1
|
Paterniti I, Cordaro M, Navarra M, Esposito E, Cuzzocrea S. Emerging pharmacotherapy for treatment of traumatic brain injury: targeting hypopituitarism and inflammation. Expert Opin Emerg Drugs 2015; 20:583-96. [PMID: 26087316 DOI: 10.1517/14728214.2015.1058358] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Traumatic brain injury (TBI) is a common cause of morbidity and mortality in the developed world. In particular, TBI is an important cause of death and disability in young adults with consequences ranging from physical disabilities to long-term cognitive, behavioural, psychological and social defects. AREAS COVERED There is a large body of evidence that suggest that TBI conditions may adversely affect pituitary function in both the acute and chronic phases of recovery. Prevalence of hypopituitarism, from total to isolated pituitary deficiency, ranges from 5 to 90%. The time interval between TBI and pituitary function evaluation is one of the major factors responsible for variations in the prevalence of hypopituitarism reported. Diagnosis of hypopituitarism and accurate treatment of pituitary disorders offers the opportunity to improve mortality and outcome in TBI conditions. EXPERT OPINION The aim of this paper is to review the history and pathophysiology of TBI and to summarize the best evidence of TBI as a cause of pituitary deficiency. Moreover, in this article we will describe the multiple changes which occur within the hypothalamic-pituitary-thyroid axis in critical illness, giving rise to 'sick euthyroid syndrome', focus our attention on thyroid hormones circulating levels from the initial insult to critical illness.
Collapse
Affiliation(s)
- Irene Paterniti
- a 1 University of Messina, Department of Biological and Environmental Sciences , Viale Ferdinando Stagno D'Alcontres, 31 - 98166 Messina, Italy +390906765208 ;
| | - Marika Cordaro
- b 2 University of Messina, Department of Biological and Environmental Sciences , Messina, Italy
| | - Michele Navarra
- c 3 University of Messina, Department of Drug Sciences and Health Products , Messina, Italy
| | - Emanuela Esposito
- a 1 University of Messina, Department of Biological and Environmental Sciences , Viale Ferdinando Stagno D'Alcontres, 31 - 98166 Messina, Italy +390906765208 ;
| | - Salvatore Cuzzocrea
- a 1 University of Messina, Department of Biological and Environmental Sciences , Viale Ferdinando Stagno D'Alcontres, 31 - 98166 Messina, Italy +390906765208 ; .,d 4 Saint Louis University School of Medicine, Department of Pharmacological and Physiological Science , USA
| |
Collapse
|
2
|
MAEGELE M, BRAUN M, WAFAISADE A, SCHÄFER N, LIPPERT-GRUENER M, KREIPKE C, RAFOLS J, SCHÄFER U, ANGELOV DN, STUERMER E. Long-Term Effects of Enriched Environment on Neurofunctional Outcome and CNS Lesion Volume After Traumatic Brain Injury in Rats. Physiol Res 2015; 64:129-45. [DOI: 10.33549/physiolres.932664] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
To determine whether the exposure to long term enriched environment (EE) would result in a continuous improvement of neurological recovery and ameliorate the loss of brain tissue after traumatic brain injury (TBI) vs. standard housing (SH). Male Sprague-Dawley rats (300-350 g, n=28) underwent lateral fluid percussion brain injury or SHAM operation. One TBI group was held under complex EE for 90 days, the other under SH. Neuromotor and sensorimotor dysfunction and recovery were assessed after injury and at days 7, 15, and 90 via Composite Neuroscore (NS), RotaRod test, and Barnes Circular Maze (BCM). Cortical tissue loss was assessed using serial brain sections. After day 7 EE animals showed similar latencies and errors as SHAM in the BCM. SH animals performed notably worse with differences still significant on day 90 (p<0.001). RotaRod test and NS revealed superior results for EE animals after day 7. The mean cortical volume was significantly higher in EE vs. SH animals (p=0.003). In summary, EE animals after lateral fluid percussion (LFP) brain injury performed significantly better than SH animals after 90 days of recovery. The window of opportunity may be wide and also lends further credibility to the importance of long term interventions in patients suffering from TBI.
Collapse
Affiliation(s)
- M. MAEGELE
- Department for Traumatology and Orthopedic Surgery, Cologne-Merheim Medical Center (CMMC), University Witten-Herdecke (Campus Cologne-Merheim), Cologne, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Abstract
Traumatic brain injury (TBI) is a major cause of mortality and morbidity worldwide. Despite extensive preclinical research supporting the effectiveness of neuroprotective therapies for brain trauma, there have been no successful randomized controlled clinical trials to date. TBI results in delayed secondary tissue injury due to neurochemical, metabolic and cellular changes; modulating such effects has provided the basis for neuroprotective interventions. To establish more effective neuroprotective treatments for TBI it is essential to better understand the complex cellular and molecular events that contribute to secondary injury. Here we critically review relevant research related to causes and modulation of delayed tissue damage, with particular emphasis on cell death mechanisms and post-traumatic neuroinflammation. We discuss the concept of utilizing multipotential drugs that target multiple secondary injury pathways, rather than more specific "laser"-targeted strategies that have uniformly failed in clinical trials. Moreover, we assess data supporting use of neuroprotective drugs that are currently being evaluated in human clinical trials for TBI, as well as promising emerging experimental multipotential drug treatment strategies. Finally, we describe key challenges and provide suggestions to improve the likelihood of successful clinical translation.
Collapse
Affiliation(s)
- David J Loane
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), National Study Center for Trauma and EMS, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bogdan A Stoica
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), National Study Center for Trauma and EMS, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alan I Faden
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), National Study Center for Trauma and EMS, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
4
|
Fei F, Rao W, Zhang L, Chen BG, Li J, Fei Z, Chen Z. Downregulation of Homer1b/c improves neuronal survival after traumatic neuronal injury. Neuroscience 2014; 267:187-94. [PMID: 24607348 DOI: 10.1016/j.neuroscience.2014.02.037] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 02/04/2014] [Accepted: 02/24/2014] [Indexed: 01/01/2023]
Abstract
Homer protein, a member of the post-synaptic density protein family, plays an important role in the neuronal synaptic activity and is extensively involved in neurological disorders. The present study investigates the role of Homer1b/c in modulating neuronal survival by using an in vitro traumatic neuronal injury model, which was achieved by using a punch device that consisted of 28 stainless steel blades joined together and produced 28 parallel cuts. Downregulation of Homer1b/c by specific siRNA significantly (p<0.05) alleviated the cytoplasmic calcium levels and neuron lactate dehydrogenase release, and ultimately decreased the apoptotic rate after traumatic neuronal injury compared with non-targeting siRNA control treatment in cultured rat cortical neurons. Moreover, the expression of metabotropic glutamate receptor 1a (mGluR1a) was significantly (p<0.05) reduced in the Homer1b/c siRNA-transfected neurons after injury. Therefore, Homer1b/c not only modulated the mGluR1a-inositol 1,4,5-triphosphate receptors-Ca(2+) signal transduction pathway, but also regulated the expression of mGluR1a in mechanical neuronal injury. These findings indicate that the suppression of Homer1b/c expression potentially protects neurons from glutamate excitotoxicity after injury and might be an effective intervention target in traumatic brain injury.
Collapse
Affiliation(s)
- F Fei
- Department of Cell Biology, College of Basic Medicine, Fourth Military Medical University, Xi'an 710032, PR China
| | - W Rao
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, PR China
| | - L Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, PR China
| | - B-G Chen
- Central Laboratory, Tongji University Affiliated Shanghai East Hospital, Shanghai 200120, PR China
| | - J Li
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, PR China
| | - Z Fei
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, PR China.
| | - Z Chen
- Department of Cell Biology, College of Basic Medicine, Fourth Military Medical University, Xi'an 710032, PR China.
| |
Collapse
|
5
|
Rahman A, Khan KM, Al-Khaledi G, Khan I, Al-Shemary T. Over activation of hippocampal serine/threonine protein phosphatases PP1 and PP2A is involved in lead-induced deficits in learning and memory in young rats. Neurotoxicology 2012; 33:370-83. [DOI: 10.1016/j.neuro.2012.02.014] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Revised: 02/15/2012] [Accepted: 02/21/2012] [Indexed: 11/27/2022]
|
6
|
Khomane KS, Meena CL, Jain R, Bansal AK. Novel thyrotropin-releasing hormone analogs: a patent review. Expert Opin Ther Pat 2012; 21:1673-91. [PMID: 22017410 DOI: 10.1517/13543776.2011.623127] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION The potential therapeutic applications of thyrotropin-releasing hormone (TRH) have attracted attention, based on its broad-spectrum neuropharmacological action rather than its endocrine properties. These central nervous system (CNS)-mediated effects provide the rationale for use of TRH and its analogs in the treatment of brain and spinal injury, and CNS disorders like schizophrenia, Alzheimer's disease, epilepsy, amyotrophic lateral sclerosis, Parkinson's disease, depression, shock and ischemia. AREAS COVERED This review summarizes the patent literature and advances in the discovery and development of novel TRH analogs over the past 20 years. It provides a comprehensive overview of the development of new TRH analogs, giving emphasis to their pharmaceutical profile. EXPERT OPINION The use of TRH in the treatment of various CNS disorders has been proven clinically. However, TRH itself is a poor drug candidate due to its short plasma half-life (5 min), poor biopharmaceutical properties (low intestinal and CNS permeability) and endocrine side effect. Nevertheless, researchers have come up with metabolically stable, more potent and selective TRH analogs and prodrugs. Taltirelin, one of the TRH analogs, has been approved under the trade name of Ceredist(®) in Japan for the treatment of spinocerebellar degeneration. Several other TRH analogs are in various stages of preclinical or clinical development.
Collapse
Affiliation(s)
- Kailas S Khomane
- National Institute of Pharmaceutical Education and Research (NIPER), Department of Pharmaceutics, Mohali, Punjab, India
| | | | | | | |
Collapse
|
7
|
Jaworska-Feil L, Jantas D, Leskiewicz M, Budziszewska B, Kubera M, Basta-Kaim A, Lipkowski AW, Lason W. Protective effects of TRH and its analogues against various cytotoxic agents in retinoic acid (RA)-differentiated human neuroblastoma SH-SY5Y cells. Neuropeptides 2010; 44:495-508. [PMID: 20869113 DOI: 10.1016/j.npep.2010.08.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2010] [Revised: 08/19/2010] [Accepted: 08/30/2010] [Indexed: 12/30/2022]
Abstract
TRH (thyroliberin) and its analogues were reported to possess neuroprotective effects in cellular and animal experimental models of acute and chronic neurodegenerative diseases. In the present study we evaluated effects of TRH and its three stable analogues, montirelin (CG-3703), RGH-2202 and Z-TRH (N-(carbobenzyloxy)-pGlutamyl-Histydyl-Proline) on the neuronally differentiated human neuroblastoma SH-SY5Y cell line, which is widely accepted for studying potential neuroprotectants. We found that TRH and all the tested analogues at concentrations 0.1-50 μM attenuated cell damage induced by MPP(+) (2 mM), 3-nitropropionate (10 mM), hydrogen peroxide (0.5 mM), homocysteine (250 μM) and beta-amyloid (20μM) in retinoic acid differentiated SH-SY5Y cells. Furthermore, we demonstrated that TRH and its analogues decreased the staurosporine (0.5 μM)-induced LDH release, caspase-3 activity and DNA fragmentation, which indicate the anti-apoptotic proprieties of these peptides. The neuroprotective effects of TRH (10 μM) and RGH-2202 (10 μM) on St-induced cell death was attenuated by inhibitors of PI3-K pathway (wortmannin and LY294002), but not MAPK/ERK1/2 (PD98059 and U0126). Moreover, TRH and its analogues at neuroprotective concentrations (1 and 10 μM) increased expression of Bcl-2 protein, as confirmed by Western blot analysis. All in all, these results extend data on neuroprotective properties of TRH and its analogues and provide evidence that mechanism of anti-apoptotic effects of these peptides in SH-SY5Y cell line involves induction of PI3K/Akt pathway and Bcl-2. Furthermore, the data obtained on human cell line with a dopaminergic phenotype suggest potential utility of TRH and its analogues in the treatment of some neurodegenerative diseases including Parkinson's disease.
Collapse
Affiliation(s)
- L Jaworska-Feil
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Abstract
SummaryThe physiological role of thyreoliberin (TRH) is the preservation of homeostasis within four systems (i) the hypothalamic-hypophsysiotropic neuroendocrine system, (ii) the brain stem/midbrain/spinal cord system, (iii) the limbic/cortical system, and (iv) the chronobiological system. Thus TRH, via various cellular mechanisms, regulates a wide range of biological processes (arousal, sleep, learning, locomotive activity, mood) and possesses the potential for unique and widespread applications for treatment of human illnesses. Since the therapeutic potential of TRH is limited by its pharmacological profile (enzymatic instability, short half-life, undesirable effects), several synthetic analogues of TRH were constructed and studied in mono- or adjunct therapy of central nervous system (CNS) disturbances. The present article summarizes the current state of understanding of the physiological role of TRH and describes its putative role in clinical indications in CNS maladies with a focus on the action of TRH analogues.
Collapse
|
9
|
Loane DJ, Faden AI. Neuroprotection for traumatic brain injury: translational challenges and emerging therapeutic strategies. Trends Pharmacol Sci 2010; 31:596-604. [PMID: 21035878 DOI: 10.1016/j.tips.2010.09.005] [Citation(s) in RCA: 434] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Revised: 09/25/2010] [Accepted: 09/27/2010] [Indexed: 11/30/2022]
Abstract
Traumatic brain injury (TBI) causes secondary biochemical changes that contribute to subsequent tissue damage and associated neuronal cell death. Neuroprotective treatments that limit secondary tissue loss and/or improve behavioral outcome have been well established in multiple animal models of TBI. However, translation of such neuroprotective strategies to human injury have been disappointing, with the failure of more than thirty controlled clinical trials. Both conceptual issues and methodological differences between preclinical and clinical injury have undoubtedly contributed to these translational difficulties. More recently, changes in experimental approach, as well as altered clinical trial methodologies, have raised cautious optimism regarding the outcomes of future clinical trials. Here we critically review developing experimental neuroprotective strategies that show promise, and we propose criteria for improving the probability of successful clinical translation.
Collapse
Affiliation(s)
- David J Loane
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), National Study Center for Trauma and Emergency Medical Systems, University of Maryland School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
10
|
Cernak I, Chang T, Ahmed FA, Cruz MI, Vink R, Stoica B, Faden AI. Pathophysiological response to experimental diffuse brain trauma differs as a function of developmental age. Dev Neurosci 2010; 32:442-53. [PMID: 20948187 DOI: 10.1159/000320085] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2010] [Accepted: 08/02/2010] [Indexed: 12/28/2022] Open
Abstract
The purpose of experimental models of traumatic brain injury (TBI) is to reproduce selected aspects of human head injury such as brain edema, contusion or concussion, and functional deficits, among others. As the immature brain may be particularly vulnerable to injury during critical periods of development, and pediatric TBI may cause neurobehavioral deficits, our aim was to develop and characterize as a function of developmental age a model of diffuse TBI (DTBI) with quantifiable functional deficits. We modified a DTBI rat model initially developed by us in adult animals to study the graded response to injury as a function of developmental age - 7-, 14- and 21-day-old rats compared to young adult (3-month-old) animals. Our model caused motor deficits that persisted even after the pups reached adulthood, as well as reduced cognitive performance 2 weeks after injury. Moreover, our model induced prominent edema often seen in pediatric TBI, particularly evident in 7- and 14-day-old animals, as measured by both the wet weight/dry weight method and diffusion-weighted MRI. Blood-brain barrier permeability, as measured by the Evans blue dye technique, peaked at 20 min after trauma in all age groups, with a second peak found only in adult animals at 24 h after injury. Phosphorus MR spectroscopy showed no significant changes in the brain energy metabolism of immature rats with moderate DTBI, in contrast to significant decreases previously identified in adult animals.
Collapse
Affiliation(s)
- Ibolja Cernak
- Department of Neuroscience, Georgetown University Medical Center, Washington, D.C., USA
| | | | | | | | | | | | | |
Collapse
|
11
|
Chauhan NB, Gatto R, Chauhan MB. Neuroanatomical correlation of behavioral deficits in the CCI model of TBI. J Neurosci Methods 2010; 190:1-9. [PMID: 20385166 DOI: 10.1016/j.jneumeth.2010.04.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2010] [Revised: 04/03/2010] [Accepted: 04/05/2010] [Indexed: 01/05/2023]
Abstract
Traumatic brain injury (TBI) is the leading cause of death and disability both in combat and civilian situations with limited treatment options including surgical removal of hematoma, ventricular drainage and use of hyperosmotic agents that restrict secondary injury following TBI. Availability of appropriate model system with full-range characterization of anatomical and behavioral components correlative with brain injury provides a pre-clinical platform to test candidate therapies for clinical translation. Modeling of TBI using controlled cortical impact injury (CCI) is largely considered to be close to clinical TBI and hence CCI models have been widely used in pre-clinical TBI research. Most studies reported so far using CCI models were presented with a limited behavioral characterization and lacked its correlation with the signature histopathology of TBI. Current investigation validated a detailed sensomotor and cognitive behavioral characterization correlative with diffuse axonal injury-the signature histopathology of TBI, in the CCI mouse model of TBI. Present study offers a comprehensively characterized model of TBI that can be used to investigate cellular and molecular mechanisms underlying TBI and to test candidate therapies in developing novel and effective treatments for TBI.
Collapse
Affiliation(s)
- Neelima B Chauhan
- Research & Development, Jesse Brown VA Medical Center Chicago, IL 60612, USA.
| | | | | |
Collapse
|
12
|
Jantas D, Jaworska-Feil L, Lipkowski AW, Lason W. Effects of TRH and its analogues on primary cortical neuronal cell damage induced by various excitotoxic, necrotic and apoptotic agents. Neuropeptides 2009; 43:371-85. [PMID: 19666192 DOI: 10.1016/j.npep.2009.07.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2009] [Revised: 07/17/2009] [Accepted: 07/18/2009] [Indexed: 11/17/2022]
Abstract
The tripeptide thyrotropin-releasing hormone (TRH, pGlu-His-Pro-NH2) has been shown to possess neuroprotective activity in in vitro and in vivo models. Since its potential utility is limited by relatively rapid metabolism, metabolically stabilized analogues have been constructed. In the present study we investigated the influence of TRH and its three stable analogues: Montirelin (MON, CG-3703), RGH-2202 (L-6-keto-piperidine-2carbonyl-l-leucyl-l-prolinamide) and Z-TRH (N-carbobenzyloxy-pGlutamyl-Histydyl-Proline) in various models of mouse cortical neuronal cell injury. Twenty four hour pre-treatment with TRH and its analogues in low micromolar concentrations attenuated the neuronal cell death evoked by excitatory amino acids (EAAs: glutamate, NMDA, kainate, quisqualate) and hydrogen peroxide. All the peptides showed neuroprotective action on staurosporine (St)-evoked apoptotic neuronal cell death, but this effect was caspase-3 independent. Interestingly, in mixed neuronal-glial cell preparations only MON decreased St- and glutamate-evoked neurotoxicity. None of the peptides inhibited the doxorubicin- and lactacystin-induced neuronal cortical cell death, agents acting via activation of death receptor (FAS) or inhibition of proteasome function, respectively. Furthermore, we found that neither inhibitors of PI3-K (wortmannin, LY 294002) nor MAPK/ERK1/2 (PD 098059, U 0126) were able to inhibit neuroprotective properties of TRH and MON in St model of apoptosis. The protection mediated by TRH and MON it that model was also not connected with influence of peptides on the pro-apoptotic GSK-3beta and JNK protein kinase expression and activity. Further studies showed that calpains, calcium-activated proteases were induced by Glu, but not by St in cortical neurons. Moreover, the Glu-evoked increase in spectrin alpha II cleavage product induced by calpains was blocked by TRH. The obtained data showed that the potency of TRH and its analogues in inhibiting EAAs- and H(2)O(2)-induced neuronal cell death from the highest to lowest activity was: MON>TRH>Z-TRH>RHG. Interestingly, all peptides were active against St-induced apoptosis, however, on concentration basis MON was far more potent than the other peptides. None of the peptides inhibited Dox- and LC-evoked apoptotic cell death. Additionally, the data exclude potential role of pro-survival (PI3-K/Akt and MAPK/ERK1/2) and pro-apoptotic (GSK-3beta and JNK) pathways in neuroprotective effects of TRH and its analogues on St-induced neuronal apoptosis. Moreover, the results point to involvement of the inhibition of calpains in the TRH neuroprotective effect in Glu model of neuronal cell death.
Collapse
Affiliation(s)
- D Jantas
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Smetna 12 Street, PL 31-343 Krakow, Poland.
| | | | | | | |
Collapse
|
13
|
Abstract
Although the concepts of secondary injury and neuroprotection after neurotrauma are experimentally well supported, clinical trials of neuroprotective agents in traumatic brain injury or spinal cord injury have been disappointing. Most strategies to date have used drugs directed toward a single pathophysiological mechanism that contributes to early necrotic cell death. Given these failures, recent research has increasingly focused on multifunctional (i.e., multipotential, pluripotential) agents that target multiple injury mechanisms, particularly those that occur later after the insult. Here we review two such approaches that show particular promise in experimental neurotrauma: cell cycle inhibitors and small cyclized peptides. Both show extended therapeutic windows for treatment and appear to share at least one important target.
Collapse
Affiliation(s)
- Bogdan Stoica
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC 20057, USA.
| | | | | |
Collapse
|
14
|
Cengiz SL, Ustun ME, Topcu C, Ahmet AK. The efficacy of intratechal administration of a very low dose potirelin after acute spinal cord injury. Injury 2008; 39:1403-7. [PMID: 19036363 DOI: 10.1016/j.injury.2008.05.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2007] [Revised: 04/05/2008] [Accepted: 05/07/2008] [Indexed: 02/02/2023]
Abstract
BACKGROUND AND OBJECT The objective of this study was to determine the effect of a very low dose protirelin in cerebrospinal fluid (CSF) glucose, magnesium and lactate levels after spinal cord trauma (SCT) in rabbits. We also aimed to evaluate whether this very low dose might induce analeptic effect. MATERIAL AND METHODS Twenty rabbits were divided equally into two groups: group I (n=10) was the control group, suffered from SCT but received only saline after SCT. Group II (n=10) (treatment group), received a very low dose of 0.05 mg/kg thyrotropin releasing hormone (TRH), analogue protirelin intratechally after SCT. The basal CSF glucose, magnesium and lactate levels were recorded in both groups. CSF lactate, glucose and magnesium contents were recorded at the same time (an hour before and after) SCT. Serum thyroid stimulating hormone (TSH), freetriiodothyronine (FT3) and freethyroxine (FT4) were measured in all rabbits before and after SCT. RESULTS Before spinal cord trauma, there were not any significant differences in glucose, lactate and magnesium levels between group I and II whereas, after spinal cord trauma in group II, the significant suppression in elevation of lactate and glucose depletion (p<0.05) were observed while no significant suppression was observed in magnesium level (p>0.05) as compared with group I (Table 3). In respect of serum TSH levels, there were not any significant differences between two groups before and after SCT. CONCLUSIONS This study showed that intratechal TRH has no analeptic effect on serum TSH, FT3 and FT4 levels but can attenuate the increase of lactate levels following spinal cord trauma. No significant decrease in magnesium level and also suppression of glucose decline in group II, may be related to the neuroprotective effects of TRH.
Collapse
Affiliation(s)
- Sahika Liva Cengiz
- Neurosurgery Department, Selcuk University, Meram Faculty of Medicine, Konya, Turkey
| | | | | | | |
Collapse
|
15
|
Jam F, Tullberg M, Luthman K, Grøtli M. Microwave assisted synthesis of spiro-2,5-diketopiperazines. Tetrahedron 2007. [DOI: 10.1016/j.tet.2007.06.108] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
16
|
Guan J, Mathai S, Harris P, Wen JY, Zhang R, Brimble M, Gluckman P. Peripheral administration of a novel diketopiperazine, NNZ 2591, prevents brain injury and improves somatosensory-motor function following hypoxia-ischemia in adult rats. Neuropharmacology 2007; 53:749-62. [PMID: 17904590 DOI: 10.1016/j.neuropharm.2007.08.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2007] [Revised: 08/03/2007] [Accepted: 08/06/2007] [Indexed: 11/29/2022]
Abstract
The current study describes the neuroprotective effects of an endogenous diketopiperazine, cyclo-glycyl-proline (cyclic GP), in rats with hypoxic-ischemic brain injury and the pre-clinical development of an analogue, cyclo-L-glycyl-L-2-allylproline (NNZ 2591), modified for improved bioavailability. The compounds were given either intracerebroventricularly or subcutaneously 2h after hypoxia-ischemia. Histology, immunohistochemistry and behavior were used to evaluate treatment effects. The central uptake of NNZ 2591 was also examined in normal and hypoxic-ischemic injured rats by HPLC-mass spectrometry. Central administration of cyclic GP or NNZ 2591 reduced the extent of brain damage in the lateral cortex, the hippocampus and the striatum (p<0.001), with NNZ 2591 being more potent. NNZ 2591 was stable in the plasma and crossed the blood-brain barrier independent of hypoxic-ischemic injury. The level of NNZ 2591 in the CSF was maintained for 2 h after a single subcutaneous dose, and modest neuroprotection was seen after a bolus subcutaneous administration (overall p<0.001). Treatment with NNZ 2591 for 5 d subcutaneously improved somatosensory-motor function (p<0.05) and long-term histological outcome (overall p<0.0001). NNZ 2591 treatment not only reduced both caspase-3 mediated apoptosis and microglial activation but also enhanced astrocytic reactivity, which may mediate its protective effect. The pharmacokinetic profile and potent long-term protective effects of NNZ 2591 suggests its utility for the treatment of ischemic brain injury and other neurological conditions requiring chronic intervention.
Collapse
Affiliation(s)
- J Guan
- The Liggins Institute, University of Auckland, Private Bag 92019, Auckland, New Zealand.
| | | | | | | | | | | | | |
Collapse
|
17
|
Ottens AK, Kobeissy FH, Fuller BF, Liu MC, Oli MW, Hayes RL, Wang KKW. Novel neuroproteomic approaches to studying traumatic brain injury. PROGRESS IN BRAIN RESEARCH 2007; 161:401-18. [PMID: 17618994 DOI: 10.1016/s0079-6123(06)61029-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Neuroproteomics entails wide-scope study of the nervous system proteome in both its content and dynamics. The field employs high-end analytical mass spectrometry and novel high-throughput antibody approaches to characterize as many proteins as possible. The most common application has been differential analysis to identify a limited set of highly dynamic proteins associated with injury, disease, or other altered states of the nervous system. Traumatic brain injury (TBI) is an important neurological condition where neuroproteomics has revolutionized the characterization of protein dynamics, leading to a greater understanding of post-injury biochemistry. Further, proteins of altered abundance or post-translational modifications identified by neuroproteomic studies are candidate biochemical markers of TBI. This chapter explores the use of neuroproteomics in the study of TBI and the validation of identified putative biomarkers for subsequent clinical translation into novel injury diagnostics.
Collapse
Affiliation(s)
- Andrew K Ottens
- Department of Psychiatry, Center for Neuroproteomics and Biomarkers Research at the McKnight Brain Institute of the University of Florida, PO Box 100256, Gainesville, FL 32610, USA.
| | | | | | | | | | | | | |
Collapse
|
18
|
Lippert-Gruener M, Maegele M, Garbe J, Angelov DN. Late effects of enriched environment (EE) plus multimodal early onset stimulation (MEOS) after traumatic brain injury in rats: Ongoing improvement of neuromotor function despite sustained volume of the CNS lesion. Exp Neurol 2006; 203:82-94. [PMID: 16965773 DOI: 10.1016/j.expneurol.2006.07.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2006] [Revised: 07/05/2006] [Accepted: 07/24/2006] [Indexed: 11/30/2022]
Abstract
Recently we showed that the combination between MEOS and EE applied to rats for 7-15 days after traumatic brain injury (TBI) was associated with reduced CNS lesion volume and enhanced reversal of neuromotor dysfunction. In a continuation of this work, we tested whether these effects persisted for longer post-operative periods, e.g. 30 days post-injury (dpi). Rats were subjected to lateral fluid percussion (LFP) or to sham injury. After LFP, one third of the animals (injured and sham) was placed under conditions of standard housing (SH), one third was kept in EE-only, and one third received EE+MEOS. Standardized composite neuroscore (NS) for neurological functions and computerized analysis of the vibrissal motor performance were used to assess post-traumatic neuromotor deficits. These were followed by evaluation of the cortical lesion volume (CLV) after immunostaining for neuron-specific enolase, caspase 3 active, and GFAP. Finally, the volume of cortical lesion containing regeneration-associated proteins (CLV-RAP) was determined in sections stained for GAP-43, MAP2, and neuronal class III beta-tubulin. We found (i) no differences in the vibrissal motor performance; (ii) EE+MEOS rats performed significantly better than SH rats in NS; (iii) EE-only and EE+MEOS animals, but not SH rats, showed better recovery at 30 dpi than at 15 dpi; (iv) no differences among all groups in CLV (larger than that at 15 dpi) and CLV-RAP, despite a clear tendency to reduction in the EE-only and EE+MEOS rats. We conclude that EE+MEOS retards, but cannot prevent the increase of lesion volume. This retardation is sufficient for a continuous restoration of neurological functions.
Collapse
|
19
|
Yakovlev AG, Faden AI. Mechanisms of neural cell death: implications for development of neuroprotective treatment strategies. NeuroRx 2005; 1:5-16. [PMID: 15717003 PMCID: PMC534908 DOI: 10.1602/neurorx.1.1.5] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
It has been increasingly recognized that cell death phenotypes and their molecular mechanisms are highly diverse. Necrosis is no longer considered a single entity, passively mediated by energy failure. Moreover, caspase-dependent apoptosis is not the only pathway involved in programmed cell death or even the only apoptotic mechanism. Recent experimental work emphasizes the diverse and interrelated nature of cell death mechanisms. Thus, there are both caspase-dependent and caspase-independent forms of apoptosis, which may differ morphologically as well as mechanistically. There are also necrotic-like phenotypes that require de novo protein synthesis and are, therefore, forms of programmed cell death. In addition, forms of cell death showing certain morphological features of both necrosis and apoptosis have been identified, leading to the term aponecrosis. Considerable experimental evidence also shows that modulation of one form of cell death may lead to another. Together, these observations underscore the need to substantially revise our conceptions about neuroprotection strategies. Use of multiple treatments that target different cell death cascades, or single agents that moderate multiple cell death pathways, is likely to lead to more effective neuroprotection for clinical disorders.
Collapse
Affiliation(s)
- Alexander G Yakovlev
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC 20057, USA
| | | |
Collapse
|
20
|
Nie Y, Schoepp DD, Klaunig JE, Yard M, Lahiri DK, Kubek MJ. Thyrotropin-releasing hormone (protirelin) inhibits potassium-stimulated glutamate and aspartate release from hippocampal slices in vitro. Brain Res 2005; 1054:45-54. [PMID: 16055093 DOI: 10.1016/j.brainres.2005.06.077] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2005] [Revised: 06/17/2005] [Accepted: 06/25/2005] [Indexed: 10/25/2022]
Abstract
Excess excitatory amino acid release is involved in pathways associated with seizures and neurodegeneration. Thyrotropin-releasing hormone (TRH; protirelin), a brain-derived tripeptide, has shown efficacy in the treatment of such disorders, yet its mechanism of neuroprotection is poorly understood. Using superfused hippocampal slices, we tested the hypothesis that TRH could inhibit evoked glutamate/aspartate release in vitro. Rat hippocampal slices were first equilibrated in oxygenated Krebs buffer (KRB) (120 min) then superfused for 10 min with KRB (control), or KRB containing 0.1, 1, or 10 microM TRH respectively, prior to and during 5 min depolarization with high potassium KRB (50 mM [K(+)] +/- TRH). Fractions (1 min) were collected during the 5 min stimulation and for an additional 10 min thereafter and analyzed for glutamate and aspartate by HPLC. TRH had no effect on baseline glutamate/aspartate release, while all three TRH doses significantly (P < 0.05) inhibited peak 50 mM [K(+)]-stimulated glutamate/aspartate release, and glutamate remained below control (P < 0.05) at 15 min post stimulation. A 5 min pulse of TRH (10 microM) had no affect on basal glutamate/aspartate release, whereas the TRH pre-pulsed slices failed to release glutamate/aspartate by [K(+)]-stimulation given 15 min later. These results are the first to show a potent and prolonged inhibitory effect of TRH on evoked glutamate/aspartate release in vitro. These initial studies suggest that exogenous and/or endogenous TRH may function, in part, to modulate excess glutamate release in specific CNS loci. Additional studies are in progress to fully understand the mechanism of this potent effect of TRH and its implication in various CNS disorders.
Collapse
Affiliation(s)
- Y Nie
- Department of Anatomy and Cell Biology, MS 5035, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN 46202, USA
| | | | | | | | | | | |
Collapse
|
21
|
Abstract
Animal models of traumatic brain injury (TBI) are used to elucidate primary and secondary sequelae underlying human head injury in an effort to identify potential neuroprotective therapies for developing and adult brains. The choice of experimental model depends upon both the research goal and underlying objectives. The intrinsic ability to study injury-induced changes in behavior, physiology, metabolism, the blood/tissue interface, the blood brain barrier, and/or inflammatory- and immune-mediated responses, makes in vivo TBI models essential for neurotrauma research. Whereas human TBI is a highly complex multifactorial disorder, animal trauma models tend to replicate only single factors involved in the pathobiology of head injury using genetically well-defined inbred animals of a single sex. Although such an experimental approach is helpful to delineate key injury mechanisms, the simplicity and hence inability of animal models to reflect the complexity of clinical head injury may underlie the discrepancy between preclinical and clinical trials of neuroprotective therapeutics. Thus, a search continues for new animal models, which would more closely mimic the highly heterogeneous nature of human TBI, and address key factors in treatment optimization.
Collapse
Affiliation(s)
- Ibolja Cernak
- Department of Neuroscience, Georgetown University Medical Center, Washington, D.C. 20057, USA.
| |
Collapse
|
22
|
Fricke ST, Vink R, Chiodo C, Cernak I, Ileva L, Faden AI. Consistent and reproducible slice selection in rodent brain using a novel stereotaxic device for MRI. J Neurosci Methods 2004; 136:99-102. [PMID: 15126050 DOI: 10.1016/j.jneumeth.2004.01.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2003] [Revised: 08/25/2003] [Accepted: 01/09/2004] [Indexed: 11/20/2022]
Abstract
Typically small animal radiological images are obtained after placing the animal in the center of the imaging device using beds or platforms, and then adjusting the position after obtaining a scout image. Such a process does not permit the reproducible visualization of the same anatomical plane with repeated examinations. We have developed a device that allows stereotaxic placement of an animal in precisely the same position for repeated examinations. The instrument incorporates a full range of physiological monitoring and life support systems including temperature control, anesthesia delivery and respiratory monitoring. Using magnetic resonance imaging (MRI), the accuracy and reliability of this device is demonstrated in a rat traumatic brain injury (TBI) model.
Collapse
Affiliation(s)
- S T Fricke
- Department of Neuroscience, Research Building, Room WB-01, Georgetown University, P.O. Box 571464, Washington, DC 20057-1464, USA.
| | | | | | | | | | | |
Collapse
|
23
|
Yakovlev AG, Faden AI. Mechanisms of neural cell death: Implications for development of neuroprotective treatment strategies. Neurotherapeutics 2004. [DOI: 10.1007/bf03206563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|