd.)
- Published
- 2013
- Full Text
- View/download PDF
39. A genome-wide RNAi screen reveals a Trio-regulated Rho GTPase circuitry transducing mitogenic signals initiated by G protein-coupled receptors.
- Author
-
Vaqué JP, Dorsam RT, Feng X, Iglesias-Bartolome R, Forsthoefel DJ, Chen Q, Debant A, Seeger MA, Ksander BR, Teramoto H, and Gutkind JS
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation, Clozapine analogs & derivatives, Clozapine pharmacology, Drosophila genetics, Drosophila Proteins genetics, Drosophila Proteins metabolism, Enzyme Activation, Female, GTP-Binding Protein alpha Subunits metabolism, GTP-Binding Protein alpha Subunits, Gq-G11, Gene Knockdown Techniques, Guanine Nucleotide Exchange Factors metabolism, Humans, Mice, Mice, Nude, Mitogen-Activated Protein Kinases metabolism, Mitogens pharmacology, NIH 3T3 Cells, Neoplasm Transplantation, Neoplasms pathology, Protein Serine-Threonine Kinases metabolism, RNA Interference, Receptors, G-Protein-Coupled genetics, Guanine Nucleotide Exchange Factors physiology, Mitosis, Protein Serine-Threonine Kinases physiology, Receptors, G-Protein-Coupled metabolism, Signal Transduction, Transcription Factor AP-1 metabolism, rho GTP-Binding Proteins metabolism
- Abstract
Activating mutations in GNAQ and GNA11, encoding members of the Gα(q) family of G protein α subunits, are the driver oncogenes in uveal melanoma, and mutations in Gq-linked G protein-coupled receptors have been identified recently in numerous human malignancies. How Gα(q) and its coupled receptors transduce mitogenic signals is still unclear because of the complexity of signaling events perturbed upon Gq activation. Using a synthetic-biology approach and a genome-wide RNAi screen, we found that a highly conserved guanine nucleotide exchange factor, Trio, is essential for activating Rho- and Rac-regulated signaling pathways acting on JNK and p38, and thereby transducing proliferative signals from Gα(q) to the nucleus independently of phospholipase C-β. Indeed, whereas many biological responses elicited by Gq depend on the transient activation of second-messenger systems, Gq utilizes a hard-wired protein-protein-interaction-based signaling circuitry to achieve the sustained stimulation of proliferative pathways, thereby controlling normal and aberrant cell growth., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
40. Exploiting the mTOR paradox for disease prevention.
- Author
-
Iglesias-Bartolome R and Gutkind JS
- Subjects
- Humans, TOR Serine-Threonine Kinases metabolism, Antineoplastic Agents therapeutic use, Neoplasms prevention & control, TOR Serine-Threonine Kinases antagonists & inhibitors
- Published
- 2012
- Full Text
- View/download PDF
41. mTOR inhibition prevents epithelial stem cell senescence and protects from radiation-induced mucositis.
- Author
-
Iglesias-Bartolome R, Patel V, Cotrim A, Leelahavanichkul K, Molinolo AA, Mitchell JB, and Gutkind JS
- Subjects
- Animals, Carcinoma, Squamous Cell enzymology, Carcinoma, Squamous Cell pathology, Cell Compartmentation drug effects, Cell Compartmentation radiation effects, Cell Death drug effects, Cell Death radiation effects, Cell Proliferation drug effects, Cell Proliferation radiation effects, Cells, Cultured, Clone Cells, Epithelial Cells drug effects, Epithelial Cells enzymology, Epithelial Cells radiation effects, Head and Neck Neoplasms enzymology, Head and Neck Neoplasms pathology, Humans, Keratinocytes drug effects, Keratinocytes enzymology, Keratinocytes pathology, Keratinocytes radiation effects, Mice, Mouth Mucosa drug effects, Mouth Mucosa pathology, Mouth Mucosa radiation effects, Mucositis enzymology, Mucositis pathology, Oxidative Stress drug effects, Oxidative Stress radiation effects, Radiation Injuries enzymology, Radiation Injuries pathology, Radiation, Ionizing, Sirolimus pharmacology, Stem Cells drug effects, Stem Cells enzymology, Stem Cells radiation effects, Superoxide Dismutase metabolism, TOR Serine-Threonine Kinases metabolism, Cellular Senescence drug effects, Cellular Senescence radiation effects, Cytoprotection drug effects, Cytoprotection radiation effects, Epithelial Cells pathology, Mucositis prevention & control, Radiation Injuries prevention & control, Stem Cells pathology, TOR Serine-Threonine Kinases antagonists & inhibitors
- Abstract
The integrity of the epidermis and mucosal epithelia is highly dependent on resident self-renewing stem cells, which makes them vulnerable to physical and chemical insults compromising the repopulating capacity of the epithelial stem cell compartment. This is frequently the case in cancer patients receiving radiation or chemotherapy, many of whom develop mucositis, a debilitating condition involving painful and deep mucosal ulcerations. Here, we show that inhibiting the mammalian target of rapamycin (mTOR) with rapamycin increases the clonogenic capacity of primary human oral keratinocytes and their resident self-renewing cells by preventing stem cell senescence. This protective effect of rapamycin is mediated by the increase in expression of mitochondrial superoxide dismutase (MnSOD), and the consequent inhibition of ROS formation and oxidative stress. mTOR inhibition also protects from the loss of proliferative basal epithelial stem cells upon ionizing radiation in vivo, thereby preserving the integrity of the oral mucosa and protecting from radiation-induced mucositis., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
42. Nuclear mapping of nanodrug delivery systems in dynamic cellular environments.
- Author
-
Bhirde AA, Kapoor A, Liu G, Iglesias-Bartolome R, Jin A, Zhang G, Xing R, Lee S, Leapman RD, Gutkind JS, and Chen X
- Subjects
- Algorithms, Cell Line, Tumor, Cell Nucleus metabolism, Cell Nucleus pathology, Humans, Molecular Imaging methods, Doxorubicin administration & dosage, Doxorubicin pharmacokinetics, Microscopy, Fluorescence methods, Nanocapsules ultrastructure, Neoplasms, Experimental metabolism, Neoplasms, Experimental pathology, Pattern Recognition, Automated methods
- Abstract
Nanoformulations have shown great promise for delivering chemotherapeutics and hold tremendous clinical relevance. However nuclear mapping of the chemodrugs is important to predict the success of the nanoformulation. In this study fluorescence microscopy and a subcellular tracking algorithm were used to map the diffusion of chemotherapeutic drugs in cancer cells. Positively charged nanoparticles efficiently carried the chemodrug across the cell membrane. The algorithm helped map free drug and drug-loaded nanoparticles, revealing a varying nuclear diffusion pattern of the chemotherapeutics in drug-sensitive and -resistant cells in a live dynamic cellular environment. While the drug-sensitive cells showed an exponential uptake of the drug with time, resistant cells showed random and asymmetric drug distribution. Moreover nanoparticles carrying the drug remained in the perinuclear region, while the drug accumulated in the cell nuclei. The tracking approach has enabled us to predict the therapeutic success of different nanoscale formulations of doxorubicin.
- Published
- 2012
- Full Text
- View/download PDF
43. Signaling circuitries controlling stem cell fate: to be or not to be.
- Author
-
Iglesias-Bartolome R and Gutkind JS
- Subjects
- Adult, Adult Stem Cells cytology, Adult Stem Cells physiology, Animals, Cell Differentiation, Cell Transformation, Neoplastic, Homeostasis, Humans, Mice, Stem Cells physiology, Signal Transduction, Stem Cells cytology
- Abstract
The integration of extrinsic and intrinsic signals is required to preserve the self-renewal and tissue regenerative capacity of adult stem cells, while protecting them from malignant conversion or loss of proliferative potential by death, differentiation or senescence. Here we review emerging signaling circuitries regulating stem cell fate, with emphasis on epithelial stem cells. Wnt, mTOR, GPCRs, Notch, Rho GTPases, YAP and DNA and histone methylases are some of the mechanisms that allow stem cells to balance their regenerative potential and the initiation of terminal differentiation programs, guaranteeing appropriate tissue homeostasis. Understanding the signaling circuitries regulating stem cell fate decisions might provide important insights into cancer initiation and numerous human pathologies that involve the progressive loss of tissue-specific adult stem cells., (Published by Elsevier Ltd.)
- Published
- 2011
- Full Text
- View/download PDF
44. Cellular systems for studying human oral squamous cell carcinomas.
- Author
-
Patel V, Iglesias-Bartolome R, Siegele B, Marsh CA, Leelahavanichkul K, Molinolo AA, and Gutkind JS
- Subjects
- Cell Separation, Cells, Cultured, Epithelial Cells physiology, Humans, Keratinocytes cytology, Mouth Mucosa cytology, Stem Cells cytology, Carcinoma, Squamous Cell pathology, Mouth Neoplasms pathology
- Abstract
The human oral squamous epithelium plays an important role in maintaining a barrier function against mechanical, physical, and pathological injury. However, the self-renewing cells residing on the basement membrane of the epithelium can give rise to oral squamous cell carcinomas (OSCC), now the sixth most common cancer in the developed world, which is still associated with poor prognosis. This is due, in part, to the limited availability of well-defined culture systems for studying oral epithelial cell biology, which could advance our understanding of the molecular basis of OSCC. Here, we describe methods to successfully isolate large cultures of human oral epithelial cells and fibroblasts from small pieces of donor tissues for use in techniques such as three-dimensional cultures and animal grafts to validate genes suspected of playing a role in OSCC development and progression. Finally, the use of isolated oral epithelial cells in generating iPS cells is discussed which holds promise in the field of oral regenerative medicine.
- Published
- 2011
- Full Text
- View/download PDF
45. Combining portable Raman probes with nanotubes for theranostic applications.
- Author
-
Bhirde AA, Liu G, Jin A, Iglesias-Bartolome R, Sousa AA, Leapman RD, Gutkind JS, Lee S, and Chen X
- Abstract
Recently portable Raman probes have emerged along with a variety of applications, including carbon nanotube (CNT) characterization. Aqueous dispersed CNTs have shown promise for biomedical applications such as drug/gene delivery vectors, photo-thermal therapy, and photoacoustic imaging. In this study we report the simultaneous detection and irradiation of carbon nanotubes in 2D monolayers of cancer cells and in 3D spheroids using a portable Raman probe. A portable handheld Raman instrument was utilized for dual purposes: as a CNT detector and as an irradiating laser source. Single-walled carbon nanotubes (SWCNTs) and multi-walled carbon nanotubes (MWCNTs) were dispersed aqueously using a lipid-polymer (LP) coating, which formed highly stable dispersions both in buffer and cell media. The LP coated SWCNT and MWCNT aqueous dispersions were characterized by atomic force microscopy, transmission electron microscopy, dynamic light scattering, Fourier transform infrared spectroscopy and Raman spectroscopy. The cellular uptake of the LP-dispersed SWCNTs and MWCNTs was observed using confocal microscopy, and fluorescein isothiocyanate (FITC)-nanotube conjugates were found to be internalized by ovarian cancer cells by using Z-stack fluorescence confocal imaging. Biocompatibility of SWCNTs and MWCNTs was assessed using a cell viability MTT assay, which showed that the nanotube dispersions did not hinder the proliferation of ovarian cancer cells at the dosage tested. Ovarian cancer cells treated with SWCNTs and MWCNTs were simultaneously detected and irradiated live in 2D layers of cancer cells and in 3D environments using the portable Raman probe. An apoptotic terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay carried out after laser irradiation confirmed that cell death occurred only in the presence of nanotube dispersions. We show for the first time that both SWCNTs and MWCNTs can be selectively irradiated and detected in cancer cells using a simple handheld Raman instrument. This approach could potentially be used to treat various diseases, including cancer.
- Published
- 2011
- Full Text
- View/download PDF
46. Keeping the epidermal stem cell niche in shape.
- Author
-
Iglesias-Bartolome R and Gutkind JS
- Abstract
Recently in Nature Cell Biology,Connelly et al. (2010) identified biomechanical sensing mechanisms that link the physical shape of the stem cell microenvironment to epithelial stem cell fate decisions. Ultimately, the integration of extrinsic and intrinsic signals controls stem cell self-renewal or differentiation., (Copyright 2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.