Network


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

Hotspot


Dive into the research topics where Maria C. Cuitiño is active.

Publication


Featured researches published by Maria C. Cuitiño.


Neoplasia | 2017

Stromal PDGFR-α Activation Enhances Matrix Stiffness, Impedes Mammary Ductal Development, and Accelerates Tumor Growth

Anisha M. Hammer; Gina M. Sizemore; Vasudha C. Shukla; Alex Avendano; Steven T. Sizemore; Jonathan J. Chang; Raleigh D. Kladney; Maria C. Cuitiño; Katie Thies; Quinn Verfurth; Arnab Chakravarti; Lisa D. Yee; Gustavo Leone; Jonathan W. Song; Samir N. Ghadiali; Michael C. Ostrowski

The extracellular matrix (ECM) is critical for mammary ductal development and differentiation, but how mammary fibroblasts regulate ECM remodeling remains to be elucidated. Herein, we used a mouse genetic model to activate platelet derived growth factor receptor-alpha (PDGFRα) specifically in the stroma. Hyperactivation of PDGFRα in the mammary stroma severely hindered pubertal mammary ductal morphogenesis, but did not interrupt the lobuloalveolar differentiation program. Increased stromal PDGFRα signaling induced mammary fat pad fibrosis with a corresponding increase in interstitial hyaluronic acid (HA) and collagen deposition. Mammary fibroblasts with PDGFRα hyperactivation also decreased hydraulic permeability of a collagen substrate in an in vitro microfluidic device assay, which was mitigated by inhibition of either PDGFRα or HA. Fibrosis seen in this model significantly increased the overall stiffness of the mammary gland as measured by atomic force microscopy. Further, mammary tumor cells injected orthotopically in the fat pads of mice with stromal activation of PDGFRα grew larger tumors compared to controls. Taken together, our data establish that aberrant stromal PDGFRα signaling disrupts ECM homeostasis during mammary gland development, resulting in increased mammary stiffness and increased potential for tumor growth.


Genes & Development | 2016

Genetic ablation of Smoothened in pancreatic fibroblasts increases acinar–ductal metaplasia

Xin Liu; Jason R. Pitarresi; Maria C. Cuitiño; Raleigh D. Kladney; Sarah Woelke; Gina M. Sizemore; Sunayana G. Nayak; Onur Egriboz; Patrick G. Schweickert; Lianbo Yu; Stefan Trela; Daniel J. Schilling; Shannon K. Halloran; Maokun Li; Shourik Dutta; Soledad Fernandez; Thomas J. Rosol; Gregory B. Lesinski; Reena Shakya; Thomas Ludwig; Stephen F. Konieczny; Gustavo Leone; Jinghai Wu; Michael C. Ostrowski

The contribution of the microenvironment to pancreatic acinar-to-ductal metaplasia (ADM), a preneoplastic transition in oncogenic Kras-driven pancreatic cancer progression, is currently unclear. Here we show that disruption of paracrine Hedgehog signaling via genetic ablation of Smoothened (Smo) in stromal fibroblasts in a Kras(G12D) mouse model increased ADM. Smo-deleted fibroblasts had higher expression of transforming growth factor-α (Tgfa) mRNA and secreted higher levels of TGFα, leading to activation of EGFR signaling in acinar cells and increased ADM. The mechanism involved activation of AKT and noncanonical activation of the GLI family transcription factor GLI2. GLI2 was phosphorylated at Ser230 in an AKT-dependent fashion and directly regulated Tgfa expression in fibroblasts lacking Smo Additionally, Smo-deleted fibroblasts stimulated the growth of Kras(G12D)/Tp53(R172H) pancreatic tumor cells in vivo and in vitro. These results define a non-cell-autonomous mechanism modulating Kras(G12D)-driven ADM that is balanced by cross-talk between Hedgehog/SMO and AKT/GLI2 pathways in stromal fibroblasts.


Genes & Development | 2015

Noncatalytic PTEN missense mutation predisposes to organ-selective cancer development in vivo

Enrico Caserta; Onur Egriboz; Hui Wang; Chelsea K. Martin; Christopher Koivisto; Thierry Pécot; Raleigh D. Kladney; Changxian Shen; Kang-Sup Shim; Thac Pham; Matthew K. Karikomi; Melissa J. Mauntel; Sarmila Majumder; Maria C. Cuitiño; Xing Tang; Arunima Srivastava; Lianbo Yu; Julie A. Wallace; Xiaokui Mo; Morag Park; Soledad Fernandez; Robert Pilarski; Krista La Perle; Thomas J. Rosol; Vincenzo Coppola; Diego H. Castrillon; Cynthia Timmers; David E. Cohn; David M. O'Malley; Floor J. Backes

Inactivation of phosphatase and tensin homology deleted on chromosome 10 (PTEN) is linked to increased PI3K-AKT signaling, enhanced organismal growth, and cancer development. Here we generated and analyzed Pten knock-in mice harboring a C2 domain missense mutation at phenylalanine 341 (Pten(FV)), found in human cancer. Despite having reduced levels of PTEN protein, homozygous Pten(FV/FV) embryos have intact AKT signaling, develop normally, and are carried to term. Heterozygous Pten(FV/+) mice develop carcinoma in the thymus, stomach, adrenal medulla, and mammary gland but not in other organs typically sensitive to Pten deficiency, including the thyroid, prostate, and uterus. Progression to carcinoma in sensitive organs ensues in the absence of overt AKT activation. Carcinoma in the uterus, a cancer-resistant organ, requires a second clonal event associated with the spontaneous activation of AKT and downstream signaling. In summary, this PTEN noncatalytic missense mutation exposes a core tumor suppressor function distinct from inhibition of canonical AKT signaling that predisposes to organ-selective cancer development in vivo.


Journal of Clinical Investigation | 2016

E2f8 mediates tumor suppression in postnatal liver development

Lindsey N. Kent; Jessica B. Rakijas; Shusil K. Pandit; Bart Westendorp; Hui-Zi Chen; Justin T. Huntington; Xing Tang; Sooin Bae; Arunima Srivastava; Shantibhusan Senapati; Christopher Koivisto; Chelsea K. Martin; Maria C. Cuitiño; Miguel Perez; Julian M. Clouse; Veda Chokshi; Neelam Shinde; Raleigh D. Kladney; Daokun Sun; Antonio Perez-Castro; Ramadhan B. Matondo; Sathidpak Nantasanti; Michal Mokry; Kun Huang; Raghu Machiraju; Soledad Fernandez; Thomas J. Rosol; Vincenzo Coppola; Kamal S. Pohar; James M. Pipas

E2F-mediated transcriptional repression of cell cycle-dependent gene expression is critical for the control of cellular proliferation, survival, and development. E2F signaling also interacts with transcriptional programs that are downstream of genetic predictors for cancer development, including hepatocellular carcinoma (HCC). Here, we evaluated the function of the atypical repressor genes E2f7 and E2f8 in adult liver physiology. Using several loss-of-function alleles in mice, we determined that combined deletion of E2f7 and E2f8 in hepatocytes leads to HCC. Temporal-specific ablation strategies revealed that E2f8s tumor suppressor role is critical during the first 2 weeks of life, which correspond to a highly proliferative stage of postnatal liver development. Disruption of E2F8s DNA binding activity phenocopied the effects of an E2f8 null allele and led to HCC. Finally, a profile of chromatin occupancy and gene expression in young and tumor-bearing mice identified a set of shared targets for E2F7 and E2F8 whose increased expression during early postnatal liver development is associated with HCC progression in mice. Increased expression of E2F8-specific target genes was also observed in human liver biopsies from HCC patients compared to healthy patients. In summary, these studies suggest that E2F8-mediated transcriptional repression is a critical tumor suppressor mechanism during postnatal liver development.


Frontiers in Oncology | 2014

Protein Kinase C Beta in the Tumor Microenvironment Promotes Mammary Tumorigenesis

Julie A. Wallace; Jason R. Pitarresi; Nandini Sharma; Marilly Palettas; Maria C. Cuitiño; Steven T. Sizemore; Lianbo Yu; Allen Sanderlin; Thomas J. Rosol; Kamal D. Mehta; Gina M. Sizemore; Michael C. Ostrowski

Protein kinase C beta (PKCβ) expression in breast cancer is associated with a more aggressive tumor phenotype, yet the mechanism for how PKCβ is pro-tumorigenic in this disease is still unclear. Interestingly, while it is known that PKCβ mediates angiogenesis, immunity, fibroblast function and adipogenesis, all components of the mammary tumor microenvironment (TME), no study to date has investigated whether stromal PKCβ is functionally relevant in breast cancer. Herein, we evaluate mouse mammary tumor virus–polyoma middle T-antigen (MMTV–PyMT) induced mammary tumorigenesis in the presence and absence of PKCβ. We utilize two model systems: one where PKCβ is deleted in both the epithelial and stromal compartments to test the global requirement for PKCβ on tumor formation, and second, where PKCβ is deleted only in the stromal compartment to test its role in the TME. MMTV–PyMT mice globally lacking PKCβ live longer and develop smaller tumors with decreased proliferation and decreased macrophage infiltration. Similarly, when PKCβ is null exclusively in the stroma, PyMT-driven B6 cells form smaller tumors with diminished collagen deposition. These experiments reveal for the first time a tumor promoting role for stromal PKCβ in MMTV–PyMT tumorigenesis. In corroboration with these results, PKCβ mRNA (Prkcb) is increased in fibroblasts isolated from MMTV–PyMT tumors. These data were confirmed in a breast cancer patient cohort. Combined these data suggest the continued investigation of PKCβ in the mammary TME is necessary to elucidate how to effectively target this signaling pathway in breast cancer.


Oncogene | 2018

Fibroblast-derived CXCL12 promotes breast cancer metastasis by facilitating tumor cell intravasation

Dinesh K. Ahirwar; Mohd W. Nasser; Madhu M. Ouseph; Mohamad Elbaz; Maria C. Cuitiño; Raleigh D. Kladney; Sanjay Varikuti; Kirti Kaul; Abhay R. Satoskar; Bhuvaneswari Ramaswamy; Xiaoli Zhang; Michael C. Ostrowski; Gustavo Leone; Ramesh K. Ganju

The chemokine CXCL12 has been shown to regulate breast tumor growth, however, its mechanism in initiating distant metastasis is not well understood. Here, we generated a novel conditional allele of Cxcl12 in mice and used a fibroblast-specific Cre transgene along with various mammary tumor models to evaluate CXCL12 function in the breast cancer metastasis. Ablation of CXCL12 in stromal fibroblasts of mice significantly delayed the time to tumor onset and inhibited distant metastasis in different mouse models. Elucidation of mechanisms using in vitro and in vivo model systems revealed that CXCL12 enhances tumor cell intravasation by increasing vascular permeability and expansion of a leaky tumor vasculature. Furthermore, our studies revealed CXCL12 enhances permeability by recruiting endothelial precursor cells and decreasing endothelial tight junction and adherence junction proteins. High expression of stromal CXCL12 in large cohort of breast cancer patients was directly correlated to blood vessel density and inversely correlated to recurrence and overall patient survival. In addition, our analysis revealed that stromal CXCL12 levels in combination with number of CD31+ blood vessels confers poorer patient survival compared to individual protein level. However, no correlation was observed between epithelial CXCL12 and patient survival or blood vessel density. Our findings describe the novel interactions between fibroblasts-derived CXCL12 and endothelial cells in facilitating tumor cell intrvasation, leading to distant metastasis. Overall, our studies indicate that cross-talk between fibroblast-derived CXCL12 and endothelial cells could be used as novel biomarker and strategy for developing tumor microenvironment based therapies against aggressive and metastatic breast cancer.


Neoplasia | 2016

Stromal ETS2 Regulates Chemokine Production and Immune Cell Recruitment during Acinar-to-Ductal Metaplasia

Jason R. Pitarresi; Xin Liu; Sudarshana M. Sharma; Maria C. Cuitiño; Raleigh D. Kladney; Thomas A. Mace; Sydney Donohue; Sunayana G. Nayak; Chunjing Qu; James Lee; Sarah Woelke; Stefan Trela; Kyle M. LaPak; Lianbo Yu; Joseph P. McElroy; Thomas J. Rosol; Reena Shakya; Thomas Ludwig; Gregory B. Lesinski; Soledad Fernandez; Stephen F. Konieczny; Gustavo Leone; Jinghai Wu; Michael C. Ostrowski

Preclinical studies have suggested that the pancreatic tumor microenvironment both inhibits and promotes tumor development and growth. Here we establish the role of stromal fibroblasts during acinar-to-ductal metaplasia (ADM), an initiating event in pancreatic cancer formation. The transcription factor V-Ets avian erythroblastosis virus E26 oncogene homolog 2 (ETS2) was elevated in smooth muscle actin–positive fibroblasts in the stroma of pancreatic ductal adenocarcinoma (PDAC) patient tissue samples relative to normal pancreatic controls. LSL-KrasG12D/+; LSL-Trp53R172H/+; Pdx-1-Cre (KPC) mice showed that ETS2 expression initially increased in fibroblasts during ADM and remained elevated through progression to PDAC. Conditional ablation of Ets-2 in pancreatic fibroblasts in a KrasG12D-driven mouse ADM model decreased the amount of ADM events. ADMs from fibroblast Ets-2–deleted animals had reduced epithelial cell proliferation and increased apoptosis. Surprisingly, fibroblast Ets-2 deletion significantly altered immune cell infiltration into the stroma, with an increased CD8+ T-cell population, and decreased presence of regulatory T cells (Tregs), myeloid-derived suppressor cells, and mature macrophages. The mechanism involved ETS2-dependent chemokine ligand production in fibroblasts. ETS2 directly bound to regulatory sequences for Ccl3, Ccl4, Cxcl4, Cxcl5, and Cxcl10, a group of chemokines that act as potent mediators of immune cell recruitment. These results suggest an unappreciated role for ETS2 in fibroblasts in establishing an immune-suppressive microenvironment in response to oncogenic KrasG12D signaling during the initial stages of tumor development.


PLOS ONE | 2017

Generation of a pancreatic cancer model using a Pdx1-Flp recombinase knock-in allele

Jinghai Wu; Xin Liu; Sunayana G. Nayak; Jason R. Pitarresi; Maria C. Cuitiño; Lianbo Yu; Blake Eason Hildreth; Katie Thies; Daniel J. Schilling; Soledad Fernandez; Gustavo Leone; Michael C. Ostrowski

The contribution of the tumor microenvironment to the development of pancreatic adenocarcinoma (PDAC) is unclear. The LSL-KrasG12D/+;LSL-p53R172H/+;Pdx-1-Cre (KPC) tumor model, which is widely utilized to faithfully recapitulate human pancreatic cancer, depends on Cre-mediated recombination in the epithelial lineage to drive tumorigenesis. Therefore, specific Cre-loxP recombination in stromal cells cannot be applied in this model, limiting the in vivo investigation of stromal genetics in tumor initiation and progression. To address this issue, we generated a new Pdx1FlpO knock-in mouse line, which represents the first mouse model to physiologically express FlpO recombinase in pancreatic epithelial cells. This mouse specifically recombines Frt loci in pancreatic epithelial cells, including acinar, ductal, and islet cells. When combined with the Frt-STOP-Frt KrasG12D and p53Frt mouse lines, simultaneous Pdx1FlpO activation of mutant Kras and deletion of p53 results in the spectrum of pathologic changes seen in PDAC, including PanIN lesions and ductal carcinoma. Combination of this KPF mouse model with any stroma-specific Cre can be used to conditionally modify target genes of interest. This will provide an excellent in vivo tool to study the roles of genes in different cell types and multiple cell compartments within the pancreatic tumor microenvironment.


Nature Communications | 2018

Stromal PTEN determines mammary epithelial response to radiotherapy

Gina M. Sizemore; Subhasree Balakrishnan; Katie Thies; Anisha M. Hammer; Steven T. Sizemore; Anthony J. Trimboli; Maria C. Cuitiño; Sarah A. Steck; Gary Tozbikian; Raleigh D. Kladney; Neelam Shinde; Manjusri Das; Dongju Park; Sarmila Majumder; Shiva Krishnan; Lianbo Yu; Soledad Fernandez; Arnab Chakravarti; Peter G. Shields; Lisa D. Yee; Thomas J. Rosol; Thomas Ludwig; Morag Park; Gustavo Leone; Michael C. Ostrowski

The importance of the tumor–associated stroma in cancer progression is clear. However, it remains uncertain whether early events in the stroma are capable of initiating breast tumorigenesis. Here, we show that in the mammary glands of non-tumor bearing mice, stromal-specific phosphatase and tensin homolog (Pten) deletion invokes radiation-induced genomic instability in neighboring epithelium. In these animals, a single dose of whole-body radiation causes focal mammary lobuloalveolar hyperplasia through paracrine epidermal growth factor receptor (EGFR) activation, and EGFR inhibition abrogates these cellular changes. By analyzing human tissue, we discover that stromal PTEN is lost in a subset of normal breast samples obtained from reduction mammoplasty, and is predictive of recurrence in breast cancer patients. Combined, these data indicate that diagnostic or therapeutic chest radiation may predispose patients with decreased stromal PTEN expression to secondary breast cancer, and that prophylactic EGFR inhibition may reduce this risk.The tumor microenvironment influences tumor progression. Here the authors show that lack of stromal PTEN phosphatase induces DNA repair defects in the neighboring mammary gland epithelial cells via hyperactivation of EGF-receptor signaling, resulting in higher radiation-induced DNA damage and hyperplasia.


Life Science Alliance | 2018

Disruption of stromal hedgehog signaling initiates RNF5-mediated proteasomal degradation of PTEN and accelerates pancreatic tumor growth

Jason R. Pitarresi; Xin Liu; Alex Avendano; Katie Thies; Gina M. Sizemore; Anisha M. Hammer; Blake E. Hildreth; David J Wang; Sarah A. Steck; Sydney Donohue; Maria C. Cuitiño; Raleigh D. Kladney; Thomas A. Mace; Jonathan J. Chang; Christina S Ennis; Huiqing Li; Roger H. Reeves; Seth Blackshaw; Jianying Zhang; Lianbo Yu; Soledad Fernandez; Wendy L. Frankel; Mark Bloomston; Thomas J. Rosol; Gregory B. Lesinski; Stephen F. Konieczny; Denis C. Guttridge; Anil K. Rustgi; Gustavo Leone; Jonathan W. Song

Disrupting paracrine Hedgehog signaling in pancreatic cancer stroma through genetic deletion of fibroblast Smoothened leads to proteasomal degradation of fibroblast PTEN and accelerates tumor growth. The contribution of the tumor microenvironment to pancreatic ductal adenocarcinoma (PDAC) development is currently unclear. We therefore examined the consequences of disrupting paracrine Hedgehog (HH) signaling in PDAC stroma. Herein, we show that ablation of the key HH signaling gene Smoothened (Smo) in stromal fibroblasts led to increased proliferation of pancreatic tumor cells. Furthermore, Smo deletion resulted in proteasomal degradation of the tumor suppressor PTEN and activation of oncogenic protein kinase B (AKT) in fibroblasts. An unbiased proteomic screen identified RNF5 as a novel E3 ubiquitin ligase responsible for degradation of phosphatase and tensin homolog (PTEN) in Smo-null fibroblasts. Ring Finger Protein 5 (Rnf5) knockdown or pharmacological inhibition of glycogen synthase kinase 3β (GSKβ), the kinase that marks PTEN for ubiquitination, rescued PTEN levels and reversed the oncogenic phenotype, identifying a new node of PTEN regulation. In PDAC patients, low stromal PTEN correlated with reduced overall survival. Mechanistically, PTEN loss decreased hydraulic permeability of the extracellular matrix, which was reversed by hyaluronidase treatment. These results define non-cell autonomous tumor-promoting mechanisms activated by disruption of the HH/PTEN axis and identifies new targets for restoring stromal tumor-suppressive functions.

Collaboration


Dive into the Maria C. Cuitiño's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lianbo Yu

Ohio State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge