Network


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

Hotspot


Dive into the research topics where Jennifer K. Newcomb is active.

Publication


Featured researches published by Jennifer K. Newcomb.


Neuroreport | 1998

Regional calpain and caspase-3 proteolysis of α-spectrin after traumatic brain injury

Brian R. Pike; Xiurong Zhao; Jennifer K. Newcomb; Rand Posmantur; Kevin K. W. Wang; R.L. Hayes

ACTIVITY of calpains and caspase-3 inferred from proteolysis of the cytoskeletal protein α-spectrin into signature spectrin breakdown products (SBDPs) was used to provide the first systematic and simultaneous comparison of changes in activity of these two families of cysteine proteases after traumatic brain injury (TBI) in rats. Distinct regional and temporal patterns of calpain/caspase-3 processing of α-spectrin were observed in brain regions ipsilateral to the site of injury after TBI, including large increases of 145 kDa calpain-mediated SBDP in cortex (up to 30-fold), and enduring increases (up to 2 weeks) of 145 kDa SBDP in hippocampus and thalamus. By contrast, 120 kDa caspase-3-mediated SBDP was absent in cortex and showed up to a 2-fold increase in hippocampus and striatum at early (hours) after TBI. Future studies will clarify the pathological significance of large regional differences in activation of calpain and caspase-3 proteases after TBI.


Experimental Neurology | 1999

Temporal Profile of Apoptotic-like Changes in Neurons and Astrocytes Following Controlled Cortical Impact Injury in the Rat

Jennifer K. Newcomb; Xiurong Zhao; Brian R. Pike; R.L. Hayes

Apoptotic cell death has been observed in both neurodegenerative diseases and acute neurological traumas such as ischemia, spinal cord injury, and traumatic brain injury (TBI). Recent studies employing different models of TBI have described morphological and biochemical changes characteristic of apoptosis following injury. However, no study has examined the temporal profile of apoptosis following controlled cortical impact (CCI) injury in the rat. In addition, the relative frequency of apoptotic profiles in different cell types (neurons versus glia) following CCI has yet to be investigated. In the present experiments, injured cortex was subjected to DNA electrophoresis, and serial sections from the contusion area were stained with hematoxylin and eosin or Hoechst 33258 or double-labeled with TUNEL and neuronal or glial markers. The results of the present study indicate that CCI produces a substantial amount of DNA damage associated with both apoptotic-like and necrotic-like cell death phenotypes primarily at the site of cortical impact and focal contusion. DNA damage, as measured by TUNEL and DNA electrophoresis, was most apparent 1 day following injury and absent by 14 days post-TBI. However, quantitative analysis showed that the majority of TUNEL-positive cells failed to exhibit apoptotic-like morphology and were probably undergoing necrosis. In addition, apoptotic-like morphology was predominantly observed in neurons compared to astrocytes. The present study provides further evidence that apoptosis is involved in the pathology of TBI and could contribute to some of the ensuing cell death following injury.


Journal of Neurochemistry | 2002

μ-Calpain activation and calpain-mediated cytoskeletal proteolysis following traumatic brain injury

Andreas Kampfl; R. Posmantur; Ralph A. Nixon; Frida Grynspan; Xiurong Zhao; Shi Jie Liu; Jennifer K. Newcomb; Guy L. Clifton; R.L. Hayes

Abstract: Increasing evidence suggests that excessive activation of the calcium‐activated neutral protease μ‐calpain could play a major role in calcium‐mediated neuronal degeneration after acute brain injuries. To further investigate the changes of the in vivo activity of μ‐calpain after unilateral cortical impact injury in vivo, the ratio of the 76‐kDa activated isoform of μ‐calpain to its 80‐kDa precursor was measured by western blotting. This μ‐calpain activation ratio increased to threefold in the pellet of cortical samples ipsilateral to the injury site at 15 min, 1 h, 3 h, and 6 h after injury and returned to control levels at 24–48 h after injury. We also investigated the effect of μ‐calpain activation on proteolysis of the neuronal cytoskeletal protein α‐spectrin. Immunoreactivity for α‐spectrin breakdown products was detectable within 15 min after injury in cortical samples ipsilateral to the injury site. The levels of α‐spectrin breakdown products increased in a biphasic manner, with a large increase between 15 min and 6 h after injury, followed by a smaller increase between 6 and 24 h after the insult. No further accumulation of α‐spectrin breakdown products was observed between 24 and 48 h after injury. Histopathological examinations using hematoxylin and eosin staining demonstrated dark, shrunken neurons within 15 min after traumatic brain injury. No evidence of μ‐calpain autolysis, calpain‐mediated α‐spectrin degradation, or hematoxylin and eosin neuronal pathology was detected in the contralateral cortex. Although μ‐calpain autolysis and cytoskeletal proteolysis occurred concurrently with early morphological alterations, evidence of calpain‐mediated proteolysis preceded the full expression of evolutionary histopathological changes. Our results indicate that rapid and persistent μ‐calpain activation plays an important role in cortical neuronal degeneration after traumatic brain injury. Our data also suggest that specific inhibitors of calpain could be potential therapeutic agents for the treatment of traumatic brain injury in vivo.


Journal of Neuroscience Research | 1998

Temporal relationships between de novo protein synthesis, calpain and caspase 3-like protease activation, and DNA fragmentation during apoptosis in septo-hippocampal cultures

Brian R. Pike; Xiurong Zhao; Jennifer K. Newcomb; Kevin K. W. Wang; Rand Posmantur; R.L. Hayes

Caspase 3‐like proteases are key executioners in mammalian apoptosis, and the calpain family of cysteine proteases has also been implicated as an effector of the apoptotic cascade. However, the influence of upstream events on calpain/caspase activation and the role of calpain/caspase activation on subsequent downstream events are poorly understood. This investigation examined the temporal profile of apoptosis‐related events after staurosporine‐induced apoptosis in mixed glial‐neuronal septo‐hippocampal cell cultures. Following 3 hr exposure to staurosporine (0.5 μM), calpain and caspase 3‐like proteases processed α‐spectrin to their signature proteolytic fragments prior to endonuclease‐mediated DNA fragmentation (not evident until 6 hr), indicating that endonuclease activation is downstream from calpain/caspase activation. Cycloheximide, a general protein synthesis inhibitor, completely prevented processing of α‐spectrin by calpains and caspase 3‐like proteases, DNA fragmentation and cell death, indicating that de novo protein synthesis is an upstream event necessary for activation of calpains and caspase 3‐like proteases. Calpain inhibitor II and the pan‐caspase inhibitor Z‐D‐DCB each inhibited their respective protease‐specific processing of α‐spectrin and attenuated endonuclease DNA fragmentation and cell death. Thus, activation of calpains and caspase 3‐like proteases is an early event in staurosporine‐induced apoptosis, and synthesis of, as yet, unknown protein(s) is necessary for their activation. J. Neurosci. Res. 52:505–520, 1998.


Neurochemical Research | 1999

Maitotoxin induces calpain but not caspase-3 activation and necrotic cell death in primary septo-hippocampal cultures

Xiurong Zhao; Brian R. Pike; Jennifer K. Newcomb; Kevin K. W. Wang; R.M. Posmantur; R.L. Hayes

Maitotoxin is a potent toxin that activates voltage and receptor-mediated Ca2+ channels, resulting in Ca2+ overload and rapid cell death. We report that maitotoxin-induced cell death is associated with activation of calpain but not caspase-3 proteases in septo-hippocampal cell cultures. Calpain and caspase-3 activation were examined by accumulation of protease-specific breakdown products to α-spectrin. Cell death manifested exclusively necrotic-like characteristics including round, shrunken nuclei, even distribution of chromatin, absence of DNA fragmentation and failure of protein synthesis inhibition to reduce cell death. Necrotic cell death was observed in neurons and astroglia. Calpain inhibitor II inhibited calpain-specific processing of α-spectrin and significantly reduced cell death. The pan-caspase inhibitor, Z-D-DCB, nominally attenuated cell death. Results suggest that: (1) calpain, but not caspase-3, is activated as a result of maitotoxin-induced Ca2+ influx; (2) necrotic cell death caused by maitotoxin exposure is partially mediated by calpain activation; (3) maitotoxin is a useful tool to investigate pathological mechanisms of necrosis.


Experimental Neurology | 2000

Light and confocal microscopic studies of evolutionary changes in neurofilament proteins following cortical impact injury in the rat

R.M. Posmantur; Jennifer K. Newcomb; A. Kampfl; Ronald L. Hayes

Previous studies have shown that traumatic brain injury (TBI) produces progressive degradation of cytoskeletal proteins including neurofilaments (e.g., neurofilament 68 [NF68] and neurofilament 200 [NF200]) within the first 24 h after injury. Thus, we employed immunofluorescence (light and confocal microscopy) to study the histopathological correlates of progressive neurofilament protein loss observed at 15 min, 3 h, and 24 h following unilateral cortical injury in rats. TBI produced significant alterations in NF68 and NF200 immunolabeling in dendrites and cell bodies at contusion sites ipsilateral to injury, as well as in the noncontused contralateral cortex. Changes in immunolabeling were associated with, but not exclusively restricted to, regions previously shown to contain dark shrunken neurons labeled by hematoxylin and eosin staining, a morphopathological response to injury suggesting impending cell death. Immunofluorescence microscopic studies of neurofilament proteins in the ipsilateral cerebral cortex detected prominent fragmentation of apical dendrites of pyramidal neurons in layers 3-5 and loss of fine dendritic arborization within layer 1. While modest changes were observed 15 min following injury, more pronounced loss of dendritic neurofilament immunofluorescence was detected 3 and 24 h following injury. Confocal microscopy also revealed progressive alterations in NF68 immunoreactivity in dendrites following TBI. While some evidence of structural alterations was observed 15 min following TBI, dendritic breaks were readily detected in confocal micrographs from 3 to 24 h following injury. However, disturbances in axonal NF68 by immunofluorescence microscopy in the corpus callosum were not detected until 24 h after injury. These studies confirmed that derangements in dendritic neurofilament cytoskeletal proteins are not exclusively restricted to sites of impact contusion. Moreover, changes in dendritic cytoskeletal proteins are progressive and not fully expressed within the first 15 min following impact injury. These progressive dendritic disruptions are characterized by disturbances in the morphology of neurofilament proteins, resulting in fragmentation and focal loss of NF68 immunofluorescence within apical dendrites. In contrast, alterations in axonal cytoskeletal proteins are more restricted and delayed with no pronounced changes until 24 h after injury.


Journal of Cerebral Blood Flow and Metabolism | 1998

Subcellular localization and duration of μ-calpain and m-calpain activity after traumatic brain injury in the rat : A casein zymography study

Xiurong Zhao; Rand Posmantur; A. Kampfl; Shi Jie Liu; Kevin K. W. Wang; Jennifer K. Newcomb; Brian R. Pike; Guy L. Clifton; R.L. Hayes

Casein zymographic assays were performed to identify changes in μ-calpain and m-calpain activity in naive, sham-injured, and injured rat cortex at 15 minutes, 3 hours, 6 hours, and 24 hours after unilateral cortical impact brain injury. Cortical samples ipsilateral and contralateral to the site of injury were separated into cytosolic and total membrane fractions. Marked increases in μ-calpain activity in cytosolic fractions in the ipsilateral cortex occurred as early as 15 minutes, became maximal at 6 hours, and decreased at 24 hours to levels observed at 15 minutes after injury. A similar temporal profile of cytosolic μ-calpain activity in the contralateral cortex was observed, although the increases in the contralateral cortex were substantially lower than those in the ipsilateral cortex. Differences were also noted between cytosolic and total membrane fractions. The detection of a shift in μ-calpain activity to the total membrane fraction first occurred at 3 hours after traumatic brain injury and became maximal at 24 hours after traumatic brain injury. This shift in μ-calpain activity between the two fractions could be due to the redistribution of μ-calpain from the cytosol to the membrane. m-Calpain activity was detected only in cytosolic fractions. m-Calpain activity in cytosolic fractions did not differ significantly between ipsilateral and contralateral cortices, and increased in both cortices from 15 minutes to 6 hours after injury. Relative magnitudes of m-calpain versus μ-calpain activity in cytosolic fractions differed at different time points after injury. These studies suggest that traumatic brain injury can activate both calpain isoforms and that calpain activity is not restricted to sites of focal contusion and cell death at the site of impact injury but may represent a more global response to injury.


Neuroscience Letters | 1998

pH dependency of μ-calpain and m-calpain activity assayed by casein zymography following traumatic brain injury in the rat

Xiurong Zhao; Jennifer K. Newcomb; Rand Posmantur; Kevin K. W. Wang; Brian R. Pike; R.L. Hayes

Studies employing casein zymographic assays analyzed the effects of varying pH (from pH 6.8 to pH 8.0) on changes in μ-calpain and m-calpain activity in naive, sham-injured and injured rat cortex 3 h following unilateral cortical impact injury. μ-Calpain activity following cortical impact injury was enhanced between pH values of 7.2 and 7.8, with pH 7.5 being optimal. m-Calpain activity was readily detected only between pH values of 7.2 and 7.4, with pH 7.3 producing the most prominent proteolytic activity. These observations suggest that strict control of pH is an important consideration in assessments of brain pH activation by casein zymography. Moreover, activation of different calpain isoforms, especially after traumatic brain injury, may be differentially influenced by smaller changes in physiological pH than previously recognized.


Journal of Cerebral Blood Flow and Metabolism | 2000

Novel characteristics of glutamate-induced cell death in primary septohippocampal cultures: Relationship to calpain and caspase-3 protease activation

Xiurong Zhao; Jennifer K. Newcomb; Brian R. Pike; Kevin K. W. Wang; Domenico d'Avella; Ronald L. Hayes

Studies examined the phenotypic characteristics of glutamate-induced cell death and their relationship to calpain and caspase-3 activation. Cell viability was assessed by fluorescein diacetate and propidium iodide staining and lactate dehydrogenase release. Calpain and caspase-3 activity was inferred from signature proteolytic fragmentation of α-spectrin. Characterization of cell death phenotypes was assessed by Hoechst 33258 and DNA fragmentation assays. Exposure of septohippocampal cultures to 1.0, 2.0, and 4.0 mmol/L glutamate induced a dose-dependent cell death with an LD50 of 2.0 mmol/L glutamate after 24 hours of incubation. Glutamate treatment induced cell death in neurons and astroglia and produced morphological alterations that differed from necrotic or apoptotic changes observed after maitotoxin or staurosporine exposure, respectively. After glutamate treatment, cell nuclei were enlarged and eccentrically shaped, and aggregated chromatin appeared in a diffusely speckled pattern. Furthermore, no dose of glutamate produced evidence of internucleosomal DNA fragmentation. Incubation with varying doses of glutamate produced calpain and caspase-3 activation. Calpain inhibitor II (N-acetyl-Leu-Leu-methionyl) provided protection only with a narrow dose range, whereas carbobenzoxy-Asp-CH2-OC(O)-2, 6-dichlorobenzene (Z-D-DCB; pan-caspase inhibitor) and MK-801 (N-methyl-d-aspartate receptor antagonist) were potently effective across a wider dose range. Cycloheximide did not reduce cell death or protease activation.


Journal of Neurotrauma | 1997

Immunohistochemical Study of Calpain-Mediated Breakdown Products to α-Spectrin Following Controlled Cortical Impact Injury in the Rat

Jennifer K. Newcomb; Andreas Kampfl; Rand Posmantur; Xiurong Zhao; Brian R. Pike; Shi Jie Liu; Guy L. Clifton; Ronald L. Hayes

Collaboration


Dive into the Jennifer K. Newcomb's collaboration.

Top Co-Authors

Avatar

Xiurong Zhao

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

R.L. Hayes

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ronald L. Hayes

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Andreas Kampfl

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Guy L. Clifton

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Shi Jie Liu

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

A. Kampfl

University of Innsbruck

View shared research outputs
Researchain Logo
Decentralizing Knowledge