Network


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

Hotspot


Dive into the research topics where Michael J. Romanko is active.

Publication


Featured researches published by Michael J. Romanko.


Developmental Neuroscience | 2001

Hypoxia/Ischemia Depletes the Rat Perinatal Subventricular Zone of Oligodendrocyte Progenitors and Neural Stem Cells

Steven W. Levison; Raymond P. Rothstein; Michael J. Romanko; Matthew J. Snyder; Roland L. Meyers; Susan J. Vannucci

Cerebral hypoxia/ischemia of the newborn has a frequency of 4/1,000 births and remains a major cause of cerebral palsy, epilepsy, and mental retardation. Despite progress in understanding the pathogenesis of hypoxic-ischemic injury, the data are incomplete regarding the mechanisms leading to permanent brain injury. Here we tested the hypothesis that cerebral hypoxia/ischemia damages stem/progenitor cells in the subventricular zone (SVZ), resulting in a permanent depletion of oligodendrocytes. We used a widely accepted rat model and examined animals at recovery intervals ranging from 4 h to 3 weeks. Within hours after the hypoxic-ischemic insult 20% of the total cells were deleted from the SVZ. The residual damaged cells appeared necrotic. During 48 h of recovery deaths accumulated; however, these later deaths were predominantly apoptotic. Many apoptotic SVZ cells stained with a marker for immature oligodendrocytes. At 3 weeks survival, the SVZ was smaller and markedly less cellular, and it contained less than 1/4 the normal complement of neural stem cells. The corresponding subcortical white matter was dysmyelinated, relatively devoid of oligodendrocytes and enriched in astrocytes. We conclude that neural stem cells and oligodendrocyte progenitors in the SVZ are vulnerable to hypoxia/ischemia. Consequently, the developmental production of oligodendrocytes is compromised and regeneration of damaged white matter oligodendrocytes does not occur resulting in failed regeneration of CNS myelin in periventricular loci. The resulting dysgenesis of the brain that occurs subsequent to perinatal hypoxic/ischemic injury may contribute to the cognitive and motor dysfunction that results from asphyxia of the newborn.


The Journal of Neuroscience | 2006

Neural Stem/Progenitor Cells Participate in the Regenerative Response to Perinatal Hypoxia/Ischemia

Ryan J. Felling; Matthew J. Snyder; Michael J. Romanko; Raymond P. Rothstein; Amber N. Ziegler; Zhengang Yang; Maria I. Givogri; Ernesto R. Bongarzone; Steven W. Levison

Perinatal hypoxia/ischemia (H/I) is the leading cause of neurologic injury resulting from birth complications. Recent advances in critical care have dramatically improved the survival rate of infants suffering this insult, but ∼50% of survivors will develop neurologic sequelae such as cerebral palsy, epilepsy or cognitive deficits. Here we demonstrate that tripotential neural stem/progenitor cells (NSPs) participate in the regenerative response to perinatal H/I as their numbers increase 100% by 3 d and that they alter their intrinsic properties to divide using expansive symmetrical cell divisions. We further show that production of new striatal neurons follows the expansion of NSPs. Increased proliferation within the NSP niche occurs at 2 d after perinatal H/I, and the proliferating cells express nestin. Of those stem-cell related genes that change, the membrane receptors Notch1, gp-130, and the epidermal growth factor receptor, as well as the downstream transcription factor Hes5, which stimulate NSP proliferation and regulate stem cellness are induced before NSP expansion. The mechanisms for the reactive expansion of the NSPs reported here reveal potential therapeutic targets that could be exploited to amplify this response, thus enabling endogenous precursors to restore a normal pattern of brain development after perinatal H/I.


Progress in Neurobiology | 2003

Roles of the mammalian subventricular zone in brain development

Christine Y. Brazel; Michael J. Romanko; Raymond P. Rothstein; Steven W. Levison

There has been enormous progress in uncovering the contributions of the subventricular zone (SVZ) to the developing brain. Here, we review the roles of four anatomically defined embryologic divisions of the SVZ of the mammalian brain: the lateral ganglionic eminence (LGE), the medial ganglionic eminence (MGE), the caudal ganglionic eminence (CGE), and the fetal neocortical SVZ (SVZn), as well as the roles of the two major anatomically defined regions of the postnatal SVZ, the anterior SVZ (SVZa) and the dorsolateral SVZ (SVZdl). We describe the types of cells within each subdivision of the SVZ, the types of brain cells that they generate during embryonic, fetal, and perinatal development, and when known the mechanisms that regulate their differentiation. This review provides a critical analysis of the literature, from which current and future studies on the SVZ can be formulated and evaluated.


Developmental Neuroscience | 2001

Perinatal Hypoxia-Ischemia Induces Apoptotic and Excitotoxic Death of Periventricular White Matter Oligodendrocyte Progenitors

Jennifer K. Ness; Michael J. Romanko; Raymond P. Rothstein; Teresa L. Wood; Steven W. Levison

Hypoxia-ischemia (HI) is a leading cause of white matter damage, a major contributor to cerebral palsy in premature infants. Preferential white matter damage is believed to result from vulnerability of the immature oligodendrocyte (the pro-OL) to factors elevated during ischemic damage, such as oxygen free radicals and glutamate. In order to determine whether pro-OLs undergo apoptotic death after HI, we analyzed periventricular white matter OLs in P7 rats 4, 12 and 24 h after HI to analyze the time course and mode of cell death. DNA fragmentation was seen at 12 and 24 h of recovery after HI, representing a 17-fold increase over control. In addition, caspase-3 activation was found in NG2+ pro-OLs at 12 h. Electron-microscopic analysis of cell death in the white matter revealed a transition from early necrotic deaths to hybrid cell deaths to classical apoptosis between 4 and 24 h of recovery from HI. The delayed time course of apoptosis in pro-OLs supports the feasibility of interventions to improve clinical outcomes for newborns surviving birth asphyxia.


Journal of Cerebral Blood Flow and Metabolism | 2004

Neural stem cells in the subventricular zone are resilient to hypoxia/ischemia whereas progenitors are vulnerable.

Michael J. Romanko; Raymond P. Rothstein; Steven W. Levison

Perinatal hypoxic-ischemic (H/I) brain injury remains a major cause of neurologic disability. Because we have previously demonstrated that this insult depletes cells from the subventricular zone (SVZ), the goal of the present investigation was to compare the relative vulnerability to H/I of neural stem cells versus progenitors. The dorsolateral SVZs of P6 rats were examined at 2 to 48 hours of recovery from H/I using hematoxylin and eosin, in situ end labeling (ISEL), terminal deoxynucleotidyl transferase-mediated 2′-deoxyuridine 5′-triphosphate-biotin nick end labeling (TUNEL), electron microscopy, and immunofluorescence. Pyknotic nuclei and ISEL+ cells were observed by 4 hours of recovery, peaked at 12 hours, and persisted for at least 48 hours. Many active-caspase3+ cells were observed at 12 hours and they comprised one third of the total TUNEL+ population. Electron microscopy revealed that hybrid cell deaths predominated at 12 hours of recovery. Importantly, few dying cells were observed in the medial SVZ, where putative stem cells reside, and no nestin+ medial SVZ cells showed caspase-3 activation. By contrast, active-caspase-3+/PSA-NCAM+ progenitors were prominent in the lateral SVZ. These data demonstrate that early progenitors are vulnerable to H/I, whereas neural stem cells are resilient. The demise of these early progenitors may lead to the depletion of neuronal and late oligodendrocyte progenitors, contributing to cerebral dysgenesis after perinatal insults.


Brain Research | 2012

Different routes of administration of human umbilical tissue-derived cells improve functional recovery in the rat after focal cerebral ischemia

Li Zhang; Yi Li; Michael J. Romanko; Brian C. Kramer; Anna Gosiewska; Michael Chopp; Klaudyne Hong

Human umbilical tissue-derived cells (hUTC) are a potential neurorestorative candidate for stroke treatment. Here, we test the effects of hUTC treatment in a rat model of stroke via various routes of administration. Rats were treated with hUTC or phosphate-buffered saline (PBS) via different routes including intraarterial (IA), intravenous (IV), intra-cisterna magna (ICM), lumber intrathecal (IT), or intracerebral injection (IC) at 24h after stroke onset. Treatment with hUTC via IV and IC route led to significant functional improvements starting at day 14, which persisted to day 60 compared with respective PBS-treated rats. HUTC administered via IA, ICM, and IT significantly improved neurological functional recovery starting at day 14 and persisted up to day 49 compared with PBS-treated rats. Although IA administration resulted in the highest donor cell number detected within the ischemic brain compared to the other routes, hUTC treatments significantly increased ipsilateral bromodeoxyuridine incorporating subventricular zone (SVZ) cells and vascular density in the ischemic boundary compared with PBS-treated rats regardless of the route of administration. While rats received hUTC treatment via IA, IV, IC, and ICM routes showed greater synaptophysin immunoreactivity, significant reductions in TUNEL-positive cells in the ipsilateral hemisphere were observed in IA, IV, and IC routes compared with PBS-treated rats. hUTC treatments did not reduce infarct volume when compared to the PBS groups. Our data indicate that hUTC administered via multiple routes provide therapeutic benefit after stroke. The enhancement of neurorestorative events in the host brain may contribute to the therapeutic benefits of hUTC in the treatment of stroke.


Journal of Neurochemistry | 2007

Death effector activation in the subventricular zone subsequent to perinatal hypoxia/ischemia

Michael J. Romanko; Changlian Zhu; Ben A. Bahr; Klas Blomgren; Steven W. Levison

Perinatal hypoxia/ischemia (H/I) is the leading cause of neurological injury resulting from birth complications and pre‐maturity. Our studies have demonstrated that this injury depletes the subventricular zone (SVZ) of progenitors. In this study, we sought to reveal which cell death pathways are activated within these progenitors after H/I. We found that calpain activity is detected as early as 4 h of reperfusion and is sustained for 48 h, while caspase 3 activation does not occur until 8 h and peaks at 24 h post‐insult. Activated calpains and caspase 3 co‐localized within precursors situated in the lateral aspects of the SVZ (which coincides with progenitor cell death), whereas neither enzyme was activated in the medial SVZ (which harbors the neural stem cells that are resilient to this insult). These studies reveal targets for neuroprotective agents to protect precursors from cell death towards the goal of restoring normal brain development after H/I.


Cell Transplantation | 2013

Intravenous administration of human umbilical tissue-derived cells improves neurological function in aged rats after embolic stroke.

Li Zhang; Li Yi; Michael Chopp; Brian C. Kramer; Michael J. Romanko; Anna Gosiewska; Klaudyne Hong

Intravenous administration of human umbilical tissue-derived cells (hUTC) improves neurological function in young adult rats after stroke. However, stroke is a major cause of death and disability in the aged population, with the majority of stroke patients 65 years and older. The present study investigated the effect of hUTC on aged rats after embolic stroke. Rats at the age of 18–20 months were subjected to embolic middle cerebral artery (MCA) occlusion. Two groups of eight animals each were compared. The investigational group was injected intravenously with 1 × 107 cells/kg in serum-free culture medium (vehicle) 24 h after stroke onset, and the control group was treated with vehicle only at the same time poststroke. Intravenous administration of hUTC significantly improved neurological functional recovery without reducing infarct volume compared to vehicle-treated aged rats. Additionally, hUTC treatment significantly enhanced synaptogenesis and vessel density in the ischemic boundary zone (IBZ). Moreover, hUTC treatment resulted in a trend toward increased progenitor cell proliferation in the subventricular zone (SVZ) compared to vehicle-treated aged rats. Intravenous administration of hUTC improved functional recovery in aged rats after stroke. The enhancement of synaptogenesis and vessel density may contribute to the beneficial effects of hUTC in the treatment of stroke in the aged animal.


Archive | 2006

Responses of the SVZ to Hypoxia and Hypoxia/Ischemia

Ryan J. Felling; H. VanGuider; Michael J. Romanko; Steven W. Levison

Stroke is a devastating injury caused by interruption of the blood supply to the brain. Each year, about 700,000 people in the United States suffer a stroke incurring healthcare costs of more than


Progress in Neurobiology | 2004

Roles of the mammalian subventricular zone in cell replacement after brain injury

Michael J. Romanko; Radoslaw Rola; John R. Fike; Francis G. Szele; Maria L.V. Dizon; Ryan J. Felling; Christine Y. Brazel; Steven W. Levison

50 billion (Heart Disease and Stroke Statistics 2003 Update, 2002). While advances in the acute treatment of stroke have improved the survival rates, little success has been realized in decreasing the associated morbidity. Stroke survivors suffer a wide range of neurological deficits depending on the location in the brain where the stroke occurs. These effects include, but are not limited to, paralysis, sensory deficits, memory loss and personality changes. Additionally, hypoxic/ischemic (H/I) injury is the single most important cause of brain damage resulting from complications during birth, leading to permanent neurological deficits. Every year, perinatal H/I afflicts approximately 1-2 per 1000 term births and roughly half of surviving preterm infants. Many of these infants suffer long-term handicaps that include learning disabilities, mental retardation, epilepsy and cerebral palsy (Volpe, 2001). Formore than 20 years, experimental animal models have beenwidely used to study the effects of ischemic brain injury. The most common model is transient middle cerebral artery occlusion (MCAo) used primarily in adult animals to generate unilateral focal cerebral infarcts (Longa et al., 1989).

Collaboration


Dive into the Michael J. Romanko's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Raymond P. Rothstein

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ryan J. Felling

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Christine Y. Brazel

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Jennifer K. Ness

Pennsylvania State University

View shared research outputs
Researchain Logo
Decentralizing Knowledge