Graziani, V.
Crosas-Molist, E.
George, S.L.
Sanz-Moreno, V.
(2024). Organelle adaptations in response to mechanical forces during tumour dissemination. Curr opin cell biol,
Vol.88,
p. 102345.
show abstract
Cell migration plays a pivotal role in various biological processes including cancer dissemination and successful metastasis, where the role of mechanical signals is increasingly acknowledged. This review focuses on the intricate mechanisms through which cancer cells modulate their migratory strategies via organelle adaptations in response to the extracellular matrix (ECM). Specifically, the nucleus and mitochondria emerge as pivotal mediators in this process. These organelles serve as sensors, translating mechanical stimuli into rapid metabolic alterations that sustain cell migration. Importantly, prolonged exposure to such stimuli can induce transcriptional or epigenetic changes, ultimately enhancing metastatic traits. Deciphering the intricate interplay between ECM properties and organelle adaptations not only advances our understanding of cytoskeletal dynamics but also holds promise for the development of innovative anti-metastatic therapeutic strategies..
Habib, S.
Osborn, G.
Willsmore, Z.
Chew, M.W.
Jakubow, S.
Fitzpatrick, A.
Wu, Y.
Sinha, K.
Lloyd-Hughes, H.
Geh, J.L.
MacKenzie-Ross, A.D.
Whittaker, S.
Sanz-Moreno, V.
Lacy, K.E.
Karagiannis, S.N.
Adams, R.
(2024). Tumor associated macrophages as key contributors and targets in current and future therapies for melanoma. Expert rev clin immunol,
Vol.20
(8),
pp. 895-911.
show abstract
INTRODUCTION: Despite the success of immunotherapies for melanoma in recent years, there remains a significant proportion of patients who do not yet derive benefit from available treatments. Immunotherapies currently licensed for clinical use target the adaptive immune system, focussing on Tcell interactions and functions. However, the most prevalent immune cells within the tumor microenvironment (TME) of melanoma are macrophages, a diverse immune cell subset displaying high plasticity, to which no current therapies are yet directly targeted. Macrophages have been shown not only to activate the adaptive immune response, and enhance cancer cell killing, but, when influenced by factors within the TME of melanoma, these cells also promote melanoma tumorigenesis and metastasis. AREAS COVERED: We present a review of the most up-to-date literatureavailable on PubMed, focussing on studies from within the last 10 years. We also include data from ongoing and recent clinical trials targeting macrophages in melanoma listed on clinicaltrials.gov. EXPERT OPINION: Understanding the multifaceted role of macrophages in melanoma, including their interactions with immune and cancer cells, the influence of current therapies on macrophage phenotype and functions and how macrophages could be targeted with novel treatment approaches, are all critical for improving outcomes for patients with melanoma..
Jung-Garcia, Y.
Maiques, O.
Monger, J.
Rodriguez-Hernandez, I.
Fanshawe, B.
Domart, M.-.
Renshaw, M.J.
Marti, R.M.
Matias-Guiu, X.
Collinson, L.M.
Sanz-Moreno, V.
Carlton, J.G.
(2023). LAP1 supports nuclear adaptability during constrained melanoma cell migration and invasion. Nat cell biol,
Vol.25
(1),
pp. 108-119.
show abstract
Metastasis involves dissemination of cancer cells away from a primary tumour and colonization at distal sites. During this process, the mechanical properties of the nucleus must be tuned since they pose a challenge to the negotiation of physical constraints imposed by the microenvironment and tissue structure. We discovered increased expression of the inner nuclear membrane protein LAP1 in metastatic melanoma cells, at the invasive front of human primary melanoma tumours and in metastases. Human cells express two LAP1 isoforms (LAP1B and LAP1C), which differ in their amino terminus. Here, using in vitro and in vivo models that recapitulate human melanoma progression, we found that expression of the shorter isoform, LAP1C, supports nuclear envelope blebbing, constrained migration and invasion by allowing a weaker coupling between the nuclear envelope and the nuclear lamina. We propose that LAP1 renders the nucleus highly adaptable and contributes to melanoma aggressiveness..
Barcelo, J.
Samain, R.
Sanz-Moreno, V.
(2023). Preclinical to clinical utility of ROCK inhibitors in cancer. Trends cancer,
Vol.9
(3),
pp. 250-263.
show abstract
ROCK belongs to the AGC family of Ser/Thr protein kinases that are involved in many cellular processes. ROCK-driven actomyosin contractility regulates cytoskeletal dynamics underpinning cell migration, proliferation, and survival in many cancer types. ROCK1/2 play key protumorigenic roles in several subtypes and stages of cancer development. Therefore, successfully targeting ROCK and its downstream effectors presents an interesting avenue for cancer treatment. Because local use of ROCK inhibitors will reduce the side effects of systemic administration, we propose different therapeutic strategies and latest-generation ROCK inhibitors for use in the clinic..
Samain, R.
Maiques, O.
Monger, J.
Lam, H.
Candido, J.
George, S.
Ferrari, N.
KohIhammer, L.
Lunetto, S.
Varela, A.
Orgaz, J.L.
Vilardell, F.
Olsina, J.J.
Matias-Guiu, X.
Sarker, D.
Biddle, A.
Balkwill, F.R.
Eyles, J.
Wilkinson, R.W.
Kocher, H.M.
Calvo, F.
Wells, C.M.
Sanz-Moreno, V.
(2023). CD73 controls Myosin II-driven invasion, metastasis, and immunosuppression in amoeboid pancreatic cancer cells. Sci adv,
Vol.9
(42),
p. eadi0244.
show abstract
full text
Pancreatic ductal adenocarcinoma (PDAC) has a very poor prognosis because of its high propensity to metastasize and its immunosuppressive microenvironment. Using a panel of pancreatic cancer cell lines, three-dimensional (3D) invasion systems, microarray gene signatures, microfluidic devices, mouse models, and intravital imaging, we demonstrate that ROCK-Myosin II activity in PDAC cells supports a transcriptional program conferring amoeboid invasive and immunosuppressive traits and in vivo metastatic abilities. Moreover, we find that immune checkpoint CD73 is highly expressed in amoeboid PDAC cells and drives their invasive, metastatic, and immunomodulatory traits. Mechanistically, CD73 activates RhoA-ROCK-Myosin II downstream of PI3K. Tissue microarrays of human PDAC biopsies combined with bioinformatic analysis reveal that rounded-amoeboid invasive cells with high CD73-ROCK-Myosin II activity and their immunosuppressive microenvironment confer poor prognosis to patients. We propose targeting amoeboid PDAC cells as a therapeutic strategy..
Crosas-Molist, E.
Graziani, V.
Maiques, O.
Pandya, P.
Monger, J.
Samain, R.
George, S.L.
Malik, S.
Salise, J.
Morales, V.
Le Guennec, A.
Atkinson, R.A.
Marti, R.M.
Matias-Guiu, X.
Charras, G.
Conte, M.R.
Elosegui-Artola, A.
Holt, M.
Sanz-Moreno, V.
(2023). AMPK is a mechano-metabolic sensor linking cell adhesion and mitochondrial dynamics to Myosin-dependent cell migration. Nat commun,
Vol.14
(1),
p. 2740.
show abstract
Cell migration is crucial for cancer dissemination. We find that AMP-activated protein kinase (AMPK) controls cell migration by acting as an adhesion sensing molecular hub. In 3-dimensional matrices, fast-migrating amoeboid cancer cells exert low adhesion/low traction linked to low ATP/AMP, leading to AMPK activation. In turn, AMPK plays a dual role controlling mitochondrial dynamics and cytoskeletal remodelling. High AMPK activity in low adhering migratory cells, induces mitochondrial fission, resulting in lower oxidative phosphorylation and lower mitochondrial ATP. Concurrently, AMPK inactivates Myosin Phosphatase, increasing Myosin II-dependent amoeboid migration. Reducing adhesion or mitochondrial fusion or activating AMPK induces efficient rounded-amoeboid migration. AMPK inhibition suppresses metastatic potential of amoeboid cancer cells in vivo, while a mitochondrial/AMPK-driven switch is observed in regions of human tumours where amoeboid cells are disseminating. We unveil how mitochondrial dynamics control cell migration and suggest that AMPK is a mechano-metabolic sensor linking energetics and the cytoskeleton..
Crosas-Molist, E.
Samain, R.
Kohlhammer, L.
Orgaz, J.L.
George, S.L.
Maiques, O.
Barcelo, J.
Sanz-Moreno, V.
(2022). Rho GTPase signaling in cancer progression and dissemination. Physiol rev,
Vol.102
(1),
pp. 455-510.
show abstract
Rho GTPases are a family of small G proteins that regulate a wide array of cellular processes related to their key roles controlling the cytoskeleton. Cancer is a multistep disease caused by the accumulation of genetic mutations and epigenetic alterations, from the initial stages of cancer development when cells in normal tissues undergo transformation, to the acquisition of invasive and metastatic traits, responsible for a large number of cancer related deaths. In this review, we discuss the role of Rho GTPase signaling in cancer in every step of disease progression. Rho GTPases contribute to tumor initiation and progression, by regulating proliferation and apoptosis, but also metabolism, senescence, and cancer cell stemness. Rho GTPases play a major role in cell migration and in the metastatic process. They are also involved in interactions with the tumor microenvironment and regulate inflammation, contributing to cancer progression. After years of intensive research, we highlight the importance of relevant models in the Rho GTPase field, and we reflect on the therapeutic opportunities arising for cancer patients..
Maiques, O.
Sanz-Moreno, V.
(2022). Location, location, location: Melanoma cells "living at the edge". Exp dermatol,
Vol.31
(1),
pp. 82-88.
show abstract
Abnormal cell migration and invasion underlie metastatic dissemination, one of the major challenges for cancer treatment. Melanoma is one of the deadliest and most aggressive forms of skin cancer due in part to its migratory and metastatic potential. Cancer cells use a variety of migratory strategies regulated by cytoskeletal remodelling. In particular, we discuss the importance of amoeboid invasive melanoma strategies, since they have been identified at the edge of human melanomas. We hypothesize that the presence of amoeboid melanoma cells will favour tumor progression since they are invasive and metastatic; they support immunosuppression; they harbour cancer stem cell properties and they are involved in therapy resistance. The Rho-ROCK-Myosin II pathway is key to maintain amoeboid melanoma invasion but this pathway is further regulated by pro-tumorigenic/pro-metastatic/pro-survival signalling pathways such as JAK-STAT3, TGFβ-SMAD, NF-κB, Wnt11/5-FDZ7 and BRAFV600E -MEK-ERK. These pathways support amoeboid behaviour and are actionable in the clinic. After melanoma wide surgical margin removal, we propose that possible remaining melanoma cells should be eradicated using anti-amoeboid therapies..
Graziani, V.
Rodriguez-Hernandez, I.
Maiques, O.
Sanz-Moreno, V.
(2022). The amoeboid state as part of the epithelial-to-mesenchymal transition programme. Trends cell biol,
Vol.32
(3),
pp. 228-242.
show abstract
Cell migration is essential for many biological processes, while abnormal cell migration is characteristic of cancer cells. Epithelial cells become motile by undergoing epithelial-to-mesenchymal transition (EMT), and mesenchymal cells increase migration speed by adopting amoeboid features. This review highlights how amoeboid behaviour is not merely a migration mode but rather a cellular state - within the EMT spectra - by which cancer cells survive, invade and colonise challenging microenvironments. Molecular biomarkers and physicochemical triggers associated with amoeboid behaviour are discussed, including an amoeboid associated tumour microenvironment. We reflect on how amoeboid characteristics support metastasis and how their liabilities could turn into therapeutic opportunities..
Barcelo, J.
Sanz-Moreno, V.
(2022). NECTIN1 is a melanoma metastasis suppressor gene. Nat genet,
Vol.54
(12),
pp. 1776-1777.
Maiques, O.
Sanz-Moreno, V.
(2022). Multiplex chromogenic immunohistochemistry to stain and analyze paraffin tissue sections from the mouse or human. Star protoc,
Vol.3
(4),
p. 101879.
show abstract
full text
Here we describe a multiplex chromogenic immunohistochemistry platform to stain and analyze two markers in paraffin tissue sections from mouse or human. The basis of the protocol is a series of stripping and re-probing steps with subsequent image analysis, which allows the user to perform multiplex imaging in a reliable and affordable manner. Here, we describe specific usage to assess the levels of PD-L1 in tumor-associated macrophages. We have used different antibodies and assessed this protocol for up to five consecutive antibodies per slide. For complete details on the use and execution of this protocol, please refer to Orgaz et al. (2020).1..
George, S.
Georgouli, M.
Sanz-Moreno, V.
(2022). Protocol to drive human monocyte-to-macrophage polarization in vitro using tumor conditioned media. Star protoc,
Vol.3
(4),
p. 101666.
show abstract
full text
Tumor-associated macrophages (TAMs) are key contributors to antitumor immunity. Here, we present a protocol to drive human monocyte-macrophage differentiation using tumor-derived conditioned media, followed by phenotypic and functional characterization of TAMs in vitro. We describe CD14+ cell isolation from healthy human blood, and detail the procedure to induce macrophage polarization. Finally, we outline morphological assessment of macrophages, and validation of their functional behaviors with a tumor cell killing assay. This translatable-based approach can be applied to different cancer cell types. For complete details on the use and execution of this protocol, please refer to Georgouli et al. (2019)..
Roy-Luzarraga, M.
Reynolds, L.E.
de Luxán-Delgado, B.
Maiques, O.
Wisniewski, L.
Newport, E.
Rajeeve, V.
Drake, R.J.
Gómez-Escudero, J.
Richards, F.M.
Weller, C.
Dormann, C.
Meng, Y.-.
Vermeulen, P.B.
Saur, D.
Sanz-Moreno, V.
Wong, P.-.
Géraud, C.
Cutillas, P.R.
Hodivala-Dilke, K.
(2022). Suppression of Endothelial Cell FAK Expression Reduces Pancreatic Ductal Adenocarcinoma Metastasis after Gemcitabine Treatment. Cancer res,
Vol.82
(10),
pp. 1909-1925.
show abstract
UNLABELLED: Despite substantial advances in the treatment of solid cancers, resistance to therapy remains a major obstacle to prolonged progression-free survival. Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive cancers, with a high level of liver metastasis. Primary PDAC is highly hypoxic, and metastases are resistant to first-line treatment, including gemcitabine. Recent studies have indicated that endothelial cell (EC) focal adhesion kinase (FAK) regulates DNA-damaging therapy-induced angiocrine factors and chemosensitivity in primary tumor models. Here, we show that inducible loss of EC-FAK in both orthotopic and spontaneous mouse models of PDAC is not sufficient to affect primary tumor growth but reduces liver and lung metastasis load and improves survival rates in gemcitabine-treated, but not untreated, mice. EC-FAK loss did not affect primary tumor angiogenesis, tumor blood vessel leakage, or early events in metastasis, including the numbers of circulating tumor cells, tumor cell homing, or metastatic seeding. Phosphoproteomics analysis showed a downregulation of the MAPK, RAF, and PAK signaling pathways in gemcitabine-treated FAK-depleted ECs compared with gemcitabine-treated wild-type ECs. Moreover, low levels of EC-FAK correlated with increased survival and reduced relapse in gemcitabine-treated patients with PDAC, supporting the clinical relevance of these findings. Altogether, we have identified a new role of EC-FAK in regulating PDAC metastasis upon gemcitabine treatment that impacts outcome. SIGNIFICANCE: These findings establish the potential utility of combinatorial endothelial cell FAK targeting together with gemcitabine in future clinical applications to control metastasis in patients with pancreatic ductal adenocarcinoma..
Perdrix Rosell, A.
Maiques, O.
Martin, J.A.
Chakravarty, P.
Ombrato, L.
Sanz-Moreno, V.
Malanchi, I.
(2022). Early functional mismatch between breast cancer cells and their tumour microenvironment suppresses long term growth. Cancer lett,
Vol.544,
p. 215800.
show abstract
Cancer cells thrive when embedded in a fine-tuned cellular and extracellular environment or tumour microenvironment (TME). There is a general understanding of a co-evolution between cancer cells and their surrounding TME, pointing at a functional connection between cancer cells characteristics and the perturbations induced in their surrounding tissue. However, it has never been formally proven whether this functional connection needs to be set from the start or if aggressive cancer cells always dominate their microenvironmental any point in time. This would require a dedicated experimental setting where malignant cells are challenged to grow in a different TME from the one they would naturally create. Here we generated an experimental setting where we transiently perturb the secretory profile of aggressive breast cancer cells without affecting their intrinsic growth ability, which led to the initial establishment of an atypical TME. Interestingly, even if initially tumours are formed, this atypical TME evolves to impair long term in vivo cancer growth. Using a combination of in vivo transcriptomics, protein arrays and in vitro co-cultures, we found that the atypical TME culminates in the infiltration of macrophages with STAT1high activity. These macrophages show strong anti-tumoural functions which reduce long-term tumour growth, despite lacking canonical M1 markers. Importantly, gene signatures of the mesenchymal compartment of the TME, as well as the anti-tumoural macrophages, show striking prognostic power that correlates with less aggressive human breast cancers..
George, S.
Martin, J.A.
Graziani, V.
Sanz-Moreno, V.
(2022). Amoeboid migration in health and disease: Immune responses versus cancer dissemination. Front cell dev biol,
Vol.10,
p. 1091801.
show abstract
Cell migration is crucial for efficient immune responses and is aberrantly used by cancer cells during metastatic dissemination. Amoeboid migrating cells use myosin II-powered blebs to propel themselves, and change morphology and direction. Immune cells use amoeboid strategies to respond rapidly to infection or tissue damage, which require quick passage through several barriers, including blood, lymph and interstitial tissues, with complex and varied environments. Amoeboid migration is also used by metastatic cancer cells to aid their migration, dissemination and survival, whereby key mechanisms are hijacked from professionally motile immune cells. We explore important parallels observed between amoeboid immune and cancer cells. We also consider key distinctions that separate the lifespan, state and fate of these cell types as they migrate and/or fulfil their function. Finally, we reflect on unexplored areas of research that would enhance our understanding of how tumour cells use immune cell strategies during metastasis, and how to target these processes..
Adams, R.
Osborn, G.
Mukhia, B.
Laddach, R.
Willsmore, Z.
Chenoweth, A.
Geh, J.L.
MacKenzie Ross, A.D.
Healy, C.
Barber, L.
Tsoka, S.
Sanz-Moreno, V.
Lacy, K.E.
Karagiannis, S.N.
(2022). Influencing tumor-associated macrophages in malignant melanoma with monoclonal antibodies. Oncoimmunology,
Vol.11
(1),
p. 2127284.
show abstract
The application of monoclonal antibodies (mAbs) for the treatment of melanoma has significantly improved the clinical management of this malignancy over the last decade. Currently approved mAbs for melanoma enhance T cell effector immune responses by blocking immune checkpoint molecules PD-L1/PD-1 and CTLA-4. However, more than half of patients do not benefit from treatment. Targeting the prominent myeloid compartment within the tumor microenvironment, and in particular the ever-abundant tumor-associated macrophages (TAMs), may be a promising strategy to complement existing therapies and enhance treatment success. TAMs are a highly diverse and plastic subset of cells whose pro-tumor properties can support melanoma growth, angiogenesis and invasion. Understanding of their diversity, plasticity and multifaceted roles in cancer forms the basis for new promising TAM-centered treatment strategies. There are multiple mechanisms by which macrophages can be targeted with antibodies in a therapeutic setting, including by depletion, inhibition of specific pro-tumor properties, differential polarization to pro-inflammatory states and enhancement of antitumor immune functions. Here, we discuss TAMs in melanoma, their interactions with checkpoint inhibitor antibodies and emerging mAbs targeting different aspects of TAM biology and their potential to be translated to the clinic..
Candido, J.B.
Maiques, O.
Boxberg, M.
Kast, V.
Peerani, E.
Tomás-Bort, E.
Weichert, W.
Sananes, A.
Papo, N.
Magdolen, V.
Sanz-Moreno, V.
Loessner, D.
(2021). Kallikrein-Related Peptidase 6 Is Associated with the Tumour Microenvironment of Pancreatic Ductal Adenocarcinoma. Cancers (basel),
Vol.13
(16).
show abstract
As cancer-associated factors, kallikrein-related peptidases (KLKs) are components of the tumour microenvironment, which represents a rich substrate repertoire, and considered attractive targets for the development of novel treatments. Standard-of-care therapy of pancreatic cancer shows unsatisfactory results, indicating the need for alternative therapeutic approaches. We aimed to investigate the expression of KLKs in pancreatic cancer and to inhibit the function of KLK6 in pancreatic cancer cells. KLK6, KLK7, KLK8, KLK10 and KLK11 were coexpressed and upregulated in tissues from pancreatic cancer patients compared to normal pancreas. Their high expression levels correlated with each other and were linked to shorter survival compared to low KLK levels. We then validated KLK6 mRNA and protein expression in patient-derived tissues and pancreatic cancer cells. Coexpression of KLK6 with KRT19, αSMA or CD68 was independent of tumour stage, while KLK6 was coexpressed with KRT19 and CD68 in the invasive tumour area. High KLK6 levels in tumour and CD68+ cells were linked to shorter survival. KLK6 inhibition reduced KLK6 mRNA expression, cell metabolic activity and KLK6 secretion and increased the secretion of other serine and aspartic lysosomal proteases. The association of high KLK levels and poor prognosis suggests that inhibiting KLKs may be a therapeutic strategy for precision medicine..
Maiques, O.
Fanshawe, B.
Crosas-Molist, E.
Rodriguez-Hernandez, I.
Volpe, A.
Cantelli, G.
Boehme, L.
Orgaz, J.L.
Mardakheh, F.K.
Sanz-Moreno, V.
Fruhwirth, G.O.
(2021). A preclinical pipeline to evaluate migrastatics as therapeutic agents in metastatic melanoma. Br j cancer,
Vol.125
(5),
pp. 699-713.
show abstract
BACKGROUND: Metastasis is a hallmark of cancer and responsible for most cancer deaths. Migrastatics were defined as drugs interfering with all modes of cancer cell invasion and thus cancers' ability to metastasise. First anti-metastatic treatments have recently been approved. METHODS: We used bioinformatic analyses of publicly available melanoma databases. Experimentally, we performed in vitro target validation (including 2.5D cell morphology analysis and mass spectrometric analysis of RhoA binding partners), developed a new traceable spontaneously metastasising murine melanoma model for in vivo validation, and employed histology (haematoxylin/eosin and phospho-myosin II staining) to confirm drug action in harvested tumour tissues. RESULTS: Unbiased and targeted bioinformatic analyses identified the Rho kinase (ROCK)-myosin II pathway and its various components as potentially relevant targets in melanoma. In vitro validation demonstrated redundancy of several RhoGEFs upstream of RhoA and confirmed ROCK as a druggable target downstream of RhoA. The anti-metastatic effects of two ROCK inhibitors were demonstrated through in vivo melanoma metastasis tracking and inhibitor effects also confirmed ex vivo by digital pathology. CONCLUSIONS: We proposed a migrastatic drug development pipeline. As part of the pipeline, we provide a new traceable spontaneous melanoma metastasis model for in vivo quantification of metastasis and anti-metastatic effects by non-invasive imaging..
de Winde, C.M.
George, S.L.
Crosas-Molist, E.
Hari-Gupta, Y.
Arp, A.B.
Benjamin, A.C.
Millward, L.J.
Makris, S.
Carver, A.
Imperatore, V.
Martínez, V.G.
Sanz-Moreno, V.
Acton, S.E.
(2021). Podoplanin drives dedifferentiation and amoeboid invasion of melanoma. Iscience,
Vol.24
(9),
p. 102976.
show abstract
Melanoma is an aggressive skin cancer developing from melanocytes, frequently resulting in metastatic disease. Melanoma cells utilize amoeboid migration as mode of local invasion. Amoeboid invasion is characterized by rounded cell morphology and high actomyosin contractility driven by Rho GTPase signalling. Migrastatic drugs targeting actin polymerization and contractility are therefore a promising treatment option for metastatic melanoma. To predict amoeboid invasion and metastatic potential, biomarkers functionally linked to contractility pathways are needed. The glycoprotein podoplanin drives actomyosin contractility in lymphoid fibroblasts and is overexpressed in many cancers. We show that podoplanin enhances amoeboid invasion in melanoma. Podoplanin expression in murine melanoma drives rounded cell morphology, increasing motility, and invasion in vivo. Podoplanin expression is increased in a subset of dedifferentiated human melanoma, and in vitro is sufficient to upregulate melanoma-associated marker Pou3f2/Brn2. Together, our data define podoplanin as a functional biomarker for dedifferentiated invasive melanoma and a promising migrastatic therapeutic target..
Farrugia, A.J.
Rodríguez, J.
Orgaz, J.L.
Lucas, M.
Sanz-Moreno, V.
Calvo, F.
(2020). CDC42EP5/BORG3 modulates SEPT9 to promote actomyosin function, migration, and invasion. J cell biol,
Vol.219
(9).
show abstract
full text
Fast amoeboid migration is critical for developmental processes and can be hijacked by cancer cells to enhance metastatic dissemination. This migratory behavior is tightly controlled by high levels of actomyosin contractility, but how it is coupled to other cytoskeletal components is poorly understood. Septins are increasingly recognized as novel cytoskeletal components, but details on their regulation and contribution to migration are lacking. Here, we show that the septin regulator Cdc42EP5 is consistently required for amoeboid melanoma cells to invade and migrate into collagen-rich matrices and locally invade and disseminate in vivo. Cdc42EP5 associates with actin structures, leading to increased actomyosin contractility and amoeboid migration. Cdc42EP5 affects these functions through SEPT9-dependent F-actin cross-linking, which enables the generation of F-actin bundles required for the sustained stabilization of highly contractile actomyosin structures. This study provides evidence that Cdc42EP5 is a regulator of cancer cell motility that coordinates actin and septin networks and describes a unique role for SEPT9 in melanoma invasion and metastasis..
Samain, R.
Sanz-Moreno, V.
(2020). Cancer-associated fibroblasts: activin A adds another string to their bow. Embo mol med,
Vol.12
(4),
p. e12102.
show abstract
full text
Non-melanoma skin cancer (NMSC) is characterized by a strong desmoplastic reaction, largely responsible for cancer aggressiveness. Within the tumour microenvironment, cancer-associated fibroblasts (CAFs) play a key role in tumour progression, secretion of extracellular matrix proteins and recruitment of immunosuppressive cells. However, pathways involved in acquisition of CAF phenotype remain unclear. In this issue of EMBO Molecular Medicine, Cangkrama et al describe a new mechanism of fibroblast activation in squamous cell carcinoma. Cancer cell-secreted activin A induces a tumour-promoting phenotype in the fibroblast compartment, with distinct properties compared to TGF-β-activated fibroblasts. Activin A reprograms fibroblasts through transcriptional regulation of mDia2 and reduction of nuclear p53, which favours CAF marker expression, and increases tumour growth and migration. Inhibition of this pathway shows promising results in different models and could offer a new therapeutic strategy in NMSC..
Wong, P.-.
Muñoz-Félix, J.M.
Hijazi, M.
Kim, H.
Robinson, S.D.
De Luxán-Delgado, B.
Rodríguez-Hernández, I.
Maiques, O.
Meng, Y.-.
Meng, Q.
Bodrug, N.
Dukinfield, M.S.
Reynolds, L.E.
Elia, G.
Clear, A.
Harwood, C.
Wang, Y.
Campbell, J.J.
Singh, R.
Zhang, P.
Schall, T.J.
Matchett, K.P.
Henderson, N.C.
Szlosarek, P.W.
Dreger, S.A.
Smith, S.
Jones, J.L.
Gribben, J.G.
Cutillas, P.R.
Meier, P.
Sanz-Moreno, V.
Hodivala-Dilke, K.M.
(2020). Cancer Burden Is Controlled by Mural Cell-β3-Integrin Regulated Crosstalk with Tumor Cells. Cell,
Vol.181
(6),
pp. 1346-1363.e21.
show abstract
Enhanced blood vessel (BV) formation is thought to drive tumor growth through elevated nutrient delivery. However, this observation has overlooked potential roles for mural cells in directly affecting tumor growth independent of BV function. Here we provide clinical data correlating high percentages of mural-β3-integrin-negative tumor BVs with increased tumor sizes but no effect on BV numbers. Mural-β3-integrin loss also enhances tumor growth in implanted and autochthonous mouse tumor models with no detectable effects on BV numbers or function. At a molecular level, mural-cell β3-integrin loss enhances signaling via FAK-p-HGFR-p-Akt-p-p65, driving CXCL1, CCL2, and TIMP-1 production. In particular, mural-cell-derived CCL2 stimulates tumor cell MEK1-ERK1/2-ROCK2-dependent signaling and enhances tumor cell survival and tumor growth. Overall, our data indicate that mural cells can control tumor growth via paracrine signals regulated by β3-integrin, providing a previously unrecognized mechanism of cancer growth control..
Orgaz, J.L.
Crosas-Molist, E.
Sadok, A.
Perdrix-Rosell, A.
Maiques, O.
Rodriguez-Hernandez, I.
Monger, J.
Mele, S.
Georgouli, M.
Bridgeman, V.
Karagiannis, P.
Lee, R.
Pandya, P.
Boehme, L.
Wallberg, F.
Tape, C.
Karagiannis, S.N.
Malanchi, I.
Sanz-Moreno, V.
(2020). Myosin II Reactivation and Cytoskeletal Remodeling as a Hallmark and a Vulnerability in Melanoma Therapy Resistance. Cancer cell,
Vol.37
(1),
pp. 85-103.e9.
show abstract
Despite substantial clinical benefit of targeted and immune checkpoint blockade-based therapies in melanoma, resistance inevitably develops. We show cytoskeletal remodeling and changes in expression and activity of ROCK-myosin II pathway during acquisition of resistance to MAPK inhibitors. MAPK regulates myosin II activity, but after initial therapy response, drug-resistant clones restore myosin II activity to increase survival. High ROCK-myosin II activity correlates with aggressiveness, identifying targeted therapy- and immunotherapy-resistant melanomas. Survival of resistant cells is myosin II dependent, regardless of the therapy. ROCK-myosin II ablation specifically kills resistant cells via intrinsic lethal reactive oxygen species and unresolved DNA damage and limits extrinsic myeloid and lymphoid immunosuppression. Efficacy of targeted therapies and immunotherapies can be improved by combination with ROCK inhibitors..
McLeod, R.
Kumar, R.
Papadatos-Pastos, D.
Mateo, J.
Brown, J.S.
Garces, A.H.
Ruddle, R.
Decordova, S.
Jueliger, S.
Ferraldeschi, R.
Maiques, O.
Sanz-Moreno, V.
Jones, P.
Traub, S.
Halbert, G.
Mellor, S.
Swales, K.E.
Raynaud, F.I.
Garrett, M.D.
Banerji, U.
(2020). First-in-Human Study of AT13148, a Dual ROCK-AKT Inhibitor in Patients with Solid Tumors. Clin cancer res,
Vol.26
(18),
pp. 4777-4784.
show abstract
full text
PURPOSE: AT13148 is an oral AGC kinase inhibitor, which potently inhibits ROCK and AKT kinases. In preclinical models, AT13148 has been shown to have antimetastatic and antiproliferative activity. PATIENTS AND METHODS: The trial followed a rolling six design during dose escalation. An intrapatient dose escalation arm to evaluate tolerability and a biopsy cohort to study pharmacodynamic effects were later added. AT13148 was administered orally three days a week (Mon-Wed-Fri) in 28-day cycles. Pharmacokinetic profiles were assessed using mass spectrometry and pharmacodynamic studies included quantifying p-GSK3β levels in platelet-rich plasma (PRP) and p-cofilin and p-MLC2 levels in tumor biopsies. RESULTS: Fifty-one patients were treated on study. The safety of 5-300 mg of AT13148 was studied. Further, the doses of 120-180-240 mg were studied in an intrapatient dose escalation cohort. The dose-limiting toxicities included hypotension (300 mg), pneumonitis, and elevated liver enzymes (240 mg), and skin rash (180 mg). The most common side effects were fatigue, nausea, headaches, and hypotension. On the basis of tolerability, 180 mg was considered the maximally tolerated dose. At 180 mg, mean C max and AUC were 400 nmol/L and 13,000 nmol/L/hour, respectively. At 180 mg, ≥50% reduction of p-cofilin was observed in 3 of 8 posttreatment biopsies. CONCLUSIONS: AT13148 was the first dual potent ROCK-AKT inhibitor to be investigated for the treatment of solid tumors. The narrow therapeutic index and the pharmacokinetic profile led to recommend not developing this compound further. There are significant lessons learned in designing and testing agents that simultaneously inhibit multiple kinases including AGC kinases in cancer..
Rodriguez-Hernandez, I.
Maiques, O.
Kohlhammer, L.
Cantelli, G.
Perdrix-Rosell, A.
Monger, J.
Fanshawe, B.
Bridgeman, V.L.
Karagiannis, S.N.
Penin, R.M.
Marcolval, J.
Marti, R.M.
Matias-Guiu, X.
Fruhwirth, G.O.
Orgaz, J.L.
Malanchi, I.
Sanz-Moreno, V.
(2020). WNT11-FZD7-DAAM1 signalling supports tumour initiating abilities and melanoma amoeboid invasion. Nat commun,
Vol.11
(1),
p. 5315.
show abstract
Melanoma is a highly aggressive tumour that can metastasize very early in disease progression. Notably, melanoma can disseminate using amoeboid invasive strategies. We show here that high Myosin II activity, high levels of ki-67 and high tumour-initiating abilities are characteristic of invasive amoeboid melanoma cells. Mechanistically, we find that WNT11-FZD7-DAAM1 activates Rho-ROCK1/2-Myosin II and plays a crucial role in regulating tumour-initiating potential, local invasion and distant metastasis formation. Importantly, amoeboid melanoma cells express both proliferative and invasive gene signatures. As such, invasive fronts of human and mouse melanomas are enriched in amoeboid cells that are also ki-67 positive. This pattern is further enhanced in metastatic lesions. We propose eradication of amoeboid melanoma cells after surgical removal as a therapeutic strategy..
Orgaz, J.L.
Sanz-Moreno, V.
(2020). What does not kill you makes you stronger: surviving anti-cancer therapies by cytoskeletal remodeling and Myosin II reactivation. Mol cell oncol,
Vol.7
(3),
p. 1735911.
show abstract
Myosin II and its regulator Rho-associated coiled-coil containing protein kinase (ROCK) are essential for cell invasion and metastatic dissemination. Our recent findings show that this molecular machinery is also involved in drug resistance in melanoma by playing a dual role: protection of tumor cells from reactive oxygen species (ROS) and DNA damage (intrinsic), and co-option of myeloid and lymphoid populations to establish immunosuppression (extrinsic)..
López-Luque, J.
Bertran, E.
Crosas-Molist, E.
Maiques, O.
Malfettone, A.
Caja, L.
Serrano, T.
Ramos, E.
Sanz-Moreno, V.
Fabregat, I.
(2019). Downregulation of Epidermal Growth Factor Receptor in hepatocellular carcinoma facilitates Transforming Growth Factor-β-induced epithelial to amoeboid transition. Cancer lett,
Vol.464,
pp. 15-24.
show abstract
full text
The Epidermal Growth Factor Receptor (EGFR) and the Transforming Growth Factor-beta (TGF-β) are key regulators of hepatocarcinogenesis. Targeting EGFR was proposed as a promising therapy; however, poor success was obtained in human hepatocellular carcinoma (HCC) clinical trials. Here, we describe how EGFR is frequently downregulated in HCC patients while TGF-β is upregulated. Using 2D/3D cellular models, we show that after EGFR loss, TGF-β is more efficient in its pro-migratory and invasive effects, inducing epithelial to amoeboid transition. EGFR knock-down promotes loss of cell-cell and cell-to-matrix adhesion, favouring TGF-β-induced actomyosin contractility and acquisition of an amoeboid migratory phenotype. Moreover, TGF-β upregulates RHOC and CDC42 after EGFR silencing, promoting Myosin II in amoeboid cells. Importantly, low EGFR combined with high TGFB1 or RHOC/CDC42 levels confer poor patient prognosis. In conclusion, this work reveals a new tumour suppressor function for EGFR counteracting TGF-β-mediated epithelial to amoeboid transitions in HCC, supporting a rational for targeting the TGF-β pathway in patients with low EGFR expression. Our work also highlights the relevance of epithelial to amoeboid transition in human tumours and the need to better target this process in the clinic..
Georgouli, M.
Herraiz, C.
Crosas-Molist, E.
Fanshawe, B.
Maiques, O.
Perdrix, A.
Pandya, P.
Rodriguez-Hernandez, I.
Ilieva, K.M.
Cantelli, G.
Karagiannis, P.
Mele, S.
Lam, H.
Josephs, D.H.
Matias-Guiu, X.
Marti, R.M.
Nestle, F.O.
Orgaz, J.L.
Malanchi, I.
Fruhwirth, G.O.
Karagiannis, S.N.
Sanz-Moreno, V.
(2019). Regional Activation of Myosin II in Cancer Cells Drives Tumor Progression via a Secretory Cross-Talk with the Immune Microenvironment. Cell,
Vol.176
(4),
pp. 757-774.e23.
show abstract
ROCK-Myosin II drives fast rounded-amoeboid migration in cancer cells during metastatic dissemination. Analysis of human melanoma biopsies revealed that amoeboid melanoma cells with high Myosin II activity are predominant in the invasive fronts of primary tumors in proximity to CD206+CD163+ tumor-associated macrophages and vessels. Proteomic analysis shows that ROCK-Myosin II activity in amoeboid cancer cells controls an immunomodulatory secretome, enabling the recruitment of monocytes and their differentiation into tumor-promoting macrophages. Both amoeboid cancer cells and their associated macrophages support an abnormal vasculature, which ultimately facilitates tumor progression. Mechanistically, amoeboid cancer cells perpetuate their behavior via ROCK-Myosin II-driven IL-1α secretion and NF-κB activation. Using an array of tumor models, we show that high Myosin II activity in tumor cells reprograms the innate immune microenvironment to support tumor growth. We describe an unexpected role for Myosin II dynamics in cancer cells controlling myeloid function via secreted factors..
Foxall, E.
Staszowska, A.
Hirvonen, L.M.
Georgouli, M.
Ciccioli, M.
Rimmer, A.
Williams, L.
Calle, Y.
Sanz-Moreno, V.
Cox, S.
Jones, G.E.
Wells, C.M.
(2019). PAK4 Kinase Activity Plays a Crucial Role in the Podosome Ring of Myeloid Cells. Cell rep,
Vol.29
(11),
pp. 3385-3393.e6.
show abstract
p21-Activated kinase 4 (PAK4), a serine/threonine kinase, is purported to localize to podosomes: transient adhesive structures that degrade the extracellular matrix to facilitate rapid myeloid cell migration. We find that treatment of transforming growth factor β (TGF-β)-differentiated monocytic (THP-1) cells with a PAK4-targeted inhibitor significantly reduces podosome formation and induces the formation of focal adhesions. This switch in adhesions confers a diminution of matrix degradation and reduced cell migration. Furthermore, reduced PAK4 expression causes a significant reduction in podosome number that cannot be rescued by kinase-dead PAK4, supporting a kinase-dependent role. Concomitant with PAK4 depletion, phosphorylation of Akt is perturbed, whereas a specific phospho-Akt signal is detected within the podosomes. Using superresolution analysis, we find that PAK4 specifically localizes in the podosome ring, nearer to the actin core than other ring proteins. We propose PAK4 kinase activity intersects with the Akt pathway at the podosome ring:core interface to drive regulation of macrophage podosome turnover..
Rosel, D.
Fernandes, M.
Sanz-Moreno, V.
Brábek, J.
(2019). Migrastatics: Redirecting R&D in Solid Cancer Towards Metastasis?. Trends cancer,
Vol.5
(12),
pp. 755-756.
show abstract
The concept of 'migrastatics' allows the development of a new drug class that is neither cytotoxic nor antiproliferative but is solely directed towards inhibition of cancer cell motility. Given that the regulatory pathway is open, and migrastatic candidates have been described, it is the right time to enter a new era of antimetastatic treatment..
Dukinfield, M.
Maniati, E.
Reynolds, L.E.
Aubdool, A.
Baliga, R.S.
D'Amico, G.
Maiques, O.
Wang, J.
Bedi, K.C.
Margulies, K.B.
Sanz-Moreno, V.
Hobbs, A.
Hodivala-Dilke, K.
(2019). Repurposing an anti-cancer agent for the treatment of hypertrophic heart disease. J pathol,
Vol.249
(4),
pp. 523-535.
show abstract
Coronary microvascular dysfunction combined with maladaptive cardiomyocyte morphology and energetics is a major contributor to heart failure advancement. Thus, dually enhancing cardiac angiogenesis and targeting cardiomyocyte function to slow, or reverse, the development of heart failure is a logical step towards improved therapy. We present evidence for the potential to repurpose a former anti-cancer Arg-Gly-Asp (RGD)-mimetic pentapeptide, cilengitide, here used at low doses. Cilengitide targets αvβ3 integrin and this protein is upregulated in human dilated and ischaemic cardiomyopathies. Treatment of mice after abdominal aortic constriction (AAC) surgery with low-dose cilengitide (ldCil) enhances coronary angiogenesis and directly affects cardiomyocyte hypertrophy with an associated reduction in disease severity. At a molecular level, ldCil treatment has a direct effect on cardiac endothelial cell transcriptomic profiles, with a significant enhancement of pro-angiogenic signalling pathways, corroborating the enhanced angiogenic phenotype after ldCil treatment. Moreover, ldCil treatment of Angiotensin II-stimulated AngII-stimulated cardiomyocytes significantly restores transcriptomic profiles similar to those found in normal human heart. The significance of this finding is enhanced by transcriptional similarities between AngII-treated cardiomyocytes and failing human hearts. Taken together, our data provide evidence supporting a possible new strategy for improved heart failure treatment using low-dose RGD-mimetics with relevance to human disease. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland..
Valcarcel-Jimenez, L.
Macchia, A.
Crosas-Molist, E.
Schaub-Clerigué, A.
Camacho, L.
Martín-Martín, N.
Cicogna, P.
Viera-Bardón, C.
Fernández-Ruiz, S.
Rodriguez-Hernandez, I.
Hermanova, I.
Astobiza, I.
Cortazar, A.R.
Corres-Mendizabal, J.
Gomez-Muñoz, A.
Sanz-Moreno, V.
Torrano, V.
Carracedo, A.
(2019). PGC1α Suppresses Prostate Cancer Cell Invasion through ERRα Transcriptional Control. Cancer res,
Vol.79
(24),
pp. 6153-6165.
show abstract
The PPARγ coactivator 1 alpha (PGC1α) is a prostate tumor suppressor that controls the balance between anabolism and catabolism. PGC1A downregulation in prostate cancer is causally associated with the development of metastasis. Here we show that the transcriptional complex formed by PGC1α and estrogen-related receptor 1 alpha (ERRα) controls the aggressive properties of prostate cancer cells. PGC1α expression significantly decreased migration and invasion of various prostate cancer cell lines. This phenotype was consistent with remarkable cytoskeletal remodeling and inhibition of integrin alpha 1 and beta 4 expression, both in vitro and in vivo. CRISPR/Cas9-based deletion of ERRα suppressed PGC1α regulation of cytoskeletal organization and invasiveness. Mechanistically, PGC1α expression decreased MYC levels and activity prior to inhibition of invasiveness. In addition, PGC1α and ERRα associated at the MYC promoter, supporting the inhibitory activity PGC1α. The inverse correlation between PGC1α-ERRα activity and MYC levels was corroborated in multiple prostate cancer datasets. Altogether, these results support that PGC1α-ERRα functions as a tumor-suppressive transcriptional complex through the regulation of metabolic and signaling events. SIGNIFICANCE: These findings describe how downregulation of the prostate tumor suppressor PGC1 drives invasiveness and migration of prostate cancer cells..
Maiques, O.
Georgouli, M.
Sanz-Moreno, V.
(2019). Recent advances in tissue imaging for cancer research. F1000res,
Vol.8.
show abstract
full text
Image analysis in clinical research has evolved at fast pace in the last decade. This review discusses basic concepts ranging from immunohistochemistry to advanced techniques such as multiplex imaging, digital pathology, flow cytometry and intravital microscopy. Tissue imaging ex vivo is still one of the gold-standards in the field due to feasibility. We describe here different protocols and applications of digital analysis providing basic and clinical researchers with an overview on how to analyse tissue images. In vivo imaging is not accessible to researchers; however, it provides invaluable dynamic information easily. Overall, we discuss a plethora of techniques that - when combined - constitute a powerful platform for basic and translational cancer research..
O'Donnell, M.A.
(2018). Victoria Sanz-Moreno: Rho together for cancer research. J cell biol,
Vol.217
(6),
pp. 1885-1886.
show abstract
Sanz-Moreno investigates how the cytoskeleton controls tumor biology..
Peregrin, S.
Jurado-Pueyo, M.
Campos, P.M.
Sanz-Moreno, V.
Ruiz-Gomez, A.
Crespo, P.
Mayor, F.
Murga, C.
(2018). Phosphorylation of p38 by GRK2 at the Docking Groove Unveils a Novel Mechanism for Inactivating p38MAPK. Curr biol,
Vol.28
(15),
p. 2513.
Villaronga, M.Á.
Hermida-Prado, F.
Granda-Díaz, R.
Menéndez, S.T.
Álvarez-Teijeiro, S.
Quer, M.
Vilaseca, I.
Allonca, E.
Garzón-Arango, M.
Sanz-Moreno, V.
Astudillo, A.
Rodrigo, J.P.
García-Pedrero, J.M.
(2018). Immunohistochemical Expression of Cortactin and Focal Adhesion Kinase Predicts Recurrence Risk and Laryngeal Cancer Risk Beyond Histologic Grading. Cancer epidemiol biomarkers prev,
Vol.27
(7),
pp. 805-813.
show abstract
Background: Cortactin (CTTN) and the focal adhesion kinase (FAK) are two major candidate genes to, respectively, drive 11q13- and 8q24-associated aggressive behavior in various cancers. Recent evidence uncovered their clinical relevance in early stages of tumorigenesis as promising biomarkers for cancer risk assessment.Methods: Using a multicenter validation study, CTTN and FAK expression was evaluated by immunohistochemistry (IHC) in a cohort of 109 patients with laryngeal precancerous lesions, and correlated with clinicopathologic parameters and laryngeal cancer risk. The pathophysiologic role of CTTN and FAK was further investigated using functional studies in cellular models.Results: Positive CTTN and FAK expression (scores 2 and 3) was detected in 49 (41%) and 35 (32%) laryngeal dysplasias, respectively. Univariate Cox analysis showed that CTTN and FAK expression but not histologic grading was significantly associated with both recurrence risk and laryngeal cancer risk. Patients carrying strong CTTN- or FAK-expressing lesions (score 3) experienced the highest laryngeal cancer incidence (log-rank P < 0.001). In multivariate stepwise analysis, FAK expression [HR = 13.91; 95% CI, 4.82-40.15; P < 0.001] and alcohol consumption (HR = 2.22; 95% confidence interval, 1.17-4.20; P = 0.014) were significant independent predictors of laryngeal cancer development. Targeting FAK by either RNAi or pharmacologic inhibitors effectively blocked cell growth, colony formation, and invasion into 3D collagen matrices.Conclusions: CTTN and FAK emerge as powerful predictors of laryngeal cancer risk and recurrence risk beyond histologic grading.Impact: Our work supports the applicability of IHC CTTN and FAK as complementary markers for risk stratification in patients with laryngeal precancerous lesions. Cancer Epidemiol Biomarkers Prev; 27(7); 805-13. ©2018 AACR..
Maiques, O.
Barceló, C.
Panosa, A.
Pijuan, J.
Orgaz, J.L.
Rodriguez-Hernandez, I.
Matas-Nadal, C.
Tell, G.
Vilella, R.
Fabra, A.
Puig, S.
Sanz-Moreno, V.
Matias-Guiu, X.
Canti, C.
Herreros, J.
Marti, R.M.
Macià, A.
(2018). T-type calcium channels drive migration/invasion in BRAFV600E melanoma cells through Snail1. Pigment cell melanoma res,
Vol.31
(4),
pp. 484-495.
show abstract
Melanoma is a malignant tumor derived from melanocytes. Once disseminated, it is usually highly resistant to chemotherapy and is associated with poor prognosis. We have recently reported that T-type calcium channels (TTCCs) are overexpressed in melanoma cells and play an important role in melanoma progression. Importantly, TTCC pharmacological blockers reduce proliferation and deregulate autophagy leading to apoptosis. Here, we analyze the role of autophagy during migration/invasion of melanoma cells. TTCC Cav3.1 and LC3-II proteins are highly expressed in BRAFV600E compared with NRAS mutant melanomas, both in cell lines and biopsies. Chloroquine, pharmacological blockade, or gene silencing of TTCCs inhibit the autophagic flux and impair the migration and invasion capabilities, specifically in BRAFV600E melanoma cells. Snail1 plays an important role in motility and invasion of melanoma cells. We show that Snail1 is strongly expressed in BRAFV600E melanoma cells and patient biopsies, and its expression decreases when autophagy is blocked. These results demonstrate a role of Snail1 during BRAFV600E melanoma progression and strongly suggest that targeting macroautophagy and, particularly TTCCs, might be a good therapeutic strategy to inhibit metastasis of the most common melanoma type (BRAFV600E)..
Sanz-Moreno, V.
Balkwill, F.R.
(2018). Mets and NETs: The Awakening Force. Immunity,
Vol.49
(5),
pp. 798-800.
show abstract
In a recent study published in Science, Albrengues et al. (2018) unveil an intriguing mechanism whereby the release of neutrophil extra-cellular traps during chronic lung inflammation awakens dormant malignant cells and contributes to cancer progression..
Muliaditan, T.
Caron, J.
Okesola, M.
Opzoomer, J.W.
Kosti, P.
Georgouli, M.
Gordon, P.
Lall, S.
Kuzeva, D.M.
Pedro, L.
Shields, J.D.
Gillett, C.E.
Diebold, S.S.
Sanz-Moreno, V.
Ng, T.
Hoste, E.
Arnold, J.N.
(2018). Macrophages are exploited from an innate wound healing response to facilitate cancer metastasis. Nat commun,
Vol.9
(1),
p. 2951.
show abstract
Tumour-associated macrophages (TAMs) play an important role in tumour progression, which is facilitated by their ability to respond to environmental cues. Here we report, using murine models of breast cancer, that TAMs expressing fibroblast activation protein alpha (FAP) and haem oxygenase-1 (HO-1), which are also found in human breast cancer, represent a macrophage phenotype similar to that observed during the wound healing response. Importantly, the expression of a wound-like cytokine response within the tumour is clinically associated with poor prognosis in a variety of cancers. We show that co-expression of FAP and HO-1 in macrophages results from an innate early regenerative response driven by IL-6, which both directly regulates HO-1 expression and licenses FAP expression in a skin-like collagen-rich environment. We show that tumours can exploit this response to facilitate transendothelial migration and metastatic spread of the disease, which can be pharmacologically targeted using a clinically relevant HO-1 inhibitor..
Krupina, K.
Kleiss, C.
Awal, S.
Rodriguez-Hernandez, I.
Sanz-Moreno, V.
Sumara, I.
(2018). UBASH3B-mediated silencing of the mitotic checkpoint: Therapeutic perspectives in cancer. Mol cell oncol,
Vol.5
(2),
p. e1271494.
show abstract
Defects in mitosis can lead to aneuploidy, which is a common feature of human cancers. Spindle Assembly Checkpoint (SAC) controls fidelity of chromosome segregation in mitosis to prevent aneuploidy. The ubiquitin receptor protein Ubiquitin Associated and SH3 Domain Containing B (UBASH3B) was recently found to control SAC silencing and faithful chromosome segregation by relocalizing Aurora B kinase to the mitotic microtubules. Accordingly, loss and gain of function of UBASH3B have strong effects on mitotic progression. Downregulation of UBASH3B prevents SAC satisfaction leading to inhibition of chromosome segregation, mitotic arrest, and cell death. In contrast, increased cellular levels of UBASH3B trigger premature and uncontrolled chromosome segregation. Interestingly, elevated levels of UBASH3B were found in aggressive tumors. Therefore, we raised the question whether the oncogenic potential of UBASH3B is linked to its role in chromosome segregation. Here we show that in cancer cells expressing high levels of UBASH3B and SAC proteins, downregulation of UBASH3B, can further potentiate SAC response inducing mitotic arrest and cell death. Moreover, data mining approaches identified a correlation between mRNA levels of UBASH3B and SAC components in a set of primary patient tumors including kidney and liver carcinomas. Thus, inhibition of UBASH3B may offer an attractive therapeutic perspective for cancers..
Josephs, D.H.
Bax, H.J.
Dodev, T.
Georgouli, M.
Nakamura, M.
Pellizzari, G.
Saul, L.
Karagiannis, P.
Cheung, A.
Herraiz, C.
Ilieva, K.M.
Correa, I.
Fittall, M.
Crescioli, S.
Gazinska, P.
Woodman, N.
Mele, S.
Chiaruttini, G.
Gilbert, A.E.
Koers, A.
Bracher, M.
Selkirk, C.
Lentfer, H.
Barton, C.
Lever, E.
Muirhead, G.
Tsoka, S.
Canevari, S.
Figini, M.
Montes, A.
Downes, N.
Dombrowicz, D.
Corrigan, C.J.
Beavil, A.J.
Nestle, F.O.
Jones, P.S.
Gould, H.J.
Sanz-Moreno, V.
Blower, P.J.
Spicer, J.F.
Karagiannis, S.N.
(2017). Anti-Folate Receptor-α IgE but not IgG Recruits Macrophages to Attack Tumors via TNFα/MCP-1 Signaling. Cancer res,
Vol.77
(5),
pp. 1127-1141.
show abstract
IgE antibodies are key mediators of antiparasitic immune responses, but their potential for cancer treatment via antibody-dependent cell-mediated cytotoxicity (ADCC) has been little studied. Recently, tumor antigen-specific IgEs were reported to restrict cancer cell growth by engaging high-affinity Fc receptors on monocytes and macrophages; however, the underlying therapeutic mechanisms were undefined and in vivo proof of concept was limited. Here, an immunocompetent rat model was designed to recapitulate the human IgE-Fcε receptor system for cancer studies. We also generated rat IgE and IgG mAbs specific for the folate receptor (FRα), which is expressed widely on human ovarian tumors, along with a syngeneic rat tumor model expressing human FRα. Compared with IgG, anti-FRα IgE reduced lung metastases. This effect was associated with increased intratumoral infiltration by TNFα+ and CD80+ macrophages plus elevated TNFα and the macrophage chemoattractant MCP-1 in lung bronchoalveolar lavage fluid. Increased levels of TNFα and MCP-1 correlated with IgE-mediated tumor cytotoxicity by human monocytes and with longer patient survival in clinical specimens of ovarian cancer. Monocytes responded to IgE but not IgG exposure by upregulating TNFα, which in turn induced MCP-1 production by monocytes and tumor cells to promote a monocyte chemotactic response. Conversely, blocking TNFα receptor signaling abrogated induction of MCP-1, implicating it in the antitumor effects of IgE. Overall, these findings show how antitumor IgE reprograms monocytes and macrophages in the tumor microenvironment, encouraging the clinical use of IgE antibody technology to attack cancer beyond the present exclusive reliance on IgG. Cancer Res; 77(5); 1127-41. ©2017 AACR..
Pandya, P.
Orgaz, J.L.
Sanz-Moreno, V.
(2017). Modes of invasion during tumour dissemination. Mol oncol,
Vol.11
(1),
pp. 5-27.
show abstract
Cancer cell migration and invasion underlie metastatic dissemination, one of the major problems in cancer. Tumour cells exhibit a striking variety of invasion strategies. Importantly, cancer cells can switch between invasion modes in order to cope with challenging environments. This ability to switch migratory modes or plasticity highlights the challenges behind antimetastasis therapy design. In this Review, we present current knowledge on different tumour invasion strategies, the determinants controlling plasticity and arising therapeutic opportunities. We propose that targeting master regulators controlling plasticity is needed to hinder tumour dissemination and metastasis..
(2017). Cell scientist to watch – Victoria Sanz-Moreno. Journal of cell science,
Vol.130
(7),
pp. 1199-1200.
show abstract
ABSTRACT
Vicky Sanz-Moreno received her first degree in chemistry and later a master's degree in biochemistry from the University of Oviedo in Spain, followed by a PhD in chemical sciences in the laboratory of Piero Crespo at the University of Cantabria, awarded in 2002. After a short postdoc in the same lab funded by a Lady Tata Memorial Trust Fellowship, she moved to the Institute of Cancer Research in London as a CRUK and Marie Curie Intra-European Fellow to work with Chris Marshall. In 2008, she received the Applied Biosystems and EACR 40th Anniversary Research Award. In 2011, Vicky started her independent research group with a CRUK Career Development Fellowship at King's College London in the Randall Division of Cell and Molecular Biophysics. In 2015, she was shortlisted for the CRUK Communications and Brand Ambassador Prize for communicating science to the public through media work. Vicky has been awarded the 2017 BSCB Women in Cell Biology Early Career Medal. Her group is interested in the actomyosin cytoskeleton and in transcription factors, and how the crosstalk between the two influences invasive and metastatic behaviour in cancer cells..
Cantelli, G.
Crosas-Molist, E.
Georgouli, M.
Sanz-Moreno, V.
(2017). TGFΒ-induced transcription in cancer. Semin cancer biol,
Vol.42,
pp. 60-69.
show abstract
full text
The Transforming Growth Factor-beta (TGFβ) pathway mediates a broad spectrum of cellular processes and is involved in several diseases, including cancer. TGFβ has a dual role in tumours, acting as a tumour suppressor in the early phase of tumorigenesis and as a tumour promoter in more advanced stages. In this review, we discuss the effects of TGFβ-driven transcription on all stages of tumour progression, with special focus on lung cancer. Since some TGFβ target genes are specifically involved in promoting metastasis, we speculate that these genes might be good targets to block tumour progression without compromising the tumour suppressor effects of the TGFβ pathway..
Gandalovičová, A.
Rosel, D.
Fernandes, M.
Veselý, P.
Heneberg, P.
Čermák, V.
Petruželka, L.
Kumar, S.
Sanz-Moreno, V.
Brábek, J.
(2017). Migrastatics-Anti-metastatic and Anti-invasion Drugs: Promises and Challenges. Trends cancer,
Vol.3
(6),
pp. 391-406.
show abstract
In solid cancers, invasion and metastasis account for more than 90% of mortality. However, in the current armory of anticancer therapies, a specific category of anti-invasion and antimetastatic drugs is missing. Here, we coin the term 'migrastatics' for drugs interfering with all modes of cancer cell invasion and metastasis, to distinguish this class from conventional cytostatic drugs, which are mainly directed against cell proliferation. We define actin polymerization and contractility as target mechanisms for migrastatics, and review candidate migrastatic drugs. Critical assessment of these antimetastatic agents is warranted, because they may define new options for the treatment of solid cancers..
Pandya, P.
Orgaz, J.L.
Sanz-Moreno, V.
(2017). Actomyosin contractility and collective migration: may the force be with you. Curr opin cell biol,
Vol.48,
pp. 87-96.
show abstract
full text
Collective cell migration is essential during physiological processes such as development or wound healing and in pathological conditions such as cancer dissemination. Cells migrating within multicellular tissues experiment different forces which play an intricate role during tissue formation, development and maintenance. How cells are able to respond to these forces depends largely on how they interact with the extracellular matrix. In this review, we focus on mechanics and mechanotransduction in collective migration. In particular, we discuss current knowledge on how cells integrate mechanical signals during collective migration and we highlight actomyosin contractility as a central hub coordinating mechanosensing and mechanotransduction responses..
Talamillo, A.
Grande, L.
Ruiz-Ontañon, P.
Velasquez, C.
Mollinedo, P.
Torices, S.
Sanchez-Gomez, P.
Aznar, A.
Esparis-Ogando, A.
Lopez-Lopez, C.
Lafita, C.
Berciano, M.T.
Montero, J.A.
Vazquez-Barquero, A.
Segura, V.
Villagra, N.T.
Pandiella, A.
Lafarga, M.
Leon, J.
Martinez-Climent, J.A.
Sanz-Moreno, V.
Fernandez-Luna, J.L.
(2017). ODZ1 allows glioblastoma to sustain invasiveness through a Myc-dependent transcriptional upregulation of RhoA. Oncogene,
Vol.36
(12),
pp. 1733-1744.
show abstract
Long-term survival remains low for most patients with glioblastoma (GBM), which reveals the need for markers of disease outcome and novel therapeutic targets. We describe that ODZ1 (also known as TENM1), a type II transmembrane protein involved in fetal brain development, plays a crucial role in the invasion of GBM cells. Differentiation of glioblastoma stem-like cells drives the nuclear translocation of an intracellular fragment of ODZ1 through proteolytic cleavage by signal peptide peptidase-like 2a. The intracellular fragment of ODZ1 promotes cytoskeletal remodelling of GBM cells and invasion of the surrounding environment both in vitro and in vivo. Absence of ODZ1 by gene deletion or downregulation of ODZ1 by small interfering RNAs drastically reduces the invasive capacity of GBM cells. This activity is mediated by an ODZ1-triggered transcriptional pathway, through the E-box binding Myc protein, that promotes the expression and activation of Ras homolog family member A (RhoA) and subsequent activation of Rho-associated, coiled-coil containing protein kinase (ROCK). Overexpression of ODZ1 in GBM cells reduced survival of xenografted mice. Consistently, analysis of 122 GBM tumour samples revealed that the number of ODZ1-positive cells inversely correlated with overall and progression-free survival. Our findings establish a novel marker of invading GBM cells and consequently a potential marker of disease progression and a therapeutic target in GBM..
Crosas-Molist, E.
Bertran, E.
Rodriguez-Hernandez, I.
Herraiz, C.
Cantelli, G.
Fabra, À.
Sanz-Moreno, V.
Fabregat, I.
(2017). The NADPH oxidase NOX4 represses epithelial to amoeboid transition and efficient tumour dissemination. Oncogene,
Vol.36
(21),
pp. 3002-3014.
show abstract
Epithelial to mesenchymal transition is a common event during tumour dissemination. However, direct epithelial to amoeboid transition has not been characterized to date. Here we provide evidence that cells from hepatocellular carcinoma (HCC), a highly metastatic cancer, undergo epithelial to amoeboid transition in physiological environments, such as organoids or three-dimensional complex matrices. Furthermore, the NADPH oxidase NOX4 inhibits this transition and therefore suppresses efficient amoeboid bleb-based invasion. Moreover, NOX4 expression is associated with E-cadherin levels and inversely correlated with invasive features. NOX4 is necessary to maintain parenchymal structures, increase cell-cell and cell-to-matrix adhesion, and impair actomyosin contractility and amoeboid invasion. Importantly, NOX4 gene deletions are frequent in HCC patients, correlating with higher tumour grade. Contrary to that observed in mesenchymal cell types, here NOX4 suppresses Rho and Cdc42 GTPase expression and downstream actomyosin contractility. In HCC patients, NOX4 expression inversely correlates with RhoC and Cdc42 levels. Moreover, low expression of NOX4 combined with high expression of either RhoC or Cdc42 is associated with worse prognosis. Therefore, loss of NOX4 increases actomyosin levels and favours an epithelial to amoeboid transition contributing to tumour aggressiveness..
Herraiz, C.
Calvo, F.
Pandya, P.
Cantelli, G.
Rodriguez-Hernandez, I.
Orgaz, J.L.
Kang, N.
Chu, T.
Sahai, E.
Sanz-Moreno, V.
(2016). Reactivation of p53 by a Cytoskeletal Sensor to Control the Balance Between DNA Damage and Tumor Dissemination. J natl cancer inst,
Vol.108
(1).
show abstract
full text
BACKGROUND: Abnormal cell migration and invasion underlie metastasis, and actomyosin contractility is a key regulator of tumor invasion. The links between cancer migratory behavior and DNA damage are poorly understood. METHODS: Using 3D collagen systems to recapitulate melanoma extracellular matrix, we analyzed the relationship between the actomyosin cytoskeleton of migrating cells and DNA damage. We used multiple melanoma cell lines and microarray analysis to study changes in gene expression and in vivo intravital imaging (n = 7 mice per condition) to understand how DNA damage impacts invasive behavior. We used Protein Tissue Microarrays (n = 164 melanomas) and patient databases (n = 354 melanoma samples) to investigate the associations between markers of DNA damage and actomyosin cytoskeletal features. Data were analyzed with Student's and multiple t tests, Mann-Whitney's test, one-way analysis of variance, and Pearson correlation. All statistical tests were two-sided. RESULTS: Melanoma cells with low levels of Rho-ROCK-driven actomyosin are subjected to oxidative stress-dependent DNA damage and ATM-mediated p53 protein stabilization. This results in a specific transcriptional signature enriched in DNA damage/oxidative stress responsive genes, including Tumor Protein p53 Inducible Protein 3 (TP53I3 or PIG3). PIG3, which functions in DNA damage repair, uses an unexpected catalytic mechanism to suppress Rho-ROCK activity and impair tumor invasion in vivo. This regulation was suppressed by antioxidants. Furthermore, PIG3 levels decreased while ROCK1/2 levels increased in human metastatic melanomas (ROCK1 vs PIG3; r = -0.2261, P < .0001; ROCK2 vs PIG3: r = -0.1381, P = .0093). CONCLUSIONS: The results suggest using Rho-kinase inhibitors to reactivate the p53-PIG3 axis as a novel therapeutic strategy; we suggest that the use of antioxidants in melanoma should be very carefully evaluated..
Herraiz, C.
Crosas-Molist, E.
Sanz-Moreno, V.
(2016). Reactive oxygen species and tumor dissemination: Allies no longer. Mol cell oncol,
Vol.3
(2),
p. e1127313.
show abstract
For decades, reactive oxygen species (ROS) linked to oxidative stress have been suggested to promote carcinogenesis. However, we and others have demonstrated a protective role for ROS in metastatic dissemination. These recent studies partly explain the large failure observed in clinical trials using antioxidants for cancer prevention..
Torrano, V.
Valcarcel-Jimenez, L.
Cortazar, A.R.
Liu, X.
Urosevic, J.
Castillo-Martin, M.
Fernández-Ruiz, S.
Morciano, G.
Caro-Maldonado, A.
Guiu, M.
Zúñiga-García, P.
Graupera, M.
Bellmunt, A.
Pandya, P.
Lorente, M.
Martín-Martín, N.
Sutherland, J.D.
Sanchez-Mosquera, P.
Bozal-Basterra, L.
Zabala-Letona, A.
Arruabarrena-Aristorena, A.
Berenguer, A.
Embade, N.
Ugalde-Olano, A.
Lacasa-Viscasillas, I.
Loizaga-Iriarte, A.
Unda-Urzaiz, M.
Schultz, N.
Aransay, A.M.
Sanz-Moreno, V.
Barrio, R.
Velasco, G.
Pinton, P.
Cordon-Cardo, C.
Locasale, J.W.
Gomis, R.R.
Carracedo, A.
(2016). The metabolic co-regulator PGC1α suppresses prostate cancer metastasis. Nat cell biol,
Vol.18
(6),
pp. 645-656.
show abstract
Cellular transformation and cancer progression is accompanied by changes in the metabolic landscape. Master co-regulators of metabolism orchestrate the modulation of multiple metabolic pathways through transcriptional programs, and hence constitute a probabilistically parsimonious mechanism for general metabolic rewiring. Here we show that the transcriptional co-activator peroxisome proliferator-activated receptor gamma co-activator 1α (PGC1α) suppresses prostate cancer progression and metastasis. A metabolic co-regulator data mining analysis unveiled that PGC1α is downregulated in prostate cancer and associated with disease progression. Using genetically engineered mouse models and xenografts, we demonstrated that PGC1α opposes prostate cancer progression and metastasis. Mechanistically, the use of integrative metabolomics and transcriptomics revealed that PGC1α activates an oestrogen-related receptor alpha (ERRα)-dependent transcriptional program to elicit a catabolic state and metastasis suppression. Importantly, a signature based on the PGC1α-ERRα pathway exhibited prognostic potential in prostate cancer, thus uncovering the relevance of monitoring and manipulating this pathway for prostate cancer stratification and treatment..
Saul, L.
Ilieva, K.M.
Bax, H.J.
Karagiannis, P.
Correa, I.
Rodriguez-Hernandez, I.
Josephs, D.H.
Tosi, I.
Egbuniwe, I.U.
Lombardi, S.
Crescioli, S.
Hobbs, C.
Villanova, F.
Cheung, A.
Geh, J.L.
Healy, C.
Harries, M.
Sanz-Moreno, V.
Fear, D.J.
Spicer, J.F.
Lacy, K.E.
Nestle, F.O.
Karagiannis, S.N.
(2016). IgG subclass switching and clonal expansion in cutaneous melanoma and normal skin. Sci rep,
Vol.6,
p. 29736.
show abstract
B cells participate in immune surveillance in human circulation and tissues, including tumors such as melanoma. By contrast, the role of humoral responses in cutaneous immunity is underappreciated. We report circulating skin-homing CD22+CLA+B cells in healthy volunteers and melanoma patients (n = 73) and CD22+ cells in melanoma and normal skin samples (n = 189). Normal and malignant skin featured mature IgG and CD22 mRNA, alongside mRNA for the transiently-expressed enzyme Activation-induced cytidine Deaminase (AID). Gene expression analyses of publically-available data (n = 234 GEO, n = 384 TCGA) confirmed heightened humoral responses (CD20, CD22, AID) in melanoma. Analyses of 51 melanoma-associated and 29 normal skin-derived IgG sequence repertoires revealed lower IgG1/IgGtotal representation compared with antibodies from circulating B cells. Consistent with AID, comparable somatic hypermutation frequencies and class-switching indicated affinity-matured antibodies in normal and malignant skin. A melanoma-associated antibody subset featured shorter complementarity-determining (CDR3) regions relative to those from circulating B cells. Clonal amplification in melanoma-associated antibodies and homology modeling indicated differential potential antigen recognition profiles between normal skin and melanoma sequences, suggesting distinct antibody repertoires. Evidence for IgG-expressing B cells, class switching and antibody maturation in normal and malignant skin and clonally-expanded antibodies in melanoma, support the involvement of mature B cells in cutaneous immunity..
Rodriguez-Hernandez, I.
Cantelli, G.
Bruce, F.
Sanz-Moreno, V.
(2016). Rho, ROCK and actomyosin contractility in metastasis as drug targets. F1000res,
Vol.5.
show abstract
Metastasis is the spread of cancer cells around the body and the cause of the majority of cancer deaths. Metastasis is a very complex process in which cancer cells need to dramatically modify their cytoskeleton and cope with different environments to successfully colonize a secondary organ. In this review, we discuss recent findings pointing at Rho-ROCK or actomyosin force (or both) as major drivers of many of the steps required for metastatic success. We propose that these are important drug targets that need to be considered in the clinic to palliate metastatic disease..
Masuzzo, P.
Martens, L.
2014 Cell Migration Workshop Participants,
Ampe, C.
Anderson, K.I.
Barry, J.
De Wever, O.
Debeir, O.
Decaestecker, C.
Dolznig, H.
Friedl, P.
Gaggioli, C.
Geiger, B.
Goldberg, I.G.
Horn, E.
Horwitz, R.
Kam, Z.
Le Dévédec, S.E.
Vignjevic, D.M.
Moore, J.
Olivo-Marin, J.-.
Sahai, E.
Sansone, S.A.
Sanz-Moreno, V.
Strömblad, S.
Swedlow, J.
Textor, J.
Van Troys, M.
Zantl, R.
(2015). An open data ecosystem for cell migration research. Trends cell biol,
Vol.25
(2),
pp. 55-58.
show abstract
Cell migration research has recently become both a high content and a high throughput field thanks to technological, computational, and methodological advances. Simultaneously, however, urgent bioinformatics needs regarding data management, standardization, and dissemination have emerged. To address these concerns, we propose to establish an open data ecosystem for cell migration research..
Albrengues, J.
Bertero, T.
Grasset, E.
Bonan, S.
Maiel, M.
Bourget, I.
Philippe, C.
Herraiz Serrano, C.
Benamar, S.
Croce, O.
Sanz-Moreno, V.
Meneguzzi, G.
Feral, C.C.
Cristofari, G.
Gaggioli, C.
(2015). Epigenetic switch drives the conversion of fibroblasts into proinvasive cancer-associated fibroblasts. Nat commun,
Vol.6,
p. 10204.
show abstract
Carcinoma-associated fibroblasts (CAF) mediate the onset of a proinvasive tumour microenvironment. The proinflammatory cytokine LIF reprograms fibroblasts into a proinvasive phenotype, which promotes extracellular matrix remodelling and collective invasion of cancer cells. Here we unveil that exposure to LIF initiates an epigenetic switch leading to the constitutive activation of JAK1/STAT3 signalling, which results in sustained proinvasive activity of CAF. Mechanistically, p300-histone acetyltransferase acetylates STAT3, which, in turn, upregulates and activates the DNMT3b DNA methyltransferase. DNMT3b methylates CpG sites of the SHP-1 phosphatase promoter, which abrogates SHP-1 expression, and results in constitutive phosphorylation of JAK1. Sustained JAK1/STAT3 signalling is maintained by DNA methyltransferase DNMT1. Consistently, in human lung and head and neck carcinomas, STAT3 acetylation and phosphorylation are inversely correlated with SHP-1 expression. Combined inhibition of DNMT activities and JAK signalling, in vitro and in vivo, results in long-term reversion of CAF-associated proinvasive activity and restoration of the wild-type fibroblast phenotype..
Cantelli, G.
Orgaz, J.L.
Rodriguez-Hernandez, I.
Karagiannis, P.
Maiques, O.
Matias-Guiu, X.
Nestle, F.O.
Marti, R.M.
Karagiannis, S.N.
Sanz-Moreno, V.
(2015). TGF-β-Induced Transcription Sustains Amoeboid Melanoma Migration and Dissemination. Curr biol,
Vol.25
(22),
pp. 2899-2914.
show abstract
Cell migration underlies metastatic dissemination of cancer cells, and fast "amoeboid" migration in the invasive fronts of tumors is controlled by high levels of actomyosin contractility. How amoeboid migration is regulated by extracellular signals and sustained over time by transcriptional changes is not fully understood. Transforming growth factor β (TGF-β) is well known to promote epithelial-to-mesenchymal transition (EMT) and contribute to metastasis, but melanocytes are neural crest derivatives that have undergone EMT during embryonic development. Surprisingly, we find that in melanoma, TGF-β promotes amoeboid features such as cell rounding, membrane blebbing, high levels of contractility, and increased invasion. Using genome-wide transcriptomics, we find that amoeboid melanoma cells are enriched in a TGF-β-driven signature. We observe that downstream of TGF-β, SMAD2 and its adaptor CITED1 control amoeboid behavior by regulating the expression of key genes that activate contractile forces. Moreover, CITED1 is highly upregulated during melanoma progression, and its high expression is associated with poor prognosis. CITED1 is coupled to a contractile-rounded, amoeboid phenotype in a panel of 16 melanoma cell lines, in mouse melanoma xenografts, and in 47 human melanoma patients. Its expression is also enriched in the invasive fronts of lesions. Functionally, we show how the TGF-β-SMAD2-CITED1 axis promotes different steps associated with progression: melanoma detachment from keratinocytes, 2D and 3D migration, attachment to endothelial cells, and in vivo lung metastatic initial colonization and outgrowth. We propose a novel mechanism by which TGF-β-induced transcription sustains actomyosin force in melanoma cells and thereby promotes melanoma progression independently of EMT..
Orgaz, J.L.
Pandya, P.
Dalmeida, R.
Karagiannis, P.
Sanchez-Laorden, B.
Viros, A.
Albrengues, J.
Nestle, F.O.
Ridley, A.J.
Gaggioli, C.
Marais, R.
Karagiannis, S.N.
Sanz-Moreno, V.
(2014). Diverse matrix metalloproteinase functions regulate cancer amoeboid migration. Nat commun,
Vol.5,
p. 4255.
show abstract
Rounded-amoeboid cancer cells use actomyosin contractility driven by Rho-ROCK and JAK-STAT3 to migrate efficiently. It has been suggested that rounded-amoeboid cancer cells do not require matrix metalloproteinases (MMPs) to invade. Here we compare MMP levels in rounded-amoeboid and elongated-mesenchymal melanoma cells. Surprisingly, we find that rounded-amoeboid melanoma cells secrete higher levels of several MMPs, including collagenase MMP-13 and gelatinase MMP-9. As a result, rounded-amoeboid melanoma cells degrade collagen I more efficiently than elongated-mesenchymal cells. Furthermore, using a non-catalytic mechanism, MMP-9 promotes rounded-amoeboid 3D migration through regulation of actomyosin contractility via CD44 receptor. MMP-9 is upregulated in a panel of rounded-amoeboid compared with elongated-mesenchymal melanoma cell lines and its levels are controlled by ROCK-JAK-STAT3 signalling. MMP-9 expression increases during melanoma progression and it is particularly prominent in the invasive fronts of lesions, correlating with cell roundness. Therefore, rounded-amoeboid cells use both catalytic and non-catalytic activities of MMPs for invasion..
Orgaz, J.L.
Herraiz, C.
Sanz-Moreno, V.
(2014). Rho GTPases modulate malignant transformation of tumor cells. Small gtpases,
Vol.5,
p. e29019.
show abstract
Rho GTPases are involved in the acquisition of all the hallmarks of cancer, which comprise 6 biological capabilities acquired during the development of human tumors. The hallmarks include proliferative signaling, evading growth suppressors, resisting cell death, enabling replicative immortality, inducing angiogenesis, and activating invasion and metastasis programs, as defined by Hanahan and Weinberg. (1) Controlling these hallmarks are genome instability and inflammation. Emerging hallmarks are reprogramming of energy metabolism and evading immune destruction. To give a different view to the readers, we will not be focusing on invasion, metastasis, or cytoskeletal remodeling, but we will review here how Rho GTPases contribute to other hallmarks of cancer with a special emphasis on malignant transformation..
Orgaz, J.L.
Sanz-Moreno, V.
(2013). Emerging molecular targets in melanoma invasion and metastasis. Pigment cell melanoma res,
Vol.26
(1),
pp. 39-57.
show abstract
Metastatic cutaneous melanoma accounts for the majority of skin cancer deaths due to its aggressiveness and high resistance to current therapies. To efficiently metastasize, invasive melanoma cells need to change their cytoskeletal organization and alter contacts with the extracellular matrix and the surrounding stromal cells. Melanoma cells can use different migratory strategies depending on varying environments to exit the primary tumour mass and invade surrounding and later distant tissues. In this review, we have focused on tumour cell plasticity or the interconvertibility that melanoma cells have as one of the factors that contribute to melanoma metastasis. This has been an area of very intense research in the last 5 yr yielding a vast number of findings. We have therefore reviewed all the possible clinical opportunities that this new knowledge offers to both stratify and treat cutaneous malignant melanoma patients..
Ruiz-Ontañon, P.
Orgaz, J.L.
Aldaz, B.
Elosegui-Artola, A.
Martino, J.
Berciano, M.T.
Montero, J.A.
Grande, L.
Nogueira, L.
Diaz-Moralli, S.
Esparís-Ogando, A.
Vazquez-Barquero, A.
Lafarga, M.
Pandiella, A.
Cascante, M.
Segura, V.
Martinez-Climent, J.A.
Sanz-Moreno, V.
Fernandez-Luna, J.L.
(2013). Cellular plasticity confers migratory and invasive advantages to a population of glioblastoma-initiating cells that infiltrate peritumoral tissue. Stem cells,
Vol.31
(6),
pp. 1075-1085.
show abstract
Glioblastoma (GBM) is associated with infiltration of peritumoral (PT) parenchyma by isolated tumor cells that leads to tumor regrowth. Recently, GBM stem-like or initiating cells (GICs) have been identified in the PT area, but whether these GICs have enhanced migratory and invasive capabilities compared with GICs from the tumor mass (TM) is presently unknown. We isolated GICs from the infiltrated PT tissue and the TM of three patients and found that PT cells have an advantage over TM cells in two-dimensional and three-dimensional migration and invasion assays. Interestingly, PT cells display a high plasticity in protrusion formation and cell shape and their migration is insensitive to substrate stiffness, which represent advantages to infiltrate microenvironments of different rigidity. Furthermore, mouse and chicken embryo xenografts revealed that only PT cells showed a dispersed distribution pattern, closely associated to blood vessels. Consistent with cellular plasticity, simultaneous Rac and RhoA activation are required for the enhanced invasive capacity of PT cells. Moreover, Rho GTPase signaling modulators αVβ3 and p27 play key roles in GIC invasiveness. Of note, p27 is upregulated in TM cells and inhibits RhoA activity. Gene silencing of p27 increased the invasive capacity of TM GICs. Additionally, β3 integrin is upregulated in PT cells. Blockade of dimeric integrin αVβ3, a Rac activator, reduced the invasive capacity of PT GICs in vitro and abrogated the spreading of PT cells into chicken embryos. Thus, our results describe the invasive features acquired by a unique subpopulation of GICs that infiltrate neighboring tissue..
Ahn, J.
Sanz-Moreno, V.
Marshall, C.J.
(2012). The metastasis gene NEDD9 product acts through integrin β3 and Src to promote mesenchymal motility and inhibit amoeboid motility. J cell sci,
Vol.125
(Pt 7),
pp. 1814-1826.
show abstract
Neural precursor expressed, developmentally down-regulated 9 (NEDD9), a member of the Cas family of signal transduction molecules, is amplified at the genetic level in melanoma, and elevated expression levels have been shown to correlate with melanoma progression and metastasis. NEDD9 interacts with the guanine nucleotide exchange factor DOCK3 to promote Rac activation and the elongated, mesenchymal-type of tumour cell invasion, but the molecular mechanisms through which NEDD9 promotes melanoma metastasis are not fully understood. We show that signalling through increased NEDD9 levels requires integrin β3 signalling, which leads to elevated phosphorylation of integrin β3. This results in increased Src and FAK but decreased ROCK signalling to drive elongated, mesenchymal-type invasion in environments that contain vitronectin. NEDD9 overexpression does not affect ROCK signalling through activation of RhoA but decreases ROCKII signalling through Src-dependent phosphorylation of a negative regulatory site Tyr722. In NEDD9-overexpressing melanoma cells, inhibition of Src with dasatinib results in a switch from Rac-driven elongated, mesenchymal-type invasion to ROCK-dependent rounded, amoeboid invasion. These findings brings into question whether dasatinib would work as a therapeutic agent to block melanoma invasion and metastasis. On the basis of the in vitro data presented here, a combination treatment of dasatinib and a ROCK inhibitor might be a better alternative in order to inhibit both elongated, mesenchymal-type and rounded, amoeboid motility..
Sanz-Moreno, V.
(2012). Tumour invasion: a new twist on Rac-driven mesenchymal migration. Curr biol,
Vol.22
(11),
pp. R449-R451.
show abstract
Elongated mesenchymal migration of cancer cells is driven by Rac1 activation mediated by the adaptor NEDD9 and the exchange factor DOCK3. A new study reports a role for the transcription factor Twist1 in inducing mesenchymal migration by relieving the suppression of NEDD9 and DOCK3 by the microRNA let-7i..
Crespo, P.
Calvo, F.
Sanz-Moreno, V.
(2011). Ras and Rho GTPases on the move: The RasGRF connection. Bioarchitecture,
Vol.1
(4),
pp. 200-204.
show abstract
Metastasis involves tumor cells moving through tissues and crossing tissue boundaries, which requires cell migration, remodeling of cell-to-cell contacts and interactions with the extracellular matrix. Individual tumor cells move in three-dimensional environments with either a rounded "ameboid" or an elongated "mesenchymal" morphology. These two modes of movement are tightly regulated by Rho family GTPases: elongated movement requires activation of Rac1, whereas rounded movement engages specific Cdc42 and Rho signaling pathways. It has been known for some time that events unfolding downstream of Ras GTPases are also involved in regulating multiple aspects of cell migration and invasion. More recently, RasGRF2-a Ras activator-has been identified as a suppressor of rounded movement, by inhibiting the activation of Cdc42, independently of its capacity to activate Ras. Here, we discuss how Rho and Ras signals can either cooperate or oppose each other in the regulation of cell migration and invasion..
Sanz-Moreno, V.
Gaggioli, C.
Yeo, M.
Albrengues, J.
Wallberg, F.
Viros, A.
Hooper, S.
Mitter, R.
Féral, C.C.
Cook, M.
Larkin, J.
Marais, R.
Meneguzzi, G.
Sahai, E.
Marshall, C.J.
(2011). ROCK and JAK1 signaling cooperate to control actomyosin contractility in tumor cells and stroma. Cancer cell,
Vol.20
(2),
pp. 229-245.
show abstract
Proinflammatory cytokines are frequently observed in the tumor microenvironment, and chronic inflammation is involved in cancer initiation and progression. We show that cytokine signaling through the receptor subunit GP130-IL6ST and the kinase JAK1 generates actomyosin contractility through Rho-kinase dependent signaling. This pathway generates contractile force in stromal fibroblasts to remodel the extracellular matrix to create tracks for collective migration of squamous carcinoma cells and provides the high levels of actomyosin contractility required for migration of individual melanoma cells in the rounded, "amoeboid" mode. Thus, cytokine signaling can generate actomyosin contractility in both stroma and tumor cells. Strikingly, actomyosin contractility itself positively modulates activity of the transcription factor STAT3 downstream of JAK1, demonstrating positive feedback within the signaling network..
Calvo, F.
Sanz-Moreno, V.
Agudo-Ibáñez, L.
Wallberg, F.
Sahai, E.
Marshall, C.J.
Crespo, P.
(2011). RasGRF suppresses Cdc42-mediated tumour cell movement, cytoskeletal dynamics and transformation. Nat cell biol,
Vol.13
(7),
pp. 819-826.
show abstract
Individual tumour cells move in three-dimensional environments with either a rounded or an elongated 'mesenchymal' morphology. These two modes of movement are tightly regulated by Rho family GTPases: elongated movement requires activation of Rac1, whereas rounded/amoeboid movement engages specific Cdc42 and Rho signalling pathways. In siRNA screens targeting the genes encoding guanine nucleotide exchange factors (GEFs), we found that the Ras GEF RasGRF2 regulates conversion between elongated- and rounded-type movement. RasGRF2 suppresses rounded movement by inhibiting the activation of Cdc42 independently of its capacity to activate Ras. RasGRF2 and RasGRF1 directly bind to Cdc42, outcompeting Cdc42 GEFs, thereby preventing Cdc42 activation. By this mechanism, RasGRFs regulate other Cdc42-mediated cellular processes such as the formation of actin spikes, transformation and invasion in vitro and in vivo. These results demonstrate a role for RasGRF GEFs as negative regulators of Cdc42 activation..
Sanz-Moreno, V.
Marshall, C.J.
(2010). The plasticity of cytoskeletal dynamics underlying neoplastic cell migration. Curr opin cell biol,
Vol.22
(5),
pp. 690-696.
show abstract
Due to the use of intra-vital imaging techniques and assays for cell migration into 3D matrices there has recently been much interest in different modes of tumour cell migration. Individually moving tumour cells can move either in an elongated-protrusive manner or in rounded, so-called 'amoeboid' modes. This review summarises ongoing efforts to delineate the cell signalling pathways that underlie these different forms of movement..
Casar, B.
Arozarena, I.
Sanz-Moreno, V.
Pinto, A.
Agudo-Ibáñez, L.
Marais, R.
Lewis, R.E.
Berciano, M.T.
Crespo, P.
(2009). Ras subcellular localization defines extracellular signal-regulated kinase 1 and 2 substrate specificity through distinct utilization of scaffold proteins. Mol cell biol,
Vol.29
(5),
pp. 1338-1353.
show abstract
full text
Subcellular localization influences the nature of Ras/extracellular signal-regulated kinase (ERK) signals by unknown mechanisms. Herein, we demonstrate that the microenvironment from which Ras signals emanate determines which substrates will be preferentially phosphorylated by the activated ERK1/2. We show that the phosphorylation of epidermal growth factor receptor (EGFr) and cytosolic phospholipase A(2) (cPLA(2)) is most prominent when ERK1/2 are activated from lipid rafts, whereas RSK1 is mainly activated by Ras signals from the disordered membrane. We present evidence indicating that the underlying mechanism of this substrate selectivity is governed by the participation of different scaffold proteins that distinctively couple ERK1/2, activated at defined microlocalizations, to specific substrates. As such, we show that for cPLA(2) activation, ERK1/2 activated at lipid rafts interact with KSR1, whereas ERK1/2 activated at the endoplasmic reticulum utilize Sef-1. To phosphorylate the EGFr, ERK1/2 activated at lipid rafts require the participation of IQGAP1. Furthermore, we demonstrate that scaffold usage markedly influences the biological outcome of Ras site-specific signals. These results disclose an unprecedented spatial regulation of ERK1/2 substrate specificity, dictated by the microlocalization from which Ras signals originate and by the selection of specific scaffold proteins..
Sanz-Moreno, V.
Marshall, C.J.
(2009). Rho-GTPase signaling drives melanoma cell plasticity. Cell cycle,
Vol.8
(10),
pp. 1484-1487.
show abstract
To investigate mechanisms that underlie different modes of tumor cell movement we have studied how regulation of the activity of the Rho family GTPases determines the mode of tumor cell movement. Guanine nucleotide exchange factors (GEFs) and GTPase accelerating proteins (GAPs) are key regulators of the activity of small GTPases with GEFs promoting activation to the GTP bound state and GAPs promoting inactivation by stimulating GTP hydrolysis. We identified two important signaling pathways regulating amoeboid and mesenchymal types of motility in melanoma. Here, we discuss our findings in the context of how specificity of Rho signaling is achieved by GEFs and GAPs..
Gadea, G.
Sanz-Moreno, V.
Self, A.
Godi, A.
Marshall, C.J.
(2008). DOCK10-mediated Cdc42 activation is necessary for amoeboid invasion of melanoma cells. Curr biol,
Vol.18
(19),
pp. 1456-1465.
show abstract
BACKGROUND: Tumor cells can move in a three-dimensional (3D) environment in either mesenchymal-type or amoeboid modes. In mesenchymal-type movement, cells have an elongated morphology with Rac-induced protrusions at the leading edge. Amoeboid cells have high levels of actomyosin contractility, and movement is associated with deformation of the cell body through the matrix without proteolysis. Because signaling pathways that control the activation of GTPases for amoeboid movement are poorly understood, we sought to identify regulators of amoeboid movement by screening an siRNA library targeting guanine nucleotide exchange factors (GEFs) for Rho-family GTPases. RESULTS: We identified DOCK10, a Cdc42 GEF, as a key player in amoeboid migration; accordingly, we find that expression of activated Cdc42 induces a mesenchymal-amoeboid transition and increases cell invasion. Silencing DOCK10 expression promotes conversion to mesenchymal migration and is associated with decreased MLC2 phosphorylation and increased Rac1 activation. Consequently, abrogating DOCK10 and Rac1 expression suppresses both amoeboid and mesenchymal migration and results in decreased invasion. We show that the Cdc42 effectors N-WASP and Pak2 are required for the maintenance of the rounded-amoeboid phenotype. Blocking Cdc42 results in loss of mesenchymal morphology, arguing that Cdc42 is also involved in mesenchymal morphology through different activation and effector pathways. CONCLUSIONS: Previous work has identified roles of Rho and Rac signaling in tumor cell movement, and we now elucidate novel roles of Cdc42 signaling in amoeboid and mesenchymal movement and tumor cell invasion..
Sanz-Moreno, V.
Gadea, G.
Ahn, J.
Paterson, H.
Marra, P.
Pinner, S.
Sahai, E.
Marshall, C.J.
(2008). Rac activation and inactivation control plasticity of tumor cell movement. Cell,
Vol.135
(3),
pp. 510-523.
show abstract
Tumor cells exhibit two different modes of individual cell movement. Mesenchymal-type movement is characterized by an elongated cellular morphology and requires extracellular proteolysis. In amoeboid movement, cells have a rounded morphology, are less dependent on proteases, and require high Rho-kinase signaling to drive elevated levels of actomyosin contractility. These two modes of cell movement are interconvertible. We show that mesenchymal-type movement in melanoma cells is driven by activation of the GTPase Rac through a complex containing NEDD9, a recently identified melanoma metastasis gene, and DOCK3, a Rac guanine nucleotide exchange factor. Rac signals through WAVE2 to direct mesenchymal movement and suppress amoeboid movement through decreasing actomyosin contractility. Conversely, in amoeboid movement, Rho-kinase signaling activates a Rac GAP, ARHGAP22, that suppresses mesenchymal movement by inactivating Rac. We demonstrate tight interplay between Rho and Rac in determining different modes of tumor cell movement, revealing how tumor cells switch between different modes of movement..
Casar, B.
Sanz-Moreno, V.
Yazicioglu, M.N.
Rodríguez, J.
Berciano, M.T.
Lafarga, M.
Cobb, M.H.
Crespo, P.
(2007). Mxi2 promotes stimulus-independent ERK nuclear translocation. Embo j,
Vol.26
(3),
pp. 635-646.
show abstract
full text
Spatial regulation of ERK1/2 MAP kinases is an essential yet largely unveiled mechanism for ensuring the fidelity and specificity of their signals. Mxi2 is a p38alpha isoform with the ability to bind ERK1/2. Herein we show that Mxi2 has profound effects on ERK1/2 nucleocytoplasmic distribution, promoting their accumulation in the nucleus. Downregulation of endogenous Mxi2 by RNAi causes a marked reduction of ERK1/2 in the nucleus, accompanied by a pronounced decline in cellular proliferation. We demonstrate that Mxi2 functions in nuclear shuttling of ERK1/2 by enhancing the nuclear accumulation of both phosphorylated and unphosphorylated forms in the absence of stimulation. This process requires the direct interaction of both proteins and a high-affinity binding of Mxi2 to ERK-binding sites in nucleoporins, In this respect, Mxi2 acts antagonistically to PEA15, displacing it from ERK1/2 complexes. These results point to Mxi2 as a key spatial regulator for ERK1/2 and disclose an unprecedented stimulus-independent mechanism for ERK nuclear import..
Matallanas, D.
Sanz-Moreno, V.
Arozarena, I.
Calvo, F.
Agudo-Ibáñez, L.
Santos, E.
Berciano, M.T.
Crespo, P.
(2006). Distinct utilization of effectors and biological outcomes resulting from site-specific Ras activation: Ras functions in lipid rafts and Golgi complex are dispensable for proliferation and transformation. Mol cell biol,
Vol.26
(1),
pp. 100-116.
show abstract
full text
Ras proteins are distributed in different types of plasma membrane microdomains and endomembranes. However, how microlocalization affects the signals generated by Ras and its subsequent biological outputs is largely unknown. We have approached this question by selectively targeting RasV12 to different cellular sublocalizations. We show here that compartmentalization dictates Ras utilization of effectors and the intensity of its signals. Activated Ras can evoke enhanced proliferation and transformation from most of its platforms, with the exception of the Golgi complex. Furthermore, signals that promote survival emanate primarily from the endoplasmic reticulum pool. In addition, we have investigated the need for the different pools of endogenous Ras in the conveyance of upstream mitogenic and transforming signals. Using targeted RasN17 inhibitory mutants and in physiological contexts such as H-Ras/N-Ras double knockout fibroblasts, we demonstrate that Ras functions at lipid rafts and at the Golgi complex are fully dispensable for proliferation and transformation..
Arozarena, I.
Matallanas, D.
Berciano, M.T.
Sanz-Moreno, V.
Calvo, F.
Muñoz, M.T.
Egea, G.
Lafarga, M.
Crespo, P.
(2004). Activation of H-Ras in the endoplasmic reticulum by the RasGRF family guanine nucleotide exchange factors. Mol cell biol,
Vol.24
(4),
pp. 1516-1530.
show abstract
full text
Recent findings indicate that in addition to its location in the peripheral plasma membrane, H-Ras is found in endomembranes like the endoplasmic reticulum and the Golgi complex. In these locations H-Ras is functional and can efficiently engage downstream effectors, but little is known about how its activation is regulated in these environments. Here we show that the RasGRF family exchange factors, both endogenous and ectopically expressed, are present in the endoplasmic reticulum but not in the Golgi complex. With the aid of H-Ras constructs specifically tethered to the plasma membrane, endoplasmic reticulum, and Golgi complex, we demonstrate that RasGRF1 and RasGRF2 can activate plasma membrane and reticular, but not Golgi-associated, H-Ras. We also show that RasGRF DH domain is required for the activation of H-Ras in the endoplasmic reticulum but not in the plasma membrane. Furthermore, we demonstrate that RasGRF mediation favors the activation of reticular H-Ras by lysophosphatidic acid treatment whereas plasma membrane H-Ras is made more responsive to stimulation by ionomycin. Overall, our results provide the initial insights into the regulation of H-Ras activation in the endoplasmic reticulum..
Sanz-Moreno, V.
Casar, B.
Crespo, P.
(2003). p38alpha isoform Mxi2 binds to extracellular signal-regulated kinase 1 and 2 mitogen-activated protein kinase and regulates its nuclear activity by sustaining its phosphorylation levels. Mol cell biol,
Vol.23
(9),
pp. 3079-3090.
show abstract
Mxi2 is a p38alpha splice isoform that is distinctively activated by mitogenic stimuli. Here we show that Mxi2 immunoprecipitates carry a kinase activity that is persistently activated by epidermal growth factor in a fashion regulated by Ras, Raf, and MEK. We demonstrate that this kinase activity can be attributed not to Mxi2 but rather to extracellular signal-regulated kinases 1 and 2 (ERK1/2), which coimmunoprecipitated with Mxi2 both by ectopic expression and in a physiological environment like the kidney. Furthermore, we provide evidence that Mxi2-ERK interaction has profound effects on ERK function, demonstrating that Mxi2 prolongs the duration of the ERK signal by sustaining its phosphorylation levels. Interestingly, we show that the effects of Mxi2 on ERK are restricted to nuclear events. Mxi2 potently up-regulates ERK-mediated activation of the transcription factors Elk1 and HIF1alpha but has no effect on the activity of ERK cytoplasmic substrates RSK2 and cPLA(2), induced by epidermal growth factor or by MEK. Overall, our findings point to Mxi2 as a unique member of the p38 family that may have an unprecedented role in the regulation of the functions of ERK mitogen-activated protein kinases..
Sanz, V.
Arozarena, I.
Crespo, P.
(2000). Distinct carboxy‐termini confer divergent characteristics to the mitogen‐activated protein kinase p38α and its splice isoform Mxi2. Febs letters,
Vol.474
(2-3),
pp. 169-174.
show abstract
The p38 family of mitogen‐activated protein kinases is composed of several isoforms. Mxi2 is a splicing variant of p38α that harbors a unique carboxy‐terminus. Here we show that this sole divergence results in remarkable differences between Mxi2 and p38α. Mxi2 is distinctively activated by stress stimuli and potently activated by mitogens. Mxi2 affinity for bona fide p38 substrates is remarkably diminished and Mxi2 activity is largely unaffected by the phosphatase CL100. Also, Mxi2 sensitivity to inhibition by SB203580 is greatly reduced. Interestingly, we show that the p38 C‐terminus is involved in conferring sensitivity to this compound. Overall, our results point to the p38 carboxy‐terminus as a key determinant of the biochemical properties of this family of kinases..
Arozarena, I.
Aaronson, D.S.
Matallanas, D.
Sanz, V.
Ajenjo, N.
Tenbaum, S.P.
Teramoto, H.
Ighishi, T.
Zabala, J.C.
Gutkind, J.S.
Crespo, P.
(2000). The Rho Family GTPase Cdc42 Regulates the Activation of Ras/MAP Kinase by the Exchange Factor Ras-GRF. Journal of biological chemistry,
Vol.275
(34),
pp. 26441-26448.