Network


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

Hotspot


Dive into the research topics where Elena Romm is active.

Publication


Featured researches published by Elena Romm.


Journal of Neurochemistry | 2005

Myt1 family recruits histone deacetylase to regulate neural transcription.

Elena Romm; Joseph A. Nielsen; Jin G. Kim; Lynn D. Hudson

The myelin transcription factor 1 (Myt1) gene family is comprised of three zinc finger genes [Myt1, Myt1L (Myt1‐Like) and NZF3] of the structurally unique CCHHC class that are expressed predominantly in the developing CNS. To understand the mechanism by which this family regulates neural differentiation, we searched for interaction partners. In both yeast and a mammalian two‐hybrid system, Myt1 and Myt1L interacted with Sin3B, a protein that mediates transcriptional repression by binding to histone deacetylases (HDACs). Myt1–Sin3B complexes were co‐immunoprecipitated from transfected mammalian cells and included HDAC1 and HDAC2. Myt1 and Myt1L could partner with all three Sin3B isoforms, the long form (Sin3BLF) that includes the HDAC‐binding domain, and the two short forms (Sin3BSF293 and Sin3BSF302) that lack this domain and may consequently antagonize Sin3BLF/HDAC‐mediated co‐repression. Myt1 or Myt1L interactions with the HDAC‐binding form of Sin3B conferred repression on a heterologous promoter. Oligodendrocytes were shown to express transcripts encoding each of the Sin3B isoforms. We present a model in which the Myt1 family of zinc finger proteins, when bound to a neural promoter, can recruit Sin3B. Depending on the relative availability of Sin3B isoforms, the Myt1 gene family may favor the silencing of genes during neural development.


Annals of Neurology | 2015

Cerebrospinal fluid markers reveal intrathecal inflammation in progressive multiple sclerosis.

Mika Komori; Andrew Blake; Mark C. Greenwood; Yen Chih Lin; Peter Kosa; Danish Ghazali; Paige Winokur; Muktha Natrajan; Simone C. Wuest; Elena Romm; Anil A. Panackal; Peter R. Williamson; Tianxia Wu; Bibiana Bielekova

The management of complex patients with neuroimmunological diseases is hindered by an inability to reliably measure intrathecal inflammation. Currently implemented laboratory tests developed >40 years ago either are not dynamic or fail to capture low levels of central nervous system (CNS) inflammation. Therefore, we aimed to identify and validate biomarkers of CNS inflammation in 2 blinded, prospectively acquired cohorts of untreated patients with neuroimmunological diseases and embedded controls, with the ultimate goal of developing clinically useful tools.


Journal of Immunology | 2014

Comprehensive Immunophenotyping of Cerebrospinal Fluid Cells in Patients with Neuroimmunological Diseases

Sungpil Han; Yen Chih Lin; Tianxia Wu; Alan Salgado; Ina Mexhitaj; Simone C. Wuest; Elena Romm; Joan Ohayon; Raphaela Goldbach-Mansky; Adeline Vanderver; Adriana Marques; Camilo Toro; Peter R. Williamson; Irene Cortese; Bibiana Bielekova

We performed unbiased, comprehensive immunophenotyping of cerebrospinal fluid (CSF) and blood leukocytes in 221 subjects referred for the diagnostic work-up of neuroimmunological disorders to obtain insight about disease-specific phenotypes of intrathecal immune responses. Quantification of 14 different immune cell subsets, coupled with the assessment of their activation status, revealed physiological differences between intrathecal and systemic immunity, irrespective of final diagnosis. Our data are consistent with a model where the CNS shapes intrathecal immune responses to provide effective protection against persistent viral infections, especially by memory T cells, plasmacytoid dendritic cells, and CD56bright NK cells. Our data also argue that CSF immune cells do not simply reflect cells recruited from the periphery. Instead, they represent a mixture of cells that are recruited from the blood, have been activated intrathecally and leave the CNS after performing effector functions. Diagnosis-specific differences provide mechanistic insight into the disease process in the defined subtypes of multiple sclerosis (MS), neonatal onset multisystem inflammatory disease, and Aicardi–Goutières syndrome. This analysis also determined that secondary-progressive MS patients are immunologically closer to relapsing–remitting patients as compared with patients with primary-progressive MS. Because CSF immunophenotyping captures the biology of the intrathecal inflammatory processes, it has the potential to guide optimal selection of immunomodulatory therapies in individual patients and monitor their efficacy. Our study adds to the increasing number of publications that demonstrate poor correlation between systemic and intrathecal inflammatory biomarkers in patients with neuroimmunological diseases and stresses the importance of studying immune responses directly in the intrathecal compartment.


Annals of clinical and translational neurology | 2015

Daclizumab reverses intrathecal immune cell abnormalities in multiple sclerosis

Yen Chih Lin; Paige Winokur; Andrew Blake; Tianxia Wu; Elena Romm; Bibiana Bielekova

Novel treatments such as natalizumab and fingolimod achieve their therapeutic efficacy in multiple sclerosis (MS) by blocking access of subsets of immune cells into the central nervous system, thus creating nonphysiological intrathecal immunity. In contrast, daclizumab, a humanized monoclonal antibody against the alpha chain of the IL‐2 receptor, has a unique mechanism of action with multiple direct effects on innate immunity. As cellular intrathecal abnormalities corresponding to MS have been well defined, we asked how daclizumab therapy affects these immunological hallmarks of the MS disease process.


PLOS ONE | 2014

A Complex Role of Herpes Viruses in the Disease Process of Multiple Sclerosis

Simone C. Wuest; Ina Mexhitaj; Noo Ri Chai; Elena Romm; Joerg Scheffel; Biying Xu; Kelly Lane; Tianxia Wu; Bibiana Bielekova

Multiple sclerosis (MS) is a chronic inflammatory disorder of the central nervous system (CNS). Neither the antigenic target(s) nor the cell population(s) responsible for CNS tissue destruction in MS have been fully defined. The objective of this study was to simultaneously determine the antigen (Ag)-specificity and phenotype of un-manipulated intrathecal CD4+ and CD8+ T cells of patients with relapsing-remitting and progressive MS compared to subjects with other inflammatory neurological diseases. We applied a novel Ag-recognition assay based on co-cultures of freshly obtained cerebrospinal fluid T cells and autologous dendritic cells pre-loaded with complex candidate Ags. We observed comparably low T cell responses to complex auto-Ags including human myelin, brain homogenate, and cell lysates of apoptotically modified oligodendroglial and neuronal cells in all cohorts and both compartments. Conversely, we detected a strong intrathecal enrichment of Epstein-Barr virus- and human herpes virus 6-specific (but not cytomegalovirus-specific) reactivities of the Th1-phenotype throughout all patients. Qualitatively, the intrathecal enrichment of herpes virus reactivities was more pronounced in MS patients. This enrichment was completely reversed by long-term treatment with the IL-2 modulating antibody daclizumab, which strongly inhibits MS disease activity. Finally, we observed a striking discrepancy between diminished intrathecal T cell proliferation and enhanced cytokine production of herpes virus-specific T cells among progressive MS patients, consistent with the phenotype of terminally differentiated cells. The data suggest that intrathecal administration of novel therapeutic agents targeting immune cells outside of the proliferation cycle may be necessary to effectively eliminate intrathecal inflammation in progressive MS.


Glia | 2004

Expression of a catalytically inactive transmembrane protein tyrosine phosphatase ϵ (tm-PTPϵ) delays optic nerve myelination

Naser Muja; Gabor Lovas; Elena Romm; Dietrich Machleder; Mukul Ranjan; Vittorio Gallo; Lynn D. Hudson

Reversible tyrosine phosphorylation is integral to the process of oligodendrocyte differentiation. To interfere with the subset of the phosphorylation cycle overseen by protein tyrosine phosphatase ϵ (PTPϵ) in oligodendrocytes, we applied a substrate‐trapping approach in the development of transgenic mice overexpressing a catalytically inactive, transmembrane PTPϵ‐hemaglutinin (tm‐PTPϵ‐HA) from the dual promoter element of the gene encoding the myelin protein 2′,3′‐cyclic nucleotide 3′‐phosphodiesterase (CNP). Transgene expression peaked during the active myelinating period, at 2–3 weeks postnatal. Two tyrosine phosphoproteins, α‐enolase and β‐actin, were phosphorylated to a greater degree in transgenic mice. Despite a high degree of tm‐PTPϵ‐HA expression, myelin was grossly normal in nearly all axonal tracts. Phenotypic abnormalities were limited to optic nerve, where a decrease in the degree of myelination was reflected by reduced levels of myelin proteins on postnatal day 21 (PND21), as well as a decrease in the density of differentiated oligodendrocytes. The optic chiasm was reduced in thickness in transgenic mice; optic nerves similarly exhibited a reduction in transverse width. Further analyses of the optic pathway demonstrated that transgenic protein was unexpectedly present in retinal ganglion cells, whose axons are the targets of myelination by optic nerve oligodendrocytes. On PND28, transgenic protein declined dramatically in both oligodendrocytes and retinal ganglion cells contributing to the recovery of optic nerve myelination. Thus, delayed myelination arises only when tm‐PTPϵ‐HA is simultaneously expressed in myelin‐forming glia and their neuronal targets. While tm‐PTPϵ related signaling pathways may figure in axon‐glial interactions, interfering with tm‐PTPϵ activity does not perceptibly affect the development or myelinating capacity of most oligodendrocytes.


Clinical Infectious Diseases | 2017

Spinal Arachnoiditis as a Complication of Cryptococcal Meningoencephalitis in Non-HIV Previously Healthy Adults.

Anil A. Panackal; Mika Komori; Peter Kosa; Omar Khan; Dima A. Hammoud; Lindsey B. Rosen; Sarah K. Browne; Yen-Chih Lin; Elena Romm; Charu Ramaprasad; Bettina C. Fries; John E. Bennett; Bibiana Bielekova; Peter R. Williamson

Background. Cryptococcus can cause meningoencephalitis (CM) among previously healthy non-HIV adults. Spinal arachnoiditis is under-recognized, since diagnosis is difficult with concomitant central nervous system (CNS) pathology. Methods. We describe 6 cases of spinal arachnoiditis among 26 consecutively recruited CM patients with normal CD4 counts who achieved microbiologic control. We performed detailed neurological exams, cerebrospinal fluid (CSF) immunophenotyping and biomarker analysis before and after adjunctive immunomodulatory intervention with high dose pulse corticosteroids, affording causal inference into pathophysiology. Results. All 6 exhibited severe lower motor neuron involvement in addition to cognitive changes and gait disturbances from meningoencephalitis. Spinal involvement was associated with asymmetric weakness and urinary retention. Diagnostic specificity was improved by MRI imaging which demonstrated lumbar spinal nerve root enhancement and clumping or lesions. Despite negative fungal cultures, CSF inflammatory biomarkers, sCD27 and sCD21, as well as the neuronal damage biomarker, neurofilament light chain (NFL), were elevated compared to healthy donor (HD) controls. Elevations in these biomarkers were associated with clinical symptoms and showed improvement with adjunctive high dose pulse corticosteroids. Conclusions. These data suggest that a post-infectious spinal arachnoiditis is an important complication of CM in previously healthy individuals, requiring heightened clinician awareness. Despite microbiological control, this syndrome causes significant pathology likely due to increased inflammation and may be amenable to suppressive therapeutics.


Neuroimmunology and Neuroinflammation | 2016

Patients with MS under daclizumab therapy mount normal immune responses to influenza vaccination

Yen Chih Lin; Paige Winokur; Andrew Blake; Tianxia Wu; Jody Manischewitz; Lisa R. King; Elena Romm; Hana Golding; Bibiana Bielekova

Objective: The purpose of this study was to assess the potential immunosuppressive role of daclizumab, a humanized monoclonal antibody against the α chain of the interleukin 2 receptor, in vivo, by comparing immune responses to the 2013 seasonal influenza vaccination between patients with multiple sclerosis (MS) on long-term daclizumab therapy and controls. Methods: Previously defined subpopulations of adaptive immune cells known to correlate with the immune response to the influenza vaccination were evaluated by 12-color flow cytometry in 23 daclizumab-treated patients with MS and 14 MS or healthy controls before (D0) and 1 day (D1) and 7 days (D7) after administration of the 2013 Afluria vaccine. Neutralizing antibody titers and CD4+, CD8+ T cell, B cell, and natural killer cell proliferation to 3 strains of virus contained in the Afluria vaccine were assessed at D0, D7, and 180 days postvaccination. Results: Daclizumab-treated patients and controls demonstrated comparable, statistically significant expansions of previously defined subpopulations of activated CD8+ T cells and B cells that characterize the development of effective immune responses to the influenza vaccine, while proliferation of T cells to influenza and control antigens was diminished in the daclizumab cohort. All participants fulfilled FDA criteria for seroconversion or seroprotection in antibody assays. Conclusion: Despite the mild immunosuppressive effects of daclizumab in vivo demonstrated by an increased incidence of infectious complications in clinical trials, patients with MS under daclizumab therapy mount normal antibody responses to influenza vaccinations.


Molecular and Cellular Biochemistry | 2011

Mouse transmembrane BAX inhibitor Motif 3 (Tmbim3) encodes a 38 kDa transmembrane protein expressed in the central nervous system

Joseph A. Nielsen; Meredith A. Chambers; Elena Romm; Laurel Yong-Hwa Lee; Jo Ann Berndt; Lynn D. Hudson


Transgenic Research | 2011

A tool for examining the role of the zinc finger myelin transcription factor 1 (Myt1) in neural development: Myt1 knock-in mice

Lynn D. Hudson; Elena Romm; Jo Ann Berndt; Joseph A. Nielsen

Collaboration


Dive into the Elena Romm's collaboration.

Top Co-Authors

Avatar

Bibiana Bielekova

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Tianxia Wu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Andrew Blake

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Lynn D. Hudson

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Paige Winokur

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Simone C. Wuest

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Peter Kosa

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Yen Chih Lin

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Yen-Chih Lin

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Joseph A. Nielsen

Food and Drug Administration

View shared research outputs
Researchain Logo
Decentralizing Knowledge