Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Maria Cristina Rangel is active.

Publication


Featured researches published by Maria Cristina Rangel.


American Journal of Pathology | 2010

Role of Cripto-1 in Stem Cell Maintenance and Malignant Progression

Caterina Bianco; Maria Cristina Rangel; Nadia P. Castro; Tadahiro Nagaoka; Kelly Rollman; Monica Gonzales; David S. Salomon

Cripto-1 is critical for early embryonic development and, together with its ligand Nodal, has been found to be associated with the undifferentiated status of mouse and human embryonic stem cells. Like other embryonic genes, Cripto-1 performs important roles in the formation and progression of several types of human tumors, stimulating cell proliferation, migration, epithelial to mesenchymal transition, and tumor angiogenesis. Several studies have demonstrated that cell fate regulation during embryonic development and cell transformation during oncogenesis share common signaling pathways, suggesting that uncontrolled activation of embryonic signaling pathways might drive cell transformation and tumor progression in adult tissues. Here we review our current understanding of how Cripto-1 controls stem cell biology and how it integrates with other major embryonic signaling pathways. Because many cancers are thought to derive from a subpopulation of cancer stem-like cells, which may re-express embryonic genes, Cripto-1 signaling may drive tumor growth through the generation or expansion of tumor initiating cells bearing stem-like characteristics. Therefore, the Cripto-1/Nodal signaling may represent an attractive target for treatment in cancer, leading to the elimination of undifferentiated stem-like tumor initiating cells.


American Journal of Pathology | 2012

Role of Cripto-1 during epithelial-to-mesenchymal transition in development and cancer.

Maria Cristina Rangel; Hideaki Karasawa; Nadia P. Castro; Tadahiro Nagaoka; David S. Salomon; Caterina Bianco

Epithelial-to-mesenchymal transition (EMT) is a critical multistep process that converts epithelial cells to more motile and invasive mesenchymal cells, contributing to body patterning and morphogenesis during embryonic development. In addition, both epithelial plasticity and increased motility and invasiveness are essential for the branching morphogenesis that occurs during development of the mammary gland and during tumor formation, allowing cancer cells to escape from the primary tumor. Cripto-1, a member of the epidermal growth factor-Cripto-1/FRL-1/Cryptic (EGF/CFC) gene family, together with the transforming growth factor (TGF)-β family ligand Nodal, regulates both cell movement and EMT during embryonic development. During postnatal development, Cripto-1 regulates the branching morphogenesis of the mouse mammary gland and enhances both the invasive and migratory properties of mammary epithelial cells in vitro. Furthermore, transgenic mouse models have shown that Cripto-1 promotes the formation of mammary tumors that display properties of EMT, including the down-regulation of the cell surface adherens junctional protein E-cadherin and the up-regulation of mesenchymal markers, such as vimentin, N-cadherin, and Snail. Interestingly, Cripto-1 is enriched in a subpopulation of embryonal, melanoma, prostate, and pancreatic cancer cells that possess stem-like characteristics. Therefore, Cripto-1 may play a role during developmental EMT, and it may also be involved in the reprogramming of differentiated tumor cells into cancer stem cells through the induction of an EMT program.


Future Oncology | 2010

Cripto-1: an embryonic gene that promotes tumorigenesis

Nadia P. Castro; Maria Cristina Rangel; Tadahiro Nagaoka; David S. Salomon; Caterina Bianco

Several studies have shown that cell fate regulation during embryonic development and oncogenic transformation share common regulatory mechanisms and signaling pathways. Indeed, an embryonic gene member of the EGF-Cripto-1/FRL1/Cryptic family, Cripto-1, has been implicated in embryogenesis and in carcinogenesis. Cripto-1 together with the TGF-beta ligand Nodal is a key regulator of embryonic development and is a marker of undifferentiated human and mouse embryonic stem cells. While Cripto-1 expression is very low in normal adult tissues, Cripto-1 is re-expressed at high levels in several different human tumors, modulating cancer cell proliferation, migration, epithelial-to-mesenchymal transition and stimulating tumor angiogenesis. Therefore, inhibition of Cripto-1 expression using blocking antibodies or antisense expression vectors might be a useful modality not only to target fully differentiated cancer cells but also to target a subpopulation of tumor cells with stem-like characteristics.


Journal of Cell Biology | 2009

Enhancement of Notch receptor maturation and signaling sensitivity by Cripto-1

Kazuhide Watanabe; Tadahiro Nagaoka; Joseph M. Lee; Caterina Bianco; Monica Gonzales; Nadia P. Castro; Maria Cristina Rangel; Kei Sakamoto; Youping Sun; Robert Callahan; David S. Salomon

Cripto-1 associates with Notch1 in the endoplasmic reticulum and Golgi to enhance Notch1 localization to lipid rafts and its maturation.


Seminars in Cancer Biology | 2014

The multifaceted role of the embryonic gene Cripto-1 in cancer, stem cells and epithelial-mesenchymal transition

Malgorzata Klauzinska; Nadia P. Castro; Maria Cristina Rangel; Benjamin T. Spike; Peter C. Gray; Daniel Bertolette; Frank Cuttitta; David S. Salomon

Cripto-1 (CR-1)/Teratocarcinoma-derived growth factor1 (TDGF-1) is a cell surface glycosylphosphatidylinositol (GPI)-linked glycoprotein that can function either in cis (autocrine) or in trans (paracrine). The cell membrane cis form is found in lipid rafts and endosomes while the trans acting form lacking the GPI anchor is soluble. As a member of the epidermal growth factor (EGF)/Cripto-1-FRL-1-Cryptic (CFC) family, CR-1 functions as an obligatory co-receptor for the transforming growth factor-β (TGF-β) family members, Nodal and growth and differentiation factors 1 and 3 (GDF1/3) by activating Alk4/Alk7 signaling pathways that involve Smads 2, 3 and 4. In addition, CR-1 can activate non-Smad-dependent signaling elements such as PI3K, Akt and MAPK. Both of these pathways depend upon the 78kDa glucose regulated protein (GRP78). Finally, CR-1 can facilitate signaling through the canonical Wnt/β-catenin and Notch/Cbf-1 pathways by functioning as a chaperone protein for LRP5/6 and Notch, respectively. CR-1 is essential for early embryonic development and maintains embryonic stem cell pluripotentiality. CR-1 performs an essential role in the etiology and progression of several types of human tumors where it is expressed in a population of cancer stem cells (CSCs) and facilitates epithelial-mesenchymal transition (EMT). In this context, CR-1 can significantly enhance tumor cell migration, invasion and angiogenesis. Collectively, these facts suggest that CR-1 may be an attractive target in the diagnosis, prognosis and therapy of several types of human cancer.


Cellular Signalling | 2013

Cripto-1 enhances the canonical Wnt/β-catenin signaling pathway by binding to LRP5 and LRP6 co-receptors.

Tadahiro Nagaoka; Hideaki Karasawa; Thomas Turbyville; Maria Cristina Rangel; Nadia P. Castro; Monica Gonzales; Alyson Baker; Masaharu Seno; Stephen J. Lockett; Yoshimi Endo Greer; Jeffrey S. Rubin; David S. Salomon; Caterina Bianco

Cripto-1 is implicated in multiple cellular events, including cell proliferation, motility and angiogenesis, through the activation of an intricate network of signaling pathways. A crosstalk between Cripto-1 and the canonical Wnt/β-catenin signaling pathway has been previously described. In fact, Cripto-1 is a downstream target gene of the canonical Wnt/β-catenin signaling pathway in the embryo and in colon cancer cells and T-cell factor (Tcf)/lymphoid enhancer factor binding sites have been identified in the promoter and the first intronic region of the mouse and human Cripto-1 genes. We now demonstrate that Cripto-1 modulates signaling through the canonical Wnt/β-catenin/Tcf pathway by binding to the Wnt co-receptors low-density lipoprotein receptor-related protein (LRP) 5 and LRP6, which facilitates Wnt3a binding to LRP5 and LRP6. Cripto-1 functionally enhances Wnt3a signaling through cytoplasmic stabilization of β-catenin and elevated β-catenin/Tcf transcriptional activation. Conversely, Wnt3a further increases Cripto-1 stimulation of migration, invasion and colony formation in soft agar of HC11 mouse mammary epithelial cells, indicating that Cripto-1 and the canonical Wnt/β-catenin signaling co-operate in regulating motility and in vitro transformation of mammary epithelial cells.


Breast Cancer Research and Treatment | 2016

Developmental signaling pathways regulating mammary stem cells and contributing to the etiology of triple-negative breast cancer

Maria Cristina Rangel; Daniel Bertolette; Nadia P. Castro; Malgorzata Klauzinska; Frank Cuttitta; David S. Salomon

Cancer has been considered as temporal and spatial aberrations of normal development in tissues. Similarities between mammary embryonic development and cell transformation suggest that the underlying processes required for mammary gland development are also those perturbed during various stages of mammary tumorigenesis and breast cancer (BC) development. The master regulators of embryonic development Cripto-1, Notch/CSL, and Wnt/β-catenin play key roles in modulating mammary gland morphogenesis and cell fate specification in the embryo through fetal mammary stem cells (fMaSC) and in the adult organism particularly within the adult mammary stem cells (aMaSC), which determine mammary progenitor cell lineages that generate the basal/myoepithelial and luminal compartments of the adult mammary gland. Together with recognized transcription factors and embryonic stem cell markers, these embryonic regulatory molecules can be inappropriately augmented during tumorigenesis to support the tumor-initiating cell (TIC)/cancer stem cell (CSC) compartment, and the effects of their deregulation may contribute for the etiology of BC, in particular the most aggressive subtype of BC, triple-negative breast cancer (TNBC). This in depth review will present evidence of the involvement of Cripto-1, Notch/CSL, and Wnt/β-catenin in the normal mammary gland morphogenesis and tumorigenesis, from fMaSC/aMaSC regulation to TIC generation and maintenance in TNBC. Specific therapies for treating TNBC by targeting these embryonic pathways in TICs will be further discussed, providing new opportunities to destroy not only the bulk tumor, but also TICs that initiate and promote the metastatic spread and recurrence of this aggressive subtype of BC.


Growth Factors Journal | 2012

An evolving web of signaling networks regulated by Cripto-1

Tadahiro Nagaoka; Hideaki Karasawa; Nadia P. Castro; Maria Cristina Rangel; David S. Salomon; Caterina Bianco

Over the past few decades, our understanding of the embryonic gene Cripto-1 has considerably advanced through biochemical, cell biology, and animal studies. Cripto-1 performs key functions during embryonic development, while it dramatically disappears in adult tissues, except possibly in adult tissue stem cells. Cripto-1 is re-expressed in human tumors promoting cell proliferation, migration, invasion, epithelial to mesenchymal transition, and tumor angiogenesis. This diversity of biological effects is dependent upon interaction of Cripto-1 with an extensive array of signaling molecules. In fact, Cripto-1 modulates signaling of transforming growth factor-β family members, including Nodal, GDF-1/-3, Activin, and TGF-β1, activates c-src/MAPK/Protein Kinase B (AKT) pathway in a Glypican-1 and GRP78-dependent manner, and cross-talks with erbB4, Wnt/β-catenin, Notch, Caveolin-1, and Apelin/putative receptor protein related to Angiotensin-type I receptor (APJ) pathways. This article provides an updated survey of the various signaling pathways modulated by Cripto-1 with a focus on mechanistic insights in our understanding of the biological function of Cripto-1 in eukaryotic cells.


Journal of Cellular Physiology | 2013

Regulation of human Cripto‐1 expression by nuclear receptors and DNA promoter methylation in human embryonal and breast cancer cells

Caterina Bianco; Nadia P. Castro; Christina Baraty; Kelly Rollman; Natalie Held; Maria Cristina Rangel; Hideaki Karasawa; Monica Gonzales; Luigi Strizzi; David S. Salomon

Human Cripto‐1 (CR‐1) plays an important role in regulating embryonic development while also regulating various stages of tumor progression. However, mechanisms that regulate CR‐1 expression during embryogenesis and tumorigenesis are still not well defined. In the present study, we investigated the effects of two nuclear receptors, liver receptor homolog (LRH)‐1 and germ cell nuclear factor receptor (GCNF) and epigenetic modifications on CR‐1 gene expression in NTERA‐2 human embryonal carcinoma cells and in breast cancer cells. CR‐1 expression in NTERA‐2 cells was positively regulated by LRH‐1 through direct binding to a DR0 element within the CR‐1 promoter, while GCNF strongly suppressed CR‐1 expression in these cells. In addition, the CR‐1 promoter was unmethylated in NTERA‐2 cells, while T47D, ZR75‐1, and MCF7 breast cancer cells showed high levels of CR‐1 promoter methylation and low CR‐1 mRNA and protein expression. Treatment of breast cancer cells with a demethylating agent and histone deacetylase inhibitors reduced methylation of the CR‐1 promoter and reactivated CR‐1 mRNA and protein expression in these cells, promoting migration and invasion of breast cancer cells. Analysis of a breast cancer tissue array revealed that CR‐1 was highly expressed in the majority of human breast tumors, suggesting that CR‐1 expression in breast cancer cell lines might not be representative of in vivo expression. Collectively, these findings offer some insight into the transcriptional regulation of CR‐1 gene expression and its critical role in the pathogenesis of human cancer. J. Cell. Physiol. 228: 1174–1188, 2013.


Connective Tissue Research | 2015

Cripto-1: an extracellular protein – connecting the sequestered biological dots

Malgorzata Klauzinska; Daniel Bertolette; Sudhamsh Tippireddy; Luigi Strizzi; Peter C. Gray; Monica Gonzales; Meg Duroux; Menotti Ruvo; Christian Wechselberger; Nadia P. Castro; Maria Cristina Rangel; Annalia Focà; Annamaria Sandomenico; Mary J.C. Hendrix; David S. Salomon; Frank Cuttitta

Abstract Cripto-1 (CR-1) is a multifunctional embryonic protein that is re-expressed during inflammation, wound repair, and malignant transformation. CR-1 can function either as a tethered co-receptor or shed as a free ligand underpinning its flexible role in cell physiology. CR-1 has been shown to mediate cell growth, migration, invasion, and induce epithelial to mesenchymal transition (EMT). The main signaling pathways mediating CR-1 effects include Nodal-dependent (Smad2/3) and Nodal-independent (Src/p44/42/Akt) signaling transduction pathways. In addition, there are several naturally occurring binding partner proteins (BPPs) for CR-1 that can either agonize or antagonize its bioactivity. We will review the collective role of CR-1 as an extracellular protein, discuss caveats to consider in developing a quantitation assay, define possible mechanistic avenues applicable for drug discovery, and report on our experimental approaches to overcome these problematic issues.

Collaboration


Dive into the Maria Cristina Rangel's collaboration.

Top Co-Authors

Avatar

Nadia P. Castro

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

David S. Salomon

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Caterina Bianco

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Tadahiro Nagaoka

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Malgorzata Klauzinska

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Monica Gonzales

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Daniel Bertolette

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Robert Callahan

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ahmed Raafat

Michigan State University

View shared research outputs
Researchain Logo
Decentralizing Knowledge