Network


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

Hotspot


Dive into the research topics where Jose Otero is active.

Publication


Featured researches published by Jose Otero.


ACS Applied Materials & Interfaces | 2013

Glioblastoma Behaviors in Three-Dimensional Collagen-Hyaluronan Composite Hydrogels

Shreyas S. Rao; Jessica DeJesus; Aaron R. Short; Jose Otero; Atom Sarkar; Jessica O. Winter

Glioblastoma multiforme (GBM) tumors, which arise from glia in the central nervous system (CNS), are one of the most deadly forms of human cancer with a median survival time of ∼1 year. Their high infiltrative capacity makes them extremely difficult to treat, and even with aggressive multimodal clinical therapies, outcomes are dismal. To improve understanding of cell migration in these tumors, three-dimensional (3D) multicomponent composite hydrogels consisting of collagen and hyaluronic acid, or hyaluronan (HA), were developed. Collagen is a component of blood vessels known to be associated with GBM migration; whereas, HA is one of the major components of the native brain extracellular matrix (ECM). We characterized hydrogel microstructural features and utilized these materials to investigate patient tumor-derived, single cell morphology, spreading, and migration in 3D culture. GBM morphology was influenced by collagen type with cells adopting a rounded morphology in collagen-IV versus a spindle-shaped morphology in collagen-I/III. GBM spreading and migration were inversely dependent on HA concentration; with higher concentrations promoting little or no migration. Further, noncancerous astrocytes primarily displayed rounded morphologies at lower concentrations of HA; in contrast to the spindle-shaped (spread) morphologies of GBMs. These results suggest that GBM behaviors are sensitive to ECM mimetic materials in 3D and that these composite hydrogels could be used to develop 3D brain mimetic models for studying migration processes.


Clinical Cancer Research | 2016

Inhibition of SOAT1 suppresses glioblastoma growth via blocking SREBP-1-mediated lipogenesis

Feng Geng; Xiang Cheng; Xiaoning Wu; Ji Young Yoo; Chunming Cheng; Jeffrey Yunhua Guo; Xiaokui Mo; Peng Ru; Brian Hurwitz; Sung-Hak Kim; Jose Otero; Vinay K. Puduvalli; Etienne Lefai; Jianjie Ma; Ichiro Nakano; Craig Horbinski; Balveen Kaur; Arnab Chakravarti; Deliang Guo

Purpose: Elevated lipogenesis regulated by sterol regulatory element-binding protein-1 (SREBP-1), a transcription factor playing a central role in lipid metabolism, is a novel characteristic of glioblastoma (GBM). The aim of this study was to identify effective approaches to suppress GBM growth by inhibition of SREBP-1. As SREBP activation is negatively regulated by endoplasmic reticulum (ER) cholesterol, we sought to determine whether suppression of sterol O-acyltransferase (SOAT), a key enzyme converting ER cholesterol to cholesterol esters (CE) to store in lipid droplets (LDs), effectively suppressed SREBP-1 and blocked GBM growth. Experimental Design: The presence of LDs in glioma patient tumor tissues was analyzed using immunofluorescence, immunohistochemistry, and electronic microscopy. Western blotting and real-time PCR were performed to analyze protein levels and gene expression of GBM cells, respectively. Intracranial GBM xenografts were used to determine the effects of genetically silencing SOAT1 and SREBP-1 on tumor growth. Results: Our study unraveled that cholesterol esterification and LD formation are signature of GBM, and human patients with glioma possess elevated LDs that correlate with GBM progression and poor survival. We revealed that SOAT1 is highly expressed in GBM and functions as a key player in controlling the cholesterol esterification and storage in GBM. Targeting SOAT1 suppresses GBM growth and prolongs survival in xenograft models via inhibition of SREBP-1–regulated lipid synthesis. Conclusions: Cholesterol esterification and storage in LDs are novel characteristics of GBM, and inhibiting SOAT1 to block cholesterol esterification is a promising therapeutic strategy to treat GBM by suppressing SREBP-1. Clin Cancer Res; 22(21); 5337–48. ©2016 AACR.


Acta Neuropathologica | 2015

Dysregulation of locus coeruleus development in congenital central hypoventilation syndrome

Hiroko Nobuta; Maria Roberta Cilio; Olivier Danhaive; Hui-Hsin Tsai; Srinivasan Tupal; Sandra Chang; Alice T. Murnen; Faith Kreitzer; Verenice Bravo; Catherine Czeisler; Hamza Numan Gokozan; Patrick Gygli; Sean Bush; Debra E. Weese-Mayer; Bruce R. Conklin; Siu-Pok Yee; Eric J. Huang; Paul A. Gray; David H. Rowitch; Jose Otero

Human congenital central hypoventilation syndrome (CCHS), resulting from mutations in transcription factor PHOX2B, manifests with impaired responses to hypoxemia and hypercapnia especially during sleep. To identify brainstem structures developmentally affected in CCHS, we analyzed two postmortem neonatal-lethal cases with confirmed polyalanine repeat expansion (PARM) or Non-PARM (PHOX2B∆8) mutation of PHOX2B. Both human cases showed neuronal losses within the locus coeruleus (LC), which is important for central noradrenergic signaling. Using a conditionally active transgenic mouse model of the PHOX2B∆8 mutation, we found that early embryonic expression (<E10.5) caused failure of LC neuronal specification and perinatal respiratory lethality. In contrast, later onset (E11.5) of PHOX2B∆8 expression was not deleterious to LC development and perinatal respiratory lethality was rescued, despite failure of chemosensor retrotrapezoid nucleus formation. Our findings indicate that early-onset mutant PHOX2B expression inhibits LC neuronal development in CCHS. They further suggest that such mutations result in dysregulation of central noradrenergic signaling, and therefore, potential for early pharmacologic intervention in humans with CCHS.


Oncogene | 2015

CSF1-ETS2-induced microRNA in myeloid cells promote metastatic tumor growth

Haritha Mathsyaraja; Katie Thies; David A. Taffany; Clayton Deighan; Tom Liu; Lianbo Yu; Soledad Fernandez; Charles L. Shapiro; Jose Otero; Cynthia Timmers; Maryam B. Lustberg; Jeffrey J. Chalmers; Gustavo Leone; Michael C. Ostrowski

Metastasis of solid tumors is associated with poor prognosis and bleak survival rates. Tumor-infiltrating myeloid cells (TIMs) are known to promote metastasis, but the mechanisms underlying their collaboration with tumor cells remain unknown. Here, we report an oncogenic role for microRNA (miR) in driving M2 reprogramming in TIMs, characterized by the acquisition of pro-tumor and pro-angiogenic properties. The expression of miR-21, miR-29a, miR-142-3p and miR-223 increased in myeloid cells during tumor progression in mouse models of breast cancer and melanoma metastasis. Further, we show that these miRs are regulated by the CSF1-ETS2 pathway in macrophages. A loss-of-function approach utilizing selective depletion of the miR-processing enzyme Dicer in mature myeloid cells blocks angiogenesis and metastatic tumor growth. Ectopic expression of miR-21 and miR-29a promotes angiogenesis and tumor cell proliferation through the downregulation of anti-angiogenic genes such as Col4a2, Spry1 and Timp3, whereas knockdown of the miRs impedes these processes. miR-21 and miR-29a are expressed in Csf1r+ myeloid cells associated with human metastatic breast cancer, and levels of these miRs in CD115+ non-classical monocytes correlates with metastatic tumor burden in patients. Taken together, our results suggest that miR-21 and miR-29a are essential for the pro-tumor functions of myeloid cells and the CSF1-ETS2 pathway upstream of the miRs serves as an attractive therapeutic target for the inhibition of M2 remodeling of macrophages during malignancy. In addition, miR-21 and miR-29a in circulating myeloid cells may potentially serve as biomarkers to measure therapeutic efficacy of targeted therapies for CSF1 signaling.


Biotechnology Progress | 2015

Glioma-astrocyte interactions on white matter tract-mimetic aligned electrospun nanofibers

Joseph Grodecki; Aaron R. Short; Jessica O. Winter; Shreyas S. Rao; Jose Otero; John J. Lannutti; Atom Sarkar

Gliomas are highly invasive forms of brain cancer comprising more than 50% of brain tumor cases in adults, and astrocytomas account for ∼60‐70% of all gliomas. As a result of multiple factors, including enhanced migratory properties and extracellular matrix remodeling, even with current standards of care, mean survival time for patients is only ∼12 months. Because glioblastoma multiforme (GBM) cells arise from astrocytes, there is great interest in elucidating the interactions of these two cell types in vivo. Previous work performed on two‐dimensional assays (i.e., tissue culture plastic and Boyden chamber assays) utilizes substrates that lack the complexities of the natural microenvironment. Here, we employed a three‐dimensional, electrospun poly‐(caprolactone) (PCL) nanofiber system (NFS) to mimic some features of topographical properties evidenced in vivo. Co‐cultures of human GBM cells and rat astrocytes, as performed on the NFS, showed a significant increase in astrocyte GFAP expression, particularly in the presence of extracellular matrix (ECM) deposited by GBM cells. In addition, GBM migration increased in the presence of astrocytes or soluble factors (i.e., conditioned media). However, the presence of fixed astrocytes acted as an antagonist, lowering GBM migration rates. This data suggests that astrocytes and GBM cells interact through a multitude of pathways, including soluble factors and direct contact. This work demonstrates the potential of the NFS to duplicate some topographical features of the GBM tumor microenvironment, permitting analysis of topographical effects in GBM migration.


Developmental Biology | 2014

Cerebellar cortical lamination and foliation require cyclin A2

Jose Otero; Ilona Kalaszczynska; Wojciech Michowski; Michael Wong; Patrick Gygli; Hamza Numan Gokozan; Amelie Griveau; Junko Odajima; Catherine Czeisler; Fay Patsy Catacutan; Alice T. Murnen; Ulrich Schüller; Piotr Sicinski; David H. Rowitch

The mammalian genome encodes two A-type cyclins, which are considered potentially redundant yet essential regulators of the cell cycle. Here, we tested requirements for cyclin A1 and cyclin A2 function in cerebellar development. Compound conditional loss of cyclin A1/A2 in neural progenitors resulted in severe cerebellar hypoplasia, decreased proliferation of cerebellar granule neuron progenitors (CGNP), and Purkinje (PC) neuron dyslamination. Deletion of cyclin A2 alone showed an identical phenotype, demonstrating that cyclin A1 does not compensate for cyclin A2 loss in neural progenitors. Cyclin A2 loss lead to increased apoptosis at early embryonic time points but not at post-natal time points. In contrast, neural progenitors of the VZ/SVZ did not undergo increased apoptosis, indicating that VZ/SVZ-derived and rhombic lip-derived progenitor cells show differential requirements to cyclin A2. Conditional knockout of cyclin A2 or the SHH proliferative target Nmyc in CGNP also resulted in PC neuron dyslamination. Although cyclin E1 has been reported to compensate for cyclin A2 function in fibroblasts and is upregulated in cyclin A2 null cerebella, cyclin E1 expression was unable to compensate for loss-of cyclin A2 function.


Nature Nanotechnology | 2017

Topical tissue nano-transfection mediates non-viral stroma reprogramming and rescue

Daniel Gallego-Perez; Durba Pal; Subhadip Ghatak; Veysi Malkoc; Natalia Higuita-Castro; Surya Gnyawali; Lingqian Chang; Wei-Ching Liao; Junfeng Shi; Mithun Sinha; Kanhaiya Singh; Erin Steen; Alec Sunyecz; Richard Stewart; Jordan Moore; Thomas Ziebro; Robert Northcutt; Michael Homsy; Paul Bertani; Wu Lu; Sashwati Roy; Savita Khanna; Cameron Rink; Vishnu Baba Sundaresan; Jose Otero; L. James Lee; Chandan K. Sen

Although cellular therapies represent a promising strategy for a number of conditions, current approaches face major translational hurdles, including limited cell sources and the need for cumbersome pre-processing steps (for example, isolation, induced pluripotency). In vivo cell reprogramming has the potential to enable more-effective cell-based therapies by using readily available cell sources (for example, fibroblasts) and circumventing the need for ex vivo pre-processing. Existing reprogramming methodologies, however, are fraught with caveats, including a heavy reliance on viral transfection. Moreover, capsid size constraints and/or the stochastic nature of status quo approaches (viral and non-viral) pose additional limitations, thus highlighting the need for safer and more deterministic in vivo reprogramming methods. Here, we report a novel yet simple-to-implement non-viral approach to topically reprogram tissues through a nanochannelled device validated with well-established and newly developed reprogramming models of induced neurons and endothelium, respectively. We demonstrate the simplicity and utility of this approach by rescuing necrotizing tissues and whole limbs using two murine models of injury-induced ischaemia.


Nanomedicine: Nanotechnology, Biology and Medicine | 2016

Deterministic transfection drives efficient nonviral reprogramming and uncovers reprogramming barriers

Daniel Gallego-Perez; Jose Otero; Catherine Czeisler; Junyu Ma; Cristina Ortiz; Patrick Gygli; Fay Patsy Catacutan; Hamza Numan Gokozan; Aaron Cowgill; Thomas W. Sherwood; Subhadip Ghatak; Veysi Malkoc; Xi Zhao; Wei-Ching Liao; Surya Gnyawali; Xinmei Wang; Andrew F. Adler; Kam W. Leong; Brian C. Wulff; Traci A. Wilgus; Candice C. Askwith; Savita Khanna; Cameron Rink; Chandan K. Sen; L. James Lee

UNLABELLED Safety concerns and/or the stochastic nature of current transduction approaches have hampered nuclear reprogrammings clinical translation. We report a novel non-viral nanotechnology-based platform permitting deterministic large-scale transfection with single-cell resolution. The superior capabilities of our technology are demonstrated by modification of the well-established direct neuronal reprogramming paradigm using overexpression of the transcription factors Brn2, Ascl1, and Myt1l (BAM). Reprogramming efficiencies were comparable to viral methodologies (up to ~9-12%) without the constraints of capsid size and with the ability to control plasmid dosage, in addition to showing superior performance relative to existing non-viral methods. Furthermore, increased neuronal complexity could be tailored by varying BAM ratio and by including additional proneural genes to the BAM cocktail. Furthermore, high-throughput NEP allowed easy interrogation of the reprogramming process. We discovered that BAM-mediated reprogramming is regulated by AsclI dosage, the S-phase cyclin CCNA2, and that some induced neurons passed through a nestin-positive cell stage. FROM THE CLINICAL EDITOR In the field of regenerative medicine, the ability to direct cell fate by nuclear reprogramming is an important facet in terms of clinical application. In this article, the authors described their novel technique of cell reprogramming through overexpression of the transcription factors Brn2, Ascl1, and Myt1l (BAM) by in situ electroporation through nanochannels. This new technique could provide a platform for further future designs.


Journal of Neuropathology and Experimental Neurology | 2015

Mitotic Events in Cerebellar Granule Progenitor Cells that Expand Cerebellar Surface Area Are Critical for Normal Cerebellar Cortical Lamination in Mice

Joshua C. Chang; Mark Leung; Hamza Numan Gokozan; Patrick Gygli; Fay Patsy Catacutan; Catherine Czeisler; Jose Otero

Abstract Late embryonic and postnatal cerebellar folial surface area expansion promotes cerebellar cortical cytoarchitectural lamination. We developed a streamlined sampling scheme to generate unbiased estimates of murine cerebellar surface area and volume using stereologic principles. We demonstrate that, during the proliferative phase of the external granular layer (EGL) and folial surface area expansion, EGL thickness does not change and thus is a topological proxy for progenitor self-renewal. The topological constraints indicate that, during proliferative phases, migration out of the EGL is balanced by self-renewal. Progenitor self-renewal must, therefore, include mitotic events yielding 2 cells in the same layer to increase surface area (&bgr; events) and mitotic events yielding 2 cells, with 1 cell in a superficial layer and 1 cell in a deeper layer (&agr; events). As the cerebellum grows, therefore, &bgr; events lie upstream of &agr; events. Using a mathematical model constrained by the measurements of volume and surface area, we could quantify intermitotic times for &bgr; events on a per-cell basis in postnatal mouse cerebellum. Furthermore, we found that loss of CCNA2, which decreases EGL proliferation and secondarily induces cerebellar cortical dyslamination, shows preserved &agr;-type events. Thus, CCNA2-null cerebellar granule progenitor cells are capable of self-renewal of the EGL stem cell niche; this is concordant with prior findings of extensive apoptosis in CCNA2-null mice. Similar methodologies may provide another layer of depth to the interpretation of results from stereologic studies.


The Journal of Comparative Neurology | 2016

Area postrema undergoes dynamic postnatal changes in mice and humans

Hamza Numan Gokozan; Faisal Baig; Sarah Corcoran; Fay Patsy Catacutan; Patrick Gygli; Ana C. Takakura; Thiago S. Moreira; Catherine Czeisler; Jose Otero

The postnatal period in mammals represents a developmental epoch of significant change in the autonomic nervous system (ANS). This study focuses on postnatal development of the area postrema, a crucial ANS structure that regulates temperature, breathing, and satiety, among other activities. We find that the human area postrema undergoes significant developmental changes during postnatal development. To characterize these changes further, we used transgenic mouse reagents to delineate neuronal circuitry. We discovered that, although a well‐formed ANS scaffold exists early in embryonic development, the area postrema shows a delayed maturation. Specifically, postnatal days 0–7 in mice show no significant change in area postrema volume or synaptic input from PHOX2B‐derived neurons. In contrast, postnatal days 7–20 show a significant increase in volume and synaptic input from PHOX2B‐derived neurons. We conclude that key ANS structures show unexpected dynamic developmental changes during postnatal development. These data provide a basis for understanding ANS dysfunction and disease predisposition in premature and postnatal humans. J. Comp. Neurol. 524:1259–1269, 2016.

Collaboration


Dive into the Jose Otero's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Behiye Kaya

The Ohio State University Wexner Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge