BreakThru Your Health

Traumatic brain injury (TBI)

Traumatic brain injury (TBI)

Original article

The cysteine-rich whey protein supplement, Immunocal®, preserves brain glutathione and improves cognitive, motor, and histopathological indices of traumatic brain injury in a mouse model of controlled cortical impact

Author links open overlay panelElizabeth Ignowski a 1, Aimee N. Winter a 1, Nathan Duval b, Holly Fleming b, Tyler Wallace a, Evan Manning a, Lilia Koza a, Kendra Huber c, Natalie J. Serkova c, Daniel A. Linseman d

Show more

Add to Mendeley

Share

Cite

https://doi.org/10.1016/j.freeradbiomed.2018.06.026Get rights and content

Highlights

Immunocal® significantly enhanced resilience to TBI induced by closed head injury in mice.

Immunocal® preserved brain GSH/GSSG and attenuated demyelination and neuronal degeneration.

Most significantly, Immunocal® improved motor and cognitive deficits induced by TBI.

Immunocal® holds significant promise as a preventative agent for TBI.

Abstract

Traumatic brain injury (TBI) is a major public health problem estimated to affect nearly 1.7 million people in the United States annually. Due to the often debilitating effects of TBI, novel preventative agents are highly desirable for at risk populations. Here, we tested a whey protein supplement, Immunocal®, for its potential to enhance resilience to TBI. Immunocal® is a non-denatured whey protein preparation which has been shown to act as a cysteine delivery system to increase levels of the essential antioxidant glutathione (GSH). Twice daily oral supplementation of CD1 mice with Immunocal® for 28 days prior to receiving a moderate TBI prevented an ~ 25% reduction in brain GSH/GSSG observed in untreated TBI mice. Immunocal® had no significant effect on the primary mechanical injury induced by TBI, as assessed by MRI, changes in Tau phosphorylation, and righting reflex time or apnea. However, pre-injury supplementation with Immunocal® resulted in statistically significant improvements in motor function (beam walk and rotarod) and cognitive function (Barnes maze). We also observed a significant preservation of corpus callosum width (axonal myelination), a significant decrease in degenerating neurons, a reduction in Iba1 (microglial marker), decreased lipid peroxidation, and preservation of brain-derived neurotrophic factor (BDNF) in the brains of Immunocal®-pretreated mice compared to untreated TBI mice. Taken together, these data indicate that pre-injury supplementation with Immunocal® significantly enhances the resilience to TBI induced by a moderate closed head injury in mice. We conclude that Immunocal® may hold significant promise as a preventative agent for TBI, particularly in certain high risk populations such as athletes and military personnel.

Graphical abstract

fx1

Access through your organization

Check access to the full text by signing in through your organization.

 Access through your institution

Introduction

Approximately 1.7 million incidences of TBI occur in the United States each year [12]. Of these cases, nearly 75% are categorized as forms of mild TBI including concussions. Chronic traumatic encephalopathy and other forms of dementia have been linked to repetitive mild TBI caused by sports related concussive and subconcussive head trauma in football, hockey, soccer, and wrestling [17], [21], [34], [9]. In a similar manner, blast-related TBI which is estimated to affect 10–20% of veterans returning from the wars in Iraq and Afghanistan, is also associated with an increased risk of chronic traumatic encephalopathy and other types of dementia, as well as posttraumatic stress disorder [11], [18]. Regardless of the cause or severity of TBI, even mild TBI appears to be a significant risk factor for development of dementia including Alzheimer’s disease [15], [16], [25], [29], [43]. Thus, identification of new strategies to enhance resilience against TBI is of particular importance to people participating in “high risk” occupations, such as athletes or military personnel.

Oxidative and nitrosative stress are key elements of the secondary injury processes following TBI [1], [4]. GSH is an essential antioxidant that works in concert with GSH peroxidases, GSH transferases, and peroxiredoxins to detoxify hydroperoxides and other electrophilic species produced during periods of oxidative and nitrosative stress. Several studies suggest that endogenous GSH plays an important protective role against TBI. Brain GSH levels are significantly reduced following TBI induced by controlled cortical impact in rats [47]. Genetic variations in the activity of glutathione-S-transferase-4, a GSH-dependent enzyme that reduces 4-hydroxynonenal, is a determining factor in the extent of neurodegeneration after TBI [2]. Moreover, mice homozygous for deletion of glutathione peroxidase-1 display enhanced susceptibility to brain mitochondrial dysfunction induced by TBI [50]. These findings suggest that a strategy aimed at sustaining or enhancing brain GSH levels may be a viable approach to mitigate secondary injury and the subsequent long term cognitive, physical, and emotional deficiencies induced by TBI.

Along these lines, several studies have shown that administration of the GSH precursor, N-acetylcysteine, just prior to or immediately after TBI, significantly preserved brain tissue and mitochondrial GSH levels, reduced oxidative damage, and preserved neuronal survival [24], [49]. Similarly, treatment with gamma-glutamylcysteine ethyl ester reduced indices of oxidative and nitrosative stress and preserved blood-brain-barrier (BBB) function when given immediately post-TBI [31], [40]. Finally, the GSH analog, S-nitrosoglutathione, decreased BBB disruption, minimized neuronal loss, and increased the expression of neurotrophic factors when administered post-TBI to rats subjected to controlled cortical impact [26], [27] Each of these results indicates the potential of enhancing GSH as a therapeutic approach for TBI. Unfortunately, few of these previous studies evaluated the effects of GSH precursor supplementation on cognitive or motor deficits induced by TBI and as a result, it is presently unclear what therapeutic benefit this strategy might realistically hold for patients suffering from TBI.

The nutritional supplement, Immunocal®, is a non-denatured whey protein designed to augment the available intracellular GSH pool. Cellular GSH concentrations are highly dependent on the availability of cysteine, which is the limiting precursor in GSH synthesis [36], [46]. The cysteine precursor, cystine, occurs at high levels in Immunocal®, as does the direct GSH precursor, glutamylcysteine [6], [7]. Immunocal® has been shown to significantly increase blood or lymphocyte GSH levels in HIV-seropositive or cystic fibrosis patients, respectively [8], [19]. Immunocal® is one of only a handful of nutritional supplements that are included in the Physician’s Desk Reference and is comprised of natural food protein placing it in the FDA category of generally recognized as safe [39]. We have recently found that Immunocal® preserves blood and spinal cord GSH levels and delays disease onset in a transgenic mouse model of amyotrophic lateral sclerosis [41]. Similarly, Immunocal® restored GSH homeostasis in the CNS and ameliorated behavioral deficits in a mouse model of schizophrenia [45]. Based on the above findings, we hypothesized that supplementation with Immunocal® prior to TBI in mice would provide enhanced resilience against oxidative damage, neuronal cell death, and cognitive and motor impairments induced by a closed head impact injury.

Section snippets

Animal care and treatment

All animal work was conducted under a protocol approved by the University of Denver Institutional Animal Care and Use Committee. Male CD1 Elite mice (35 days-old) were purchased from Charles River Laboratories (Hollister, CA). Mice received a numbered ear tag upon arrival for identification purposes, and then were allowed one week to acclimate to the animal facility at the University of Denver before beginning the study. Mice were then randomly assigned and evenly distributed among one of three 

Pre-injury supplementation with Immunocal® does not affect the primary mechanical injury induced by a moderate TBI

Throughout the study, mice were equally divided into the following three groups: Group 1, Sham surgery controls; Group 2, untreated TBI mice; and Group 3, mice pretreated with Immunocal® for 28 days prior to TBI. The extent of brain injury was initially assessed at 72 h post-TBI by MRI analysis of BBB disruption. In mice subjected to TBI, T1 weighted images taken with gadolinium contrast showed areas of hyper-intensity which indicate BBB disruption (Fig. 1A and B, see asterisk in the TBI image

Discussion

The pathophysiological processes underlying the short and long term injury sequelae associated with TBI are complex. The primary injury is mechanical, resulting from an external force, and leads to tissue deformation, tearing of blood vessels and neuronal axons, necrotic cell death, and initiation of secondary injury processes. Secondary injury mechanisms may include intracranial hemorrhage, excitotoxicity, ionic disturbances, decreased cerebral blood flow, edema, inflammation, mitochondrial

Acknowledgments

None.

Funding

This study was supported in large part by funding from Immunotec Inc. (Quebec, Canada), which is the manufacturer of Immunocal®. Funding was also provided by a pilot grant from the Knoebel Institute for Healthy Aging at the University of Denver. The University of Colorado Animal Imaging Shared Resources are supported by the University of Colorado Cancer Center (NCI P30 CA046934) and the Colorado Clinical and Translational Sciences Institute (NIH/NCATS UL1 TR001082).

Conflicts of interest

The authors have received

References (51)

  • M. Bains et al.

Antioxidant therapies in traumatic brain and spinal cord injury

Biochem. Biophys. Acta

(2012)

  • R.C. Gardner et al.

Epidemiology of mild traumatic brain injury and neurodegenerative disease

Mol. Cell Neurosci.

(2015)

  • B.E. Gavett et al.

Chronic traumatic encephalopathy: a potential late effect of sportrelated concussive and subconcussive head trauma

Clin. Sports Med.

(2011)

  • V. Grey et al.

Improved glutathione status in young adult patients with cystic fibrosis supplemented with whey protein

J. Cyst. Fibros.

(2003)

  • E.D. Hall et al.

Antioxidant therapies for traumatic brain injury

Neurotherapeutics

(2010)

  • A. Kumar et al.

Neuroinflammation after traumatic brain injury: opportunities for therapeutic intervention

Brain Behav. Immun.

(2012)

  • W. Song et al.

Cysteine-rich whey protein isolate (Immunocal(®)) ameliorates deficits in the GFAP.HMOX1 mouse model of schizophrenia

Free Radic. Biol. Med.

(2017)

  • Y. Xiong et al.

The protective role of cellular glutathione peroxidase against trauma-induced mitochondrial dysfunction in the mouse brain

J. Stroke Cerebrovasc. Dis.

(2004)

  • P.M. Abdul-Muneer et al.

Interactions of oxidative stress and neurovascular inflammation in the pathogenesis of traumatic brain injury

Mol. Neurobiol.

(2015)

  • F. Al Nimer et al.

Naturally occurring variation in the glutathione-s-transferase 4 gene determines neurodegeneration after traumatic brain injury

Antioxid. Redox Signal

(2013)

View more references

Cited by (25)

·         Immunocal® limits gliosis in mouse models of repetitive mild-moderate traumatic brain injury

2023, Brain Research

Show abstract

·         Early-life N-arachidonoyl-dopamine exposure increases antioxidant capacity of the brain tissues and reduces functional deficits after neonatal hypoxia in rats

2019, International Journal of Developmental Neuroscience

Citation Excerpt :

We may propose that NADAs’ ability to reduce oxidative stress in cells (Bobrov et al., 2008; Grabiec and Dehghani, 2017) resulted in decrease of basal ROS production and increased glutathione antioxidant buffer capacity. Previous findings on traumatic brain injury suggest that a strategy aimed at sustaining or enhancing brain GSH levels may be a viable approach to mitigate secondary injury and the subsequent long term cognitive, physical, and emotional deficiencies (Ignowski et al., 2018). The functional outcomes of the neonatal hypoxic injury in rodents are well documented and include an impaired development of sensorimotor reflexes.

Show abstract

·         Therapeutic effects of chrysin in a rat model of traumatic brain injury: A behavioral, biochemical, and histological study

2019, Life Sciences

Citation Excerpt :

Although many efforts have been done to develop effective treatments for TBI, there has been no definitive accepted treatment for the improvement of neurobehavioral deficits to date [8]. Oxidative stress plays a key role in the pathophysiology of the secondary phase of TBI, which can lead to neuronal apoptosis [9] and subsequently, may trigger the development of neurobehavioral deficits [10–12]. Superoxide radical (O2−), hydrogen peroxide (H2O2) and hydroxyl radical (HO−) are the main of reactive oxygen species (ROS), which leads to neuronal damage through protein oxidation, peroxidation of membrane lipids, and DNA destruction.

Show abstract

·         The effects of dexmedetomidine on trauma-induced secondary injury in rat brain

2023, Neurological Research

·         Wool keratin as a novel alternative protein: A comprehensive review of extraction, purification, nutrition, safety, and food applications

2023, Comprehensive Reviews in Food Science and Food Safety

·         Anti-aging trait of whey protein against brain damage of senile rats

2022, Journal of Umm Al-Qura University for Applied Sciences

Leave a Comment

Scroll to Top