1
|
Sun S, Tian R, Alford A, Yin D, Shi R. An analog of phenelzine demonstrates effective acrolein scavenging and neuroprotection without monoamine oxidase inhibition in a rat SCI model. Neuroscience 2025; 574:54-64. [PMID: 40189130 DOI: 10.1016/j.neuroscience.2025.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 03/26/2025] [Accepted: 04/03/2025] [Indexed: 04/14/2025]
Abstract
Oxidative stress is widely recognized as a critical factor in the functional deficits after spinal cord injury (SCI). Oxidative stress and lipid peroxidation-derived aldehydes such as acrolein are known to play a key role in SCI pathology and have therefore emerged as valuable therapeutic targets. This study introduces a novel phenelzine analogue (PhzA), designed to retain the acrolein scavenging capability of phenelzine (Phz) while removing its undesirable monoamine oxidase (MAO) inhibition effects through structure-based modification. Using a rat model of contusion SCI, we showed that PhzA significantly reduced acrolein levels in both the acute and chronic stages of SCI with minimal MAO inhibition. In addition, PhzA reduced excessive microglial and astrocytic activation, dampening inflammation and gliosis. Furthermore, PhzA-treated rats exhibited significant improvements in motor function and reduction in mechanical hypersensitivity for up to 28 days post-injury compared to untreated rats. These findings further underscore the crucial role of aldehydes in SCI pathology and strengthen the notion that acrolein could serve as an effective therapeutic target for mitigating post-SCI neurodegeneration. These results also indicate that the expansion of acrolein-scavenging drug discovery through structure-based modification of existing repurposed drugs, such as with Phz, is a viable strategy with the benefit of a likely accelerated path towards clinical application. This effort may also benefit a range of neuronal diseases and injuries beyond SCI where acrolein is implicated, advancing the health of millions of patients.
Collapse
Affiliation(s)
- Siyuan Sun
- Department of Basic Medical Sciences, College of Veterinary Medicine Purdue University, West Lafayette, IN, USA; Center for Paralysis Research, Purdue University, West Lafayette, IN, USA
| | - Ran Tian
- Department of Basic Medical Sciences, College of Veterinary Medicine Purdue University, West Lafayette, IN, USA; Center for Paralysis Research, Purdue University, West Lafayette, IN, USA; Weldon School of Biomedical Engineering, College of Engineering, Purdue University, West Lafayette, IN, USA
| | - Anna Alford
- Center for Paralysis Research, Purdue University, West Lafayette, IN, USA; Weldon School of Biomedical Engineering, College of Engineering, Purdue University, West Lafayette, IN, USA
| | - David Yin
- Department of Basic Medical Sciences, College of Veterinary Medicine Purdue University, West Lafayette, IN, USA; West Lafayette Junior/Senior High School, West Lafayette, IN, USA
| | - Riyi Shi
- Department of Basic Medical Sciences, College of Veterinary Medicine Purdue University, West Lafayette, IN, USA; Center for Paralysis Research, Purdue University, West Lafayette, IN, USA; Weldon School of Biomedical Engineering, College of Engineering, Purdue University, West Lafayette, IN, USA; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
2
|
Hirakawa T, Taniuchi M, Iguchi Y, Bogahawaththa S, Yoshitake K, Werellagama S, Uemura T, Tsujita T. NF-E2-related factor 1 suppresses the expression of a spermine oxidase and the production of highly reactive acrolein. Sci Rep 2025; 15:12405. [PMID: 40258928 PMCID: PMC12012012 DOI: 10.1038/s41598-025-96388-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 03/27/2025] [Indexed: 04/23/2025] Open
Abstract
Polyamines (putrescine, spermidine, and spermine) are among the most abundant intracellular small molecular metabolites, with concentrations at the mM level. The ratios of these three molecules remain constant under physiological conditions. Stress (i.e. polyamine overload, oxidative stress, aging, infection, etc.) triggers the catabolic conversion of spermine to spermidine, ultimately yielding acrolein and hydrogen peroxide. The potential of acrolein to induce DNA damage and protein denaturation is 1,000 times greater than that of reactive oxygen species. We have shown that these polyamine metabolic pathways also involve the nuclear factor erythroid-2-related factor 1 (NRF1) transcription factor. In our chemically-inducible, liver-specific Nrf1-knockout mice, the polyamine catabolic pathway dominated the anabolic pathway, producing free acrolein and accumulating acrolein-conjugated proteins in vivo. This metabolic feature implicates SMOX as an important causative enzyme. Chromatin immunoprecipitation and reporter assays confirmed that NRF1 directly suppressed Smox expression. This effect was also observed in vitro. Ectopic overexpression of SMOX increased the accumulation of free acrolein and acrolein-conjugated proteins. SMOX knockdown reversed the accumulation of free acrolein and acrolein-conjugated proteins. Our results show that NRF1 typically suppresses Smox expression when NRF1 is downregulated, SMOX is upregulated, and polyamine metabolic pathways are altered, producing low molecular weight polyamines and acrolein.
Collapse
Affiliation(s)
- Tomoaki Hirakawa
- Laboratory of Biochemistry, Faculty of Agriculture, Saga University, Saga, Japan
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, Japan
| | - Megumi Taniuchi
- Laboratory of Biochemistry, Faculty of Agriculture, Saga University, Saga, Japan
| | - Yoko Iguchi
- Laboratory of Biochemistry, Faculty of Agriculture, Saga University, Saga, Japan
| | - Sudarma Bogahawaththa
- Laboratory of Biochemistry, Faculty of Agriculture, Saga University, Saga, Japan
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, Japan
| | - Kiko Yoshitake
- Laboratory of Biochemistry, Faculty of Agriculture, Saga University, Saga, Japan
| | - Shanika Werellagama
- Laboratory of Biochemistry, Faculty of Agriculture, Saga University, Saga, Japan
| | - Takeshi Uemura
- Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, Saitama, Japan
| | - Tadayuki Tsujita
- Laboratory of Biochemistry, Faculty of Agriculture, Saga University, Saga, Japan.
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, Japan.
| |
Collapse
|
3
|
Zhang Z, Xiao T, Hall MR, Crodian JS, Alford AK, Kimbrough A, Shi R. Temporal differential effects of post-injury alcohol consumption in a mouse model of blast-induced traumatic brain injury. Neuroscience 2024; 562:239-251. [PMID: 39369945 PMCID: PMC11769080 DOI: 10.1016/j.neuroscience.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/22/2024] [Accepted: 10/01/2024] [Indexed: 10/08/2024]
Abstract
Traumatic brain injury is a prevalent condition that affects millions worldwide with no clear understanding or effective therapeutic management available. Military soldiers have a high risk of exposure to blast-induced traumatic brain injury (bTBI). Furthermore, alcohol drinking is common in this population, and studies have shown that post-TBI alcohol exposure can result in memory loss. Hence, it is possible that alcohol could contribute to the overall pathological outcome of brain trauma. However, such a possibility has not been explored in detail. Here, we combined a mild bTBI (mbTBI) model with the drinking-in-the-dark (DID) paradigm to investigate the pathological synergy between mbTBI and alcohol consumption by examining brain oxidative stress levels and behavioral alterations in mice. The results revealed the anxiolytic and short-term memory improvement effects of post-trauma alcohol drinking examined at an early timepoint post mbTBI. However, extended alcohol drinking for up to three weeks post mbTBI impaired long-term memory and was accompanied by intensified oxidative stress in brain regions associated with memory and anxiety. These findings, as well as those from previous in vitro TBI/alcohol studies, suggest a pathological synergy of physical force and post-impact alcohol exposure. This knowledge could potentially aid in establishing guidelines for TBI victims to avoid further injury to their brains as well as to help maximize their recovery following TBI.
Collapse
Affiliation(s)
- Zaiyang Zhang
- Department of Basic Medical Sciences, College of Veterinary Medicine Purdue University, West Lafayette, IN, United States; Center for Paralysis Research, Purdue University, West Lafayette, IN, United States
| | - Tiange Xiao
- Department of Basic Medical Sciences, College of Veterinary Medicine Purdue University, West Lafayette, IN, United States
| | - Mekyna R Hall
- Department of Basic Medical Sciences, College of Veterinary Medicine Purdue University, West Lafayette, IN, United States; Center for Paralysis Research, Purdue University, West Lafayette, IN, United States
| | - Jennifer S Crodian
- Department of Basic Medical Sciences, College of Veterinary Medicine Purdue University, West Lafayette, IN, United States; Center for Paralysis Research, Purdue University, West Lafayette, IN, United States
| | - Anna K Alford
- Department of Basic Medical Sciences, College of Veterinary Medicine Purdue University, West Lafayette, IN, United States; Center for Paralysis Research, Purdue University, West Lafayette, IN, United States; Weldon School of Biomedical Engineering, College of Engineering, Purdue University, West Lafayette, IN, United States
| | - Adam Kimbrough
- Department of Basic Medical Sciences, College of Veterinary Medicine Purdue University, West Lafayette, IN, United States; Weldon School of Biomedical Engineering, College of Engineering, Purdue University, West Lafayette, IN, United States; The Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN, United States.
| | - Riyi Shi
- Department of Basic Medical Sciences, College of Veterinary Medicine Purdue University, West Lafayette, IN, United States; Center for Paralysis Research, Purdue University, West Lafayette, IN, United States; Weldon School of Biomedical Engineering, College of Engineering, Purdue University, West Lafayette, IN, United States; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, United States.
| |
Collapse
|
4
|
Baker E, Harris WT, Guimbellot JS, Bliton K, Rowe SM, Raju SV, Oates GR. Association between biomarkers of tobacco smoke exposure and clinical efficacy of ivacaftor in the G551D observational trial (GOAL). J Cyst Fibros 2024; 23:959-966. [PMID: 39033068 PMCID: PMC11410542 DOI: 10.1016/j.jcf.2024.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 06/15/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Acrolein, an aldehyde in smoke from tobacco products, inhibits CFTR function in vitro. Ivacaftor is an FDA-approved potentiator that improves mutant CFTR function. This human clinical study investigated the relationship between two urinary markers of tobacco smoke exposure - the acrolein metabolite 3-HPMA and the nicotine metabolite NNAL - and sweat chloride response to ivacaftor in the G551D Observational Trial (GOAL). METHODS 3-HPMA (low: <50th centile; moderate: 50-75th centile; high: >75th centile) and NNAL (detectable/undetectable) in GOAL samples was quantified with LC-MS/MS. Self-report of tobacco smoke exposure (Y/N) served as a subjective measure. Change in sweat chloride from pre- to 6 months post-ivacaftor treatment (ΔSC) was the primary CFTR-dependent readout. RESULTS The sample included 151 individuals, mean age 20.7 (SD 11.4) years, range 6-59 years. Smoke exposure prevalence was 15 % per self-reports but 27 % based on detectable NNAL. 3-HPMA was increased in those reporting tobacco smoke exposure (607 vs 354 ng/ml, p = 0.008), with a higher proportion of smoke-exposed in the high- vs low-acrolein group (31 % vs 9 %, p=0.040). Compared to low-acrolein counterparts, high-acrolein participants experienced less decrease in sweat chloride (-35.2 vs -48.2 mmol/L; p = 0.020) and had higher sweat chloride values (50.6 vs 37.6 mmol/L; p = 0.020) 6 months post-ivacaftor. The odds of ivacaftor-mediated potentiation to near normative CFTR function (defined as SC6mo <40 mmol/L) was more than twice as high in the low-acrolein cohort (OR: 2.51, p = 0.026). CONCLUSIONS Increased urinary 3-HPMA, an acrolein metabolite of tobacco smoke, is associated with a diminished sweat chloride response to ivacaftor potentiation of CFTR function.
Collapse
Affiliation(s)
- Elizabeth Baker
- Medicine University of Alabama at Birmingham 1808 7th Ave S, BDB 853 Birmingham, AL 35233 United States
| | - William T Harris
- Medicine University of Alabama at Birmingham 1808 7th Ave S, BDB 853 Birmingham, AL 35233 United States
| | - Jennifer S Guimbellot
- Medicine University of Alabama at Birmingham 1808 7th Ave S, BDB 853 Birmingham, AL 35233 United States; The University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Kyle Bliton
- Medicine University of Alabama at Birmingham 1808 7th Ave S, BDB 853 Birmingham, AL 35233 United States
| | - Steven M Rowe
- Medicine University of Alabama at Birmingham 1808 7th Ave S, BDB 853 Birmingham, AL 35233 United States
| | - S Vamsee Raju
- Medicine University of Alabama at Birmingham 1808 7th Ave S, BDB 853 Birmingham, AL 35233 United States
| | - Gabriela R Oates
- Medicine University of Alabama at Birmingham 1808 7th Ave S, BDB 853 Birmingham, AL 35233 United States.
| |
Collapse
|
5
|
Tang J, Alford A, Leung G, Tully M, Shi R. Neuroprotection by acrolein sequestration through exogenously applied scavengers and endogenous enzymatic enabling strategies in mouse EAE model. Sci Rep 2024; 14:6027. [PMID: 38472318 PMCID: PMC10933361 DOI: 10.1038/s41598-024-56035-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
We have previously shown that the pro-oxidative aldehyde acrolein is a critical factor in MS pathology. In this study, we found that the acrolein scavenger hydralazine (HZ), when applied from the day of induction, can suppress acrolein and alleviate motor and sensory deficits in a mouse experimental autoimmune encephalomyelitis (EAE) model. Furthermore, we also demonstrated that HZ can alleviate motor deficits when applied after the emergence of MS symptoms, making potential anti-acrolein treatment a more clinically relevant strategy. In addition, HZ can reduce both acrolein and MPO, suggesting a connection between acrolein and inflammation. We also found that in addition to HZ, phenelzine (PZ), a structurally distinct acrolein scavenger, can mitigate motor deficits in EAE when applied from the day of induction. This suggests that the likely chief factor of neuroprotection offered by these two structurally distinct acrolein scavengers in EAE is their common feature of acrolein neutralization. Finally, up-and-down regulation of the function of aldehyde dehydrogenase 2 (ALDH2) in EAE mice using either a pharmacological or genetic strategy led to correspondent motor and sensory changes. This data indicates a potential key role of ALDH2 in influencing acrolein levels, oxidative stress, inflammation, and behavior in EAE. These findings further consolidate the critical role of aldehydes in the pathology of EAE and its mechanisms of regulation. This is expected to reinforce and expand the possible therapeutic targets of anti-aldehyde treatment to achieve neuroprotection through both endogenous and exogenous manners.
Collapse
Affiliation(s)
- Jonathan Tang
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Center for Paralysis Research, Purdue University, West Lafayette, IN, 47907, USA
| | - Anna Alford
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Center for Paralysis Research, Purdue University, West Lafayette, IN, 47907, USA
| | - Gary Leung
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA
- Center for Paralysis Research, Purdue University, West Lafayette, IN, 47907, USA
| | - Melissa Tully
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Center for Paralysis Research, Purdue University, West Lafayette, IN, 47907, USA
- MSTP Program, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Riyi Shi
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA.
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA.
- Center for Paralysis Research, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
6
|
Rogers EA, Beauclair T, Thyen A, Shi R. Utilizing novel TBI-on-a-chip device to link physical impacts to neurodegeneration and decipher primary and secondary injury mechanisms. Sci Rep 2022; 12:11838. [PMID: 35821510 PMCID: PMC9276772 DOI: 10.1038/s41598-022-14937-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 06/15/2022] [Indexed: 11/17/2022] Open
Abstract
While clinical observations have confirmed a link between the development of neurodegenerative diseases and traumatic brain injuries (TBI), there are currently no treatments available and the underlying mechanisms remain elusive. In response, we have developed an in vitro pendulum trauma model capable of imparting rapid acceleration injuries to neuronal networks grown on microelectrode arrays within a clinically relevant range of g forces, with real-time electrophysiological and morphological monitoring. By coupling a primary physical insult with the quantification of post-impact levels of known biochemical pathological markers, we demonstrate the capability of our system to delineate and investigate the primary and secondary injury mechanisms leading to post-impact neurodegeneration. Specifically, impact experiments reveal significant, force-dependent increases in the pro-inflammatory, oxidative stress marker acrolein at 24 h post-impact. The elevation of acrolein was augmented by escalating g force exposures (30-200 g), increasing the number of rapidly repeated impacts (4-6 s interval, 3, 5 and 10×), and by exposing impacted cells to 40 mM ethanol, a known comorbidity of TBI. The elevated levels of acrolein following multiple impacts could be reduced by increasing time-intervals between repeated hits. In addition, we show that conditioned media from maximally-impacted cultures can cause cellular acrolein elevation when introduced to non-impact, control networks, further solidifying acrolein's role as a diffusive-factor in post-TBI secondary injuries. Finally, morphological data reveals post-impact acrolein generation to be primarily confined to soma, with some emergence in cellular processes. In conclusion, this novel technology provides accurate, physical insults with a unique level of structural and temporal resolution, facilitating the investigation of post-TBI neurodegeneration.
Collapse
Affiliation(s)
- Edmond A Rogers
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Department of Basic Medical Sciences, School of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA
- Center for Paralysis Research, Purdue University, West Lafayette, IN, 47907, USA
| | - Timothy Beauclair
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Department of Basic Medical Sciences, School of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA
- Center for Paralysis Research, Purdue University, West Lafayette, IN, 47907, USA
| | - Andrew Thyen
- Indiana University School of Medicine, Indianapolis, IN, 46033, USA
| | - Riyi Shi
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA.
- Department of Basic Medical Sciences, School of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA.
- Center for Paralysis Research, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
7
|
Clayton DB, Tong CMC, Li B, Taylor AS, De S, Mason MD, Dudley AG, Davidoff O, Kobayashi H, Haase VH. Inhibition of hypoxia-inducible factor-prolyl hydroxylation protects from cyclophosphamide-induced bladder injury and urinary dysfunction. Am J Physiol Renal Physiol 2022; 323:F81-F91. [PMID: 35499237 PMCID: PMC9236868 DOI: 10.1152/ajprenal.00344.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 04/25/2022] [Accepted: 04/25/2022] [Indexed: 11/22/2022] Open
Abstract
Disruption of the blood-urine barrier can result in acute or chronic inflammatory bladder injury. Activation of the oxygen-regulated hypoxia-inducible factor (HIF) pathway has been shown to protect mucosal membranes by increasing the expression of cytoprotective genes and by suppressing inflammation. The activity of HIF is controlled by prolyl hydroxylase domain (PHD) dioxygenases, which have been exploited as therapeutic targets for the treatment of anemia of chronic kidney disease. Here, we established a mouse model of acute cyclophosphamide (CYP)-induced blood-urine barrier disruption associated with inflammation and severe urinary dysfunction to investigate the HIF-PHD axis in inflammatory bladder injury. We found that systemic administration of dimethyloxalylglycine or molidustat, two small-molecule inhibitors of HIF-prolyl hydroxylases, profoundly mitigated CYP-induced bladder injury and inflammation as assessed by morphological analysis of transmural edema and urothelial integrity and by measuring tissue cytokine expression. Void spot analysis to examine bladder function quantitatively demonstrated that HIF-prolyl hydroxylase inhibitor administration normalized micturition patterns and protected against CYP-induced alteration of urinary frequency and micturition patterns. Our study highlights the therapeutic potential of HIF-activating small-molecule compounds for the prevention or therapy of bladder injury and urinary dysfunction due to blood-urine barrier disruption.NEW & NOTEWORTHY Disruption of the blood-urine barrier can result in acute or chronic inflammatory bladder injury. Here, we demonstrate that pharmacological inhibition of hypoxia-inducible factor (HIF)-prolyl hydroxylation prevented bladder injury and protected from urinary dysfunction in a mouse model of cyclophosphamide-induced disruption of the blood-urine barrier. Our study highlights a potential role for HIF-activating small-molecule compounds in the prevention or therapy of bladder injury and urinary dysfunction and provides a rationale for future clinical studies.
Collapse
Affiliation(s)
- Douglass B Clayton
- Division of Pediatric Urology, Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Ching Man Carmen Tong
- Division of Pediatric Urology, Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Belinda Li
- Division of Pediatric Urology, Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Abby S Taylor
- Division of Pediatric Urology, Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Shuvro De
- Division of Pediatric Urology, Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Matthew D Mason
- Division of Pediatric Urology, Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Anne G Dudley
- Division of Pediatric Urology, Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Olena Davidoff
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Medical and Research Services, Department of Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee
| | - Hanako Kobayashi
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Medical and Research Services, Department of Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee
| | - Volker H Haase
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Medical and Research Services, Department of Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
8
|
Pradipta AR, Tanaka K. Biofunctional chemistry and reactivity of biogenic acrolein for cancer diagnosis and therapy. Chem Commun (Camb) 2021; 57:9798-9806. [PMID: 34581321 DOI: 10.1039/d1cc03590h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Acrolein holds excellent potential as a biomarker in various oxidative stress-related diseases, including cancer, Alzheimer's, Parkinson's, and inflammatory disorders. Consequently, a direct method to target and visualize acrolein in biological systems might be essential to provide tools for diagnosis and therapeutic purposes. Previously, we discovered 1,3-dipolar cycloaddition between aryl azides and acrolein, which proceeds without a catalyst to give α-diazocarbonyl derivatives. The reaction proceeds with high reactivity and selectivity even under physiological conditions. We have successfully utilized the reaction as a robust method for detecting acrolein generated by cancer cells. This review discusses the utilization of the endogenous acrolein reaction with aryl azide to (1) distinguish breast cancer from normal tissue during breast-conserving surgery and (2) treat cancer through selective prodrug activation in a mouse model without causing adverse effects. The methods have potential clinical application for breast-conserving surgery and are highly advantageous for cancer therapy.
Collapse
Affiliation(s)
- Ambara R Pradipta
- Department of Chemical Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology, 2-12-1 Ookayama, Meguro, 152-8552, Tokyo, Japan. .,Biofunctional Synthetic Chemistry Laboratory, Cluster for Pioneering Research, RIKEN, 2-1 Hirosawa, Wako, 351-0198, Saitama, Japan
| | - Katsunori Tanaka
- Department of Chemical Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology, 2-12-1 Ookayama, Meguro, 152-8552, Tokyo, Japan. .,Biofunctional Synthetic Chemistry Laboratory, Cluster for Pioneering Research, RIKEN, 2-1 Hirosawa, Wako, 351-0198, Saitama, Japan.,Biofunctional Chemistry Laboratory, Alexander Butlerov Institute of Chemistry, Kazan Federal University, 18 Kremlyovskaya Street, 420008, Kazan, Russian Federation
| |
Collapse
|
9
|
Race NS, Andrews KD, Lungwitz EA, Vega Alvarez SM, Warner TR, Acosta G, Cao J, Lu KH, Liu Z, Dietrich AD, Majumdar S, Shekhar A, Truitt WA, Shi R. Psychosocial impairment following mild blast-induced traumatic brain injury in rats. Behav Brain Res 2021; 412:113405. [PMID: 34097900 PMCID: PMC9284795 DOI: 10.1016/j.bbr.2021.113405] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 06/02/2021] [Accepted: 06/02/2021] [Indexed: 01/30/2023]
Abstract
Traumatic brain injury (TBI) is associated with increased risk for mental health disorders, impacting post-injury quality of life and societal reintegration. TBI is also associated with deficits in psychosocial processing, defined as the cognitive integration of social and emotional behaviors, however little is known about how these deficits manifest and their contributions to post-TBI mental health. In this pre-clinical investigation using rats, a single mild blast TBI (mbTBI) induced impairment of psychosocial processing in the absence of confounding physical polytrauma, post-injury motor deficits, affective abnormalities, or deficits in non-social behavior. Impairment severity correlated with acute upregulations of a known oxidative stress metabolite, 3-hydroxypropylmercapturic acid (3-HPMA), in urine. Resting state fMRI alterations in the acute post-injury period implicated key brain regions known to regulate psychosocial behavior, including orbitofrontal cortex (OFC), which is congruent with our previous report of elevated acrolein, a marker of neurotrauma and 3-HPMA precursor, in this region following mbTBI. OFC of mbTBI-exposed rats demonstrated elevated mRNA expression of metabotropic glutamate receptors 1 and 5 (mGluR1/5) and injection of mGluR1/5-selective agonist in OFC of uninjured rats approximated mbTBI-induced psychosocial processing impairment, demonstrating a novel role for OFC in this psychosocial behavior. Furthermore, OFC may serve as a hotspot for TBI-induced disruption of psychosocial processing and subsequent mental health disorders.
Collapse
Affiliation(s)
- Nicholas S Race
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA; Medical Scientist Training Program, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Katharine D Andrews
- Medical Scientist Training Program, Indiana University School of Medicine, Indianapolis, IN, USA; Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA; Program in Medical Neuroscience, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Elizabeth A Lungwitz
- Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA; Program in Medical Neuroscience, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sasha M Vega Alvarez
- PULSe Interdisciplinary Life Science Program, Purdue University, West Lafayette, IN, USA
| | - Timothy R Warner
- Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Anatomy, Cellular Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Glen Acosta
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - Jiayue Cao
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, USA
| | - Kun-Han Lu
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, USA; School of Electrical and Computer Engineering, Purdue University, West Lafayette, IN, USA
| | - Zhongming Liu
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, USA; School of Electrical and Computer Engineering, Purdue University, West Lafayette, IN, USA
| | - Amy D Dietrich
- Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Anatomy, Cellular Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sreeparna Majumdar
- Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA; Program in Medical Neuroscience, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Anantha Shekhar
- Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana Clinical and Translational Sciences Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - William A Truitt
- Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Anatomy, Cellular Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Riyi Shi
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA; PULSe Interdisciplinary Life Science Program, Purdue University, West Lafayette, IN, USA; Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, USA; Center for Paralysis Research, West Lafayette, IN, USA.
| |
Collapse
|
10
|
Wong ET, Luettich K, Krishnan S, Wong SK, Lim WT, Yeo D, Büttner A, Leroy P, Vuillaume G, Boué S, Hoeng J, Vanscheeuwijck P, Peitsch MC. Reduced Chronic Toxicity and Carcinogenicity in A/J Mice in Response to Life-Time Exposure to Aerosol From a Heated Tobacco Product Compared With Cigarette Smoke. Toxicol Sci 2021; 178:44-70. [PMID: 32780830 PMCID: PMC7657344 DOI: 10.1093/toxsci/kfaa131] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
We conducted an inhalation study, in accordance with Organisation for Economic Co-operation and Development Test Guideline 453, exposing A/J mice to tobacco heating system (THS) 2.2 aerosol or 3R4F reference cigarette smoke (CS) for up to 18 months to evaluate chronic toxicity and carcinogenicity. All exposed mice showed lower thymus and spleen weight, blood lymphocyte counts, and serum lipid concentrations than sham mice, most likely because of stress and/or nicotine effects. Unlike THS 2.2 aerosol-exposed mice, CS-exposed mice showed increased heart weight, changes in red blood cell profiles and serum liver function parameters. Similarly, increased pulmonary inflammation, altered lung function, and emphysematous changes were observed only in CS-exposed mice. Histopathological changes in other respiratory tract organs were significantly lower in the THS 2.2 aerosol-exposed groups than in the CS-exposed group. Chronic exposure to THS 2.2 aerosol also did not increase the incidence or multiplicity of bronchioloalveolar adenomas or carcinomas relative to sham, whereas CS exposure did. Male THS 2.2 aerosol-exposed mice had a lower survival rate than sham mice, related to an increased incidence of urogenital issues that appears to be related to congenital factors rather than test item exposure. The lower impact of THS 2.2 aerosol exposure on tumor development and chronic toxicity is consistent with the significantly reduced levels of harmful and potentially harmful constituents in THS 2.2 aerosol relative to CS. The totality of the evidence from this study further supports the risk reduction potential of THS 2.2 for lung diseases in comparison with cigarettes.
Collapse
Affiliation(s)
- Ee Tsin Wong
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| | - Karsta Luettich
- Department of Life Sciences, Systems Toxicology, PMI R&D, Philip Morris Products S.A, CH-2000 Neuchâtel, Switzerland
| | - Subash Krishnan
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| | - Sin Kei Wong
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| | - Wei Ting Lim
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| | - Demetrius Yeo
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| | | | - Patrice Leroy
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| | - Grégory Vuillaume
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| | - Stéphanie Boué
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| | - Julia Hoeng
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| | - Patrick Vanscheeuwijck
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| | - Manuel C Peitsch
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| |
Collapse
|
11
|
Boyko A, Tsepkova P, Aleshin V, Artiukhov A, Mkrtchyan G, Ksenofontov A, Baratova L, Ryabov S, Graf A, Bunik V. Severe Spinal Cord Injury in Rats Induces Chronic Changes in the Spinal Cord and Cerebral Cortex Metabolism, Adjusted by Thiamine That Improves Locomotor Performance. Front Mol Neurosci 2021; 14:620593. [PMID: 33867932 PMCID: PMC8044794 DOI: 10.3389/fnmol.2021.620593] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 03/05/2021] [Indexed: 01/23/2023] Open
Abstract
Our study aims at developing knowledge-based strategies minimizing chronic changes in the brain after severe spinal cord injury (SCI). The SCI-induced long-term metabolic alterations and their reactivity to treatments shortly after the injury are characterized in rats. Eight weeks after severe SCI, significant mitochondrial lesions outside the injured area are demonstrated in the spinal cord and cerebral cortex. Among the six tested enzymes essential for the TCA cycle and amino acid metabolism, mitochondrial 2-oxoglutarate dehydrogenase complex (OGDHC) is the most affected one. SCI downregulates this complex by 90% in the spinal cord and 30% in the cerebral cortex. This is associated with the tissue-specific changes in other enzymes of the OGDHC network. Single administrations of a pro-activator (thiamine, or vitamin B1, 1.2 mmol/kg) or a synthetic pro-inhibitor (triethyl glutaryl phosphonate, TEGP, 0.02 mmol/kg) of OGDHC within 15–20 h after SCI are tested as protective strategies. The biochemical and physiological assessments 8 weeks after SCI reveal that thiamine, but not TEGP, alleviates the SCI-induced perturbations in the rat brain metabolism, accompanied by the decreased expression of (acetyl)p53, increased expression of sirtuin 5 and an 18% improvement in the locomotor recovery. Treatment of the non-operated rats with the OGDHC pro-inhibitor TEGP increases the p53 acetylation in the brain, approaching the brain metabolic profiles to those after SCI. Our data testify to an important contribution of the OGDHC regulation to the chronic consequences of SCI and their control by p53 and sirtuin 5.
Collapse
Affiliation(s)
- Alexandra Boyko
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
| | - Polina Tsepkova
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
| | - Vasily Aleshin
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia.,Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Artem Artiukhov
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia.,Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Garik Mkrtchyan
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
| | - Alexander Ksenofontov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Lyudmila Baratova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Sergey Ryabov
- Russian Cardiology Research-and-Production Complex, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Anastasia Graf
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia.,Faculty of Nano-, Bio-, Informational and Cognitive Technologies, Moscow Institute of Physics and Technology, Moscow, Russia
| | - Victoria Bunik
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia.,Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia.,Department of Biological Chemistry, Sechenov University, Moscow, Russia
| |
Collapse
|
12
|
Khachatryan Z, Haunschild J, von Aspern K, Borger MA, Etz CD. Ischemic spinal cord injury - experimental evidence and evolution of protective measures. Ann Thorac Surg 2021; 113:1692-1702. [PMID: 33434541 DOI: 10.1016/j.athoracsur.2020.12.028] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 11/19/2020] [Accepted: 12/22/2020] [Indexed: 11/01/2022]
Abstract
BACKGROUND Paraplegia remains one of the most devastating complications of descending and thoracoabdominal aortic repair. The aim of this review is to outline the current state of art in the rapidly developing field of spinal cord injury (SCI) research. METHODS A review of PubMed and Web of Science databases was performed using the following terms and their combinations: spinal cord, injury, ischemia, ischemia-reperfusion, ischemic spinal cord injury, paraplegia, paraparesis. Articles published before July 2019 were screened and included if considered relevant. RESULTS The review focuses on the topic of SCI and the developments concerning methods of monitoring, diagnostics and prevention of SCI. CONCLUSIONS Translation of novel technologies from bench to bedside and into everyday clinical practice is challenging, however each of the developing areas hold great promise in SCI prevention.
Collapse
Affiliation(s)
- Zara Khachatryan
- University Department for Cardiac Surgery, Leipzig Heart Center, Struempellstrasse 39, 04289 Leipzig, Germany
| | - Josephina Haunschild
- University Department for Cardiac Surgery, Leipzig Heart Center, Struempellstrasse 39, 04289 Leipzig, Germany
| | - Konstantin von Aspern
- University Department for Cardiac Surgery, Leipzig Heart Center, Struempellstrasse 39, 04289 Leipzig, Germany
| | - Michael A Borger
- University Department for Cardiac Surgery, Leipzig Heart Center, Struempellstrasse 39, 04289 Leipzig, Germany
| | - Christian D Etz
- University Department for Cardiac Surgery, Leipzig Heart Center, Struempellstrasse 39, 04289 Leipzig, Germany.
| |
Collapse
|
13
|
Quantification of 3-Hydroxypropyl Mercapturic Acid in the Urine of Patients with Breast Cancer to Monitor Cyclophosphamide Toxicity. Ther Drug Monit 2020; 42:548-553. [DOI: 10.1097/ftd.0000000000000737] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
14
|
Szostak J, Wong ET, Titz B, Lee T, Wong SK, Low T, Lee KM, Zhang J, Kumar A, Schlage WK, Guedj E, Phillips B, Leroy P, Buettner A, Xiang Y, Martin F, Sewer A, Kuczaj A, Ivanov NV, Luettich K, Vanscheeuwijck P, Peitsch MC, Hoeng J. A 6-month systems toxicology inhalation study in ApoE -/- mice demonstrates reduced cardiovascular effects of E-vapor aerosols compared with cigarette smoke. Am J Physiol Heart Circ Physiol 2020; 318:H604-H631. [PMID: 31975625 DOI: 10.1152/ajpheart.00613.2019] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Smoking cigarettes is harmful to the cardiovascular system. Considerable attention has been paid to the reduced harm potential of alternative nicotine-containing inhalable products such as e-cigarettes. We investigated the effects of E-vapor aerosols or cigarette smoke (CS) on atherosclerosis progression, cardiovascular function, and molecular changes in the heart and aorta of female apolipoprotein E-deficient (ApoE-/-) mice. The mice were exposed to aerosols from three different E-vapor formulations: 1) carrier (propylene glycol and vegetable glycerol), 2) base (carrier and nicotine), or 3) test (base and flavor) or to CS from 3R4F reference cigarettes for up to 6 mo. Concentrations of CS and base or test aerosols were matched at 35 µg nicotine/L. Exposure to CS, compared with sham-exposed fresh air controls, accelerated atherosclerotic plaque formation, whereas no such effect was seen for any of the three E-vapor aerosols. Molecular changes indicated disease mechanisms related to oxidative stress and inflammation in general, plus changes in calcium regulation, and altered cytoskeletal organization and microtubule dynamics in the left ventricle. While ejection fraction, fractional shortening, cardiac output, and isovolumic contraction time remained unchanged following E-vapor aerosols exposure, the nicotine-containing base and test aerosols caused an increase in isovolumic relaxation time similar to CS. A nicotine-related increase in pulse wave velocity and arterial stiffness was also observed, but it was significantly lower for base and test aerosols than for CS. These results demonstrate that in comparison with CS, E-vapor aerosols induce substantially lower biological responses associated with smoking-related cardiovascular diseases.NEW & NOTEWORTHY Analysis of key urinary oxidative stress markers and proinflammatory cytokines showed an absence of oxidative stress and inflammation in the animals exposed to E-vapor aerosols. Conversely, animals exposed to conventional cigarette smoke had high urinary levels of these markers. When compared with conventional cigarette smoke, E-vapor aerosols induced smaller atherosclerotic plaque surface area and volume. Systolic and diastolic cardiac function, as well as endothelial function, were further significantly less affected by electronic cigarette aerosols than conventional cigarette smoke. Molecular analysis demonstrated that E-vapor aerosols induce significantly smaller transcriptomic dysregulation in the heart and aorta compared with conventional cigarette smoke.
Collapse
Affiliation(s)
- Justyna Szostak
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Ee Tsin Wong
- Philip Morris International Research and development, Philip Morris International Research Laboratories, Singapore
| | - Bjoern Titz
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Tom Lee
- Philip Morris International Research and development, Philip Morris International Research Laboratories, Singapore
| | - Sin Kei Wong
- Philip Morris International Research and development, Philip Morris International Research Laboratories, Singapore
| | - Tiffany Low
- Philip Morris International Research and development, Philip Morris International Research Laboratories, Singapore
| | | | | | | | | | - Emmanuel Guedj
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Blaine Phillips
- Philip Morris International Research and development, Philip Morris International Research Laboratories, Singapore
| | - Patrice Leroy
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | | | - Yang Xiang
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Florian Martin
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Alain Sewer
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Arkadiusz Kuczaj
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Nikolai V Ivanov
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Karsta Luettich
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Patrick Vanscheeuwijck
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Manuel C Peitsch
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| | - Julia Hoeng
- Philip Morris International Research and Development, Philip Morris Products, Neuchâtel, Switzerland
| |
Collapse
|
15
|
Acosta G, Race N, Herr S, Fernandez J, Tang J, Rogers E, Shi R. Acrolein-mediated alpha-synuclein pathology involvement in the early post-injury pathogenesis of mild blast-induced Parkinsonian neurodegeneration. Mol Cell Neurosci 2019; 98:140-154. [PMID: 31201929 PMCID: PMC6690849 DOI: 10.1016/j.mcn.2019.06.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 06/09/2019] [Accepted: 06/11/2019] [Indexed: 01/17/2023] Open
Abstract
Survivors of blast-induced traumatic brain injury (bTBI) have increased susceptibility to Parkinson's disease (PD), characterized by α-synuclein aggregation and the progressive degeneration of nigrostriatal dopaminergic neurons. Using an established bTBI rat model, we evaluated the changes of α-synuclein and tyrosine hydroxylase (TH), known hallmarks of PD, and acrolein, a reactive aldehyde and marker of oxidative stress, with the aim of revealing key pathways leading to PD post-bTBI. Indicated in both animal models of PD and TBI, acrolein is likely a point of pathogenic convergence. Here we show that after a single mild bTBI, acrolein is elevated up to a week, systemically in urine, and in whole brain tissue, specifically the substantia nigra and striatum. Acrolein elevation is accompanied by heightened α-synuclein oligomerization, dopaminergic dysregulation, and acrolein/α-synuclein interaction in the same brain regions. We further show that acrolein can directly modify and oligomerize α-synuclein in vitro. Taken together, our data suggests acrolein likely plays an important role in inducing PD pathology following bTBI by encouraging α-synuclein aggregation. These results are expected to advance our understanding of the long-term post-bTBI pathological changes leading to the development of PD, and suggest intervention targets to curtail such pathology.
Collapse
Affiliation(s)
- Glen Acosta
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - Nicholas Race
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA; Indiana University, School of Medicine, Indianapolis, IN, USA
| | - Seth Herr
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA; Purdue University Interdisciplinary Life Sciences Program (PULSe), Purdue University, West Lafayette, IN, USA
| | - Joseph Fernandez
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Jonathan Tang
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Edmond Rogers
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Riyi Shi
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA; Indiana University, School of Medicine, Indianapolis, IN, USA; Purdue University Interdisciplinary Life Sciences Program (PULSe), Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
16
|
Pradipta AR, Fujii M, Tanei T, Morimoto K, Shimazu K, Noguchi S, Tanaka K. Tetramethylrhodamine is an essential scaffold of azide probe in detecting cellular acrolein. Bioorg Med Chem 2019; 27:2228-2234. [DOI: 10.1016/j.bmc.2019.04.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/15/2019] [Accepted: 04/17/2019] [Indexed: 12/29/2022]
|
17
|
Phillips B, Szostak J, Titz B, Schlage WK, Guedj E, Leroy P, Vuillaume G, Martin F, Buettner A, Elamin A, Sewer A, Sierro N, Choukrallah MA, Schneider T, Ivanov NV, Teng C, Tung CK, Lim WT, Yeo YS, Vanscheeuwijck P, Peitsch MC, Hoeng J. A six-month systems toxicology inhalation/cessation study in ApoE -/- mice to investigate cardiovascular and respiratory exposure effects of modified risk tobacco products, CHTP 1.2 and THS 2.2, compared with conventional cigarettes. Food Chem Toxicol 2019; 126:113-141. [PMID: 30763686 DOI: 10.1016/j.fct.2019.02.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 01/24/2019] [Accepted: 02/04/2019] [Indexed: 02/06/2023]
Abstract
Smoking is one of the major modifiable risk factors in the development and progression of chronic obstructive pulmonary disease (COPD) and cardiovascular disease (CVD). Modified-risk tobacco products (MRTP) are being developed to provide substitute products for smokers who are unable or unwilling to quit, to lessen the smoking-related health risks. In this study, the ApoE-/- mouse model was used to investigate the impact of cigarette smoke (CS) from the reference cigarette 3R4F, or aerosol from two potential MRTPs based on the heat-not-burn principle, carbon heated tobacco product 1.2 (CHTP1.2) and tobacco heating system 2.2 (THS 2.2), on the cardiorespiratory system over a 6-month period. In addition, cessation or switching to CHTP1.2 after 3 months of CS exposure was assessed. A systems toxicology approach combining physiology, histology and molecular measurements was used to evaluate the impact of MRTP aerosols in comparison to CS. CHTP1.2 and THS2.2 aerosols, compared with CS, demonstrated lower impact on the cardiorespiratory system, including low to absent lung inflammation and emphysematous changes, and reduced atherosclerotic plaque formation. Molecular analyses confirmed the lower engagement of pathological mechanisms by MRTP aerosols than CS. Both cessation and switching to CHTP1.2 reduced the observed CS effects to almost sham exposure levels.
Collapse
Affiliation(s)
- Blaine Phillips
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore.
| | - Justyna Szostak
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Bjoern Titz
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | | | - Emmanuel Guedj
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Patrice Leroy
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Gregory Vuillaume
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Florian Martin
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | | | - Ashraf Elamin
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Alain Sewer
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Nicolas Sierro
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | | | - Thomas Schneider
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Nikolai V Ivanov
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Charles Teng
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore.
| | - Ching Keong Tung
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore.
| | - Wei Ting Lim
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore.
| | - Ying Shan Yeo
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore.
| | - Patrick Vanscheeuwijck
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Manuel C Peitsch
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Julia Hoeng
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| |
Collapse
|
18
|
Shen S, Zhang R, Zhang J, Wei Y, Guo Y, Su L, Chen F, Christiani DC. Welding fume exposure is associated with inflammation: a global metabolomics profiling study. Environ Health 2018; 17:68. [PMID: 30134906 PMCID: PMC6106842 DOI: 10.1186/s12940-018-0412-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 08/09/2018] [Indexed: 06/01/2023]
Abstract
BACKGROUND Increasing evidence suggests that welding fume exposure is associated with systemic inflammation. Although celluar metabolites may be associated with inflammation, there is limited information on metabolomic changes during welding fume exposure. Such changes may play an important role in the occurrence, development, and prevention of metal-associated diseases. We aim to investigate human metabolomics changes pre- and post-welding fume exposure. METHODS This study included 52 boilermakers totally. We collected plasma samples pre- and post-shift welding fume exposure and prepared samples using the automated MicroLab STAR® system. Metabolite concentrations were measured using ultra performance liquid chromatography - tandem mass spectrometer (UPLC-MS/MS) methods. Two-way analysis of variance was used to test the significance of metabolite changes with false discovery rate correction. RESULTS Analysis detected several metabolic changes after welding fume exposure, mainly involved in the lipid pathway [glucocorticoid class (cortisol, corticosterone, and cortisone), acylcarnitine class, and DiHOME species (9,10-DiHOME and 12,13-DiHOME)], amino acid utilization (isoleucine, proline and phenylalanine), and S-(3-hydroxypropyl) mercapturic acid (3-HPMA). These compounds are all associated with inflammation according to previous studies. Further, additive interaction effects linked smoking and 3-HPMA levels. In the metabolite set enrichment analysis for diseases, the top two disease-associated metabolite pathways were systemic inflammation-related diseases including rheumatoid arthritis and systemic lupus erythematosus. CONCLUSIONS This global metabolomics study shows evidence that metabolite changes during welding fume exposure are closely associated with systemic inflammation. The altered metabolites detected may be potential health monitoring biomarkers for boilermakers, especially for inflammation-related disease prevention.
Collapse
Affiliation(s)
- Sipeng Shen
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Ruyang Zhang
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Jinming Zhang
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Yongyue Wei
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Yichen Guo
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Li Su
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Feng Chen
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - David C Christiani
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA.
- China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
- Department of Medicine, Pulmonary and Critical Care Division, Massachusetts General Hospital and Harvard Medical School, 665 Huntington Avenue, Building I Room 1401, Boston, MA, 02115, USA.
| |
Collapse
|
19
|
Lin Y, Chen Z, Tang J, Cao P, Shi R. Acrolein Contributes to the Neuropathic Pain and Neuron Damage after Ischemic–Reperfusion Spinal Cord Injury. Neuroscience 2018; 384:120-130. [DOI: 10.1016/j.neuroscience.2018.05.029] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 04/04/2018] [Accepted: 05/20/2018] [Indexed: 11/28/2022]
|
20
|
Tully M, Tang J, Zheng L, Acosta G, Tian R, Hayward L, Race N, Mattson D, Shi R. Systemic Acrolein Elevations in Mice With Experimental Autoimmune Encephalomyelitis and Patients With Multiple Sclerosis. Front Neurol 2018; 9:420. [PMID: 29963001 PMCID: PMC6013577 DOI: 10.3389/fneur.2018.00420] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 05/22/2018] [Indexed: 01/27/2023] Open
Abstract
Demyelination and axonal injury are the key pathological processes in multiple sclerosis (MS), driven by inflammation and oxidative stress. Acrolein, a byproduct and instigator of oxidative stress, has been demonstrated as a neurotoxin in experimental autoimmune encephalomyelitis (EAE), an animal model of MS. However, due to the invasive nature of acrolein detection using immunoblotting techniques, the investigation of acrolein in MS has been limited to animal models. Recently, detection of a specific acrolein-glutathione metabolite, 3-HPMA, has been demonstrated in urine, enabling the noninvasive quantification of acrolein for the first time in humans with neurological disorders. In this study, we have demonstrated similar elevated levels of acrolein in both urine (3-HPMA) and in spinal cord tissue (acrolein-lysine adduct) in mice with EAE, which can be reduced through systemic application of acrolein scavenger hydralazine. Furthermore, using this approach we have demonstrated an increase of 3-HPMA in both the urine and serum of MS patients relative to controls. It is expected that this noninvasive acrolein detection could facilitate the investigation of the role of acrolein in the pathology of MS in human. It may also be used to monitor putative therapies aimed at suppressing acrolein levels, reducing severity of symptoms, and slowing progression as previously demonstrated in animal studies.
Collapse
Affiliation(s)
- Melissa Tully
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States.,Medical Scientist Training Program, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jonathan Tang
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States.,Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, United States
| | - Lingxing Zheng
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States.,Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, United States
| | - Glen Acosta
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, United States
| | - Ran Tian
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States.,Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, United States
| | - Lee Hayward
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Nicholas Race
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States.,Medical Scientist Training Program, Indiana University School of Medicine, Indianapolis, IN, United States
| | - David Mattson
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Riyi Shi
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States.,Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
21
|
Burcham PC. Carbonyl scavengers as pharmacotherapies in degenerative disease: Hydralazine repurposing and challenges in clinical translation. Biochem Pharmacol 2018; 154:397-406. [PMID: 29883705 DOI: 10.1016/j.bcp.2018.06.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 06/04/2018] [Indexed: 12/26/2022]
Abstract
During cellular metabolism, spontaneous oxidative damage to unsaturated lipids generates many electrophilic carbonyl compounds that readily attack cell macromolecules, forming adducts that are potential drivers of tissue dysfunction. Since such damage is heightened in many degenerative conditions, researchers have assessed the efficacy of nucleophilic carbonyl-trapping drugs in animal models of such disorders, anticipating that they will protect tissues by intercepting toxic lipid-derived electrophiles (LDEs) within cells. This Commentary explores recent animal evidence for carbonyl scavenger efficacy in two disparate yet significant conditions known to involve LDE production, namely spinal cord injury (SCI) and alcoholic liver disease (ALD). Primary emphasis is placed on studies that utilised hydralazine, a clinically-approved "broad-spectrum" scavenger known to trap multiple LDEs. In addition to reviewing recent studies of hydralazine efficacy in animal SCI and ALD models, the Commentary reviews new insights concerning novel lifespan- and healthspan-extending properties of hydralazine obtained during studies in model invertebrate organisms, since the mechanisms involved seem of likely benefit during the treatment of degenerative disease. Finally, noting that human translation of the histoprotective properties of hydralazine have been limited, the final section of the Commentary will address two obstacles that hamper clinical translation of LDE-trapping therapies while also suggesting potential strategies for overcoming these problems.
Collapse
Affiliation(s)
- Philip C Burcham
- Discipline of Pharmacology, School of Biomedical Science, The University of Western Australia, Crawley, WA 6007, Australia.
| |
Collapse
|
22
|
Ambaw A, Zheng L, Tambe MA, Strathearn KE, Acosta G, Hubers SA, Liu F, Herr SA, Tang J, Truong A, Walls E, Pond A, Rochet JC, Shi R. Acrolein-mediated neuronal cell death and alpha-synuclein aggregation: Implications for Parkinson's disease. Mol Cell Neurosci 2018; 88:70-82. [PMID: 29414104 DOI: 10.1016/j.mcn.2018.01.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 12/01/2017] [Accepted: 01/17/2018] [Indexed: 11/28/2022] Open
Abstract
Growing evidence suggests that oxidative stress plays a critical role in neuronal destruction characteristic of Parkinson's disease (PD). However, the molecular mechanisms of oxidative stress-mediated dopaminergic cell death are far from clear. In the current investigation, we tested the hypothesis that acrolein, an oxidative stress and lipid peroxidation (LPO) product, is a key factor in the pathogenesis of PD. Using a combination of in vitro, in vivo, and cell free models, coupled with anatomical, functional, and behavioral examination, we found that acrolein was elevated in 6-OHDA-injected rats, and behavioral deficits associated with 6-OHDA could be mitigated by the application of the acrolein scavenger hydralazine, and mimicked by injection of acrolein in healthy rats. Furthermore, hydralazine alleviated neuronal cell death elicited by 6-OHDA and another PD-related toxin, rotenone, in vitro. We also show that acrolein can promote the aggregation of alpha-synuclein, suggesting that alpha-synuclein self-assembly, a key pathological phenomenon in human PD, could play a role in neurotoxic effects of acrolein in PD models. These studies suggest that acrolein is involved in the pathogenesis of PD, and the administration of anti-acrolein scavengers such as hydralazine could represent a novel strategy to alleviate tissue damage and motor deficits associated with this disease.
Collapse
Affiliation(s)
- Abeje Ambaw
- Department of Basic Medical Sciences, School of Veterinary Medicine, Purdue University, United States
| | - Lingxing Zheng
- Department of Basic Medical Sciences, School of Veterinary Medicine, Purdue University, United States
| | - Mitali A Tambe
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, United States
| | - Katherine E Strathearn
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, United States
| | - Glen Acosta
- Department of Basic Medical Sciences, School of Veterinary Medicine, Purdue University, United States
| | - Scott A Hubers
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, United States
| | - Fang Liu
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, United States
| | - Seth A Herr
- Department of Basic Medical Sciences, School of Veterinary Medicine, Purdue University, United States; Purdue University Interdisciplinary Life Sciences Program (PULSe), Purdue University, United States
| | - Jonathan Tang
- Department of Basic Medical Sciences, School of Veterinary Medicine, Purdue University, United States; Weldon School of Biomedical Engineering, Purdue University, United States
| | - Alan Truong
- Weldon School of Biomedical Engineering, Purdue University, United States
| | - Elwood Walls
- Department of Basic Medical Sciences, School of Veterinary Medicine, Purdue University, United States
| | - Amber Pond
- Department of Basic Medical Sciences, School of Veterinary Medicine, Purdue University, United States
| | - Jean-Christophe Rochet
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, United States
| | - Riyi Shi
- Department of Basic Medical Sciences, School of Veterinary Medicine, Purdue University, United States; Weldon School of Biomedical Engineering, Purdue University, United States.
| |
Collapse
|
23
|
Tsou HH, Hsu WC, Fuh JL, Chen SP, Liu TY, Wang HT. Alterations in Acrolein Metabolism Contribute to Alzheimer’s Disease. J Alzheimers Dis 2017; 61:571-580. [DOI: 10.3233/jad-170736] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Han-Hsing Tsou
- Institute of Environmental and Occupational Health Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Wen-Chin Hsu
- Department of Pharmacology, National Yang-Ming University, Taipei, Taiwan
| | - Jong-Ling Fuh
- Faculty of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
- Brain Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Shih-Pin Chen
- Faculty of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
- Brain Research Center, National Yang-Ming University, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Tsung-Yun Liu
- Institute of Environmental and Occupational Health Sciences, National Yang-Ming University, Taipei, Taiwan
- Institute of Food Safety and Health Risk Assessment, National Yang-Ming University, Taipei, Taiwan
| | - Hsiang-Tsui Wang
- Department of Pharmacology, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
24
|
Butler B, Acosta G, Shi R. Exogenous Acrolein intensifies sensory hypersensitivity after spinal cord injury in rat. J Neurol Sci 2017; 379:29-35. [DOI: 10.1016/j.jns.2017.05.039] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 04/30/2017] [Accepted: 05/21/2017] [Indexed: 10/19/2022]
|
25
|
Yao L, Wu YT, Tian GX, Xia CQ, Zhang F, Zhang W. Acrolein Scavenger Hydralazine Prevents Streptozotocin-Induced Painful Diabetic Neuropathy and Spinal Neuroinflammation in Rats. Anat Rec (Hoboken) 2017; 300:1858-1864. [PMID: 28598552 DOI: 10.1002/ar.23618] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 02/06/2017] [Accepted: 03/02/2017] [Indexed: 01/12/2023]
Abstract
Diabetes-induced neuropathic pain (DNP) substantially influences people's life qualities. Hyperglycemia-induced excess free radicals have been considered as the most critical mechanisms underlying DNP. As an unsaturated aldehyde and a reactive product of lipid peroxidation, acrolein plays critical roles in diabetic nephropathy and inflammatory pain. We sought to determine whether acrolein is involved in DNP in this study. Diabetes was induced by a single intraperitoneal (i.p.) injection of 60 mg/kg streptozotocin (STZ). An acrolein scavenger hydralazine (5 mg/kg) was administered through a daily injection for 4 weeks, starting immediately within 30 min after STZ injection. Western blot showed that hydralazine could effectively inhibit STZ-induced upregulation of acrolein in the spinal dorsal horn on day 7-28 after STZ injection. Behavioral tests showed that STZ injection induced significant mechanical allodynia and thermal hyperalgesia, which could be alleviated by hydralazine. Immunofluorescent histochemistry and Western blot showed that STZ induced significant microglial activation. ELISA data indicated upregulation of inflammatory cytokines IL-1β and TNF-α expression in the spinal dorsal horn. Furthermore, hydralazine effectively attenuated microglial activation and expression of inflammatory mediators. Our data indicate that acrolein might be involved in the development of neuroinflammation and behavioral consequences of DNP. Anat Rec, 2017. © 2017 Wiley Periodicals, Inc. Anat Rec, 300:1858-1864, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Lu Yao
- Department of Forth Cadre, PLA Army General Hospital, No. 5 Nanmencang, Dongcheng District, Beijing, 10070, China
| | - Yun-Tao Wu
- Department of Forth Cadre, PLA Army General Hospital, No. 5 Nanmencang, Dongcheng District, Beijing, 10070, China
| | - Guo-Xiang Tian
- Department of Forth Cadre, PLA Army General Hospital, No. 5 Nanmencang, Dongcheng District, Beijing, 10070, China
| | - Chang-Quan Xia
- Department of Forth Cadre, PLA Army General Hospital, No. 5 Nanmencang, Dongcheng District, Beijing, 10070, China
| | - Feng Zhang
- Department of Forth Cadre, PLA Army General Hospital, No. 5 Nanmencang, Dongcheng District, Beijing, 10070, China
| | - Wei Zhang
- Department of Forth Cadre, PLA Army General Hospital, No. 5 Nanmencang, Dongcheng District, Beijing, 10070, China
| |
Collapse
|
26
|
Cruz-Haces M, Tang J, Acosta G, Fernandez J, Shi R. Pathological correlations between traumatic brain injury and chronic neurodegenerative diseases. Transl Neurodegener 2017; 6:20. [PMID: 28702179 PMCID: PMC5504572 DOI: 10.1186/s40035-017-0088-2] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 06/27/2017] [Indexed: 02/06/2023] Open
Abstract
Traumatic brain injury is among the most common causes of death and disability in youth and young adults. In addition to the acute risk of morbidity with moderate to severe injuries, traumatic brain injury is associated with a number of chronic neurological and neuropsychiatric sequelae including neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. However, despite the high incidence of traumatic brain injuries and the established clinical correlation with neurodegeneration, the causative factors linking these processes have not yet been fully elucidated. Apart from removal from activity, few, if any prophylactic treatments against post-traumatic brain injury neurodegeneration exist. Therefore, it is imperative to understand the pathophysiological mechanisms of traumatic brain injury and neurodegeneration in order to identify potential factors that initiate neurodegenerative processes. Oxidative stress, neuroinflammation, and glutamatergic excitotoxicity have previously been implicated in both secondary brain injury and neurodegeneration. In particular, reactive oxygen species appear to be key in mediating molecular insult in neuroinflammation and excitotoxicity. As such, it is likely that post injury oxidative stress is a key mechanism which links traumatic brain injury to increased risk of neurodegeneration. Consequently, reactive oxygen species and their subsequent byproducts may serve as novel fluid markers for identification and monitoring of cellular damage. Furthermore, these reactive species may further serve as a suitable therapeutic target to reduce the risk of post-injury neurodegeneration and provide long term quality of life improvements for those suffering from traumatic brain injury.
Collapse
Affiliation(s)
- Marcela Cruz-Haces
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907 USA
| | - Jonathan Tang
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907 USA
| | - Glen Acosta
- Department of Basic Medical Sciences, Purdue University, West Lafayette, USA
| | - Joseph Fernandez
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907 USA
| | - Riyi Shi
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907 USA
- Department of Basic Medical Sciences, Purdue University, West Lafayette, USA
| |
Collapse
|
27
|
Tian R, Shi R. Dimercaprol is an acrolein scavenger that mitigates acrolein-mediated PC-12 cells toxicity and reduces acrolein in rat following spinal cord injury. J Neurochem 2017; 141:708-720. [PMID: 28301040 DOI: 10.1111/jnc.14025] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 02/23/2017] [Accepted: 03/07/2017] [Indexed: 12/22/2022]
Abstract
Acrolein is one of the most toxic byproducts of lipid peroxidation, and it has been shown to be associated with multiple pathological processes in trauma and diseases, including spinal cord injury, multiple sclerosis, and Alzheimer's disease. Therefore, suppressing acrolein using acrolein scavengers has been suggested as a novel strategy of neuroprotection. In an effort to identify effective acrolein scavengers, we have confirmed that dimercaprol, which possesses thiol functional groups, could bind and trap acrolein. We demonstrated the reaction between acrolein and dimercaprol in an abiotic condition by nuclear magnetic resonance spectroscopy. Specifically, dimercaprol is able to bind to both the carbon double bond and aldehyde group of acrolein. Its acrolein scavenging capability was further demonstrated by in vitro results that showed that dimercaprol could significantly protect PC-12 cells from acrolein-mediated cell death in a dose-dependent manner. Furthermore, dimercaprol, when applied systemically through intraperitoneal injection, could significantly reduce acrolein contents in spinal cord tissue following a spinal cord contusion injury in rats, a condition known to have elevated acrolein concentration. Taken together, dimercaprol may be an effective acrolein scavenger and a viable candidate for acrolein detoxification.
Collapse
Affiliation(s)
- Ran Tian
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana, USA.,Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Riyi Shi
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana, USA.,Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
28
|
Urinary 3-hydroxypropyl mercapturic acid (3-HPMA) concentrations in dogs with acute spinal cord injury due to intervertebral disc herniation. Vet J 2016; 219:12-14. [PMID: 28093103 DOI: 10.1016/j.tvjl.2016.11.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 11/10/2016] [Accepted: 11/22/2016] [Indexed: 11/23/2022]
Abstract
The aim of this study was to investigate urinary 3-hydroxypropyl mercapturic acid (3-HPMA), a metabolite of acrolein, as a novel biomarker in acute spinal cord injury (ASCI) due to intervertebral disc herniation in dogs. Urine from 10 client-owned dogs with ASCI collected at presentation and 10 control dogs was analyzed for 3-HPMA. The median urinary 3-HPMA concentration in ASCI dogs was significantly higher than in control dogs, but was not correlated with the severity of ASCI. The median urinary 3-HPMA concentration in intact dogs was higher than in neutered dogs. Higher urinary 3-HPMA concentrations in dogs after ASCI support a role for acrolein, a cytotoxic by-product of lipid peroxidation, in canine ASCI. Urinary 3-HPMA could be used as a biomarker in future clinical trials to measure the effect of therapeutic intervention of reducing acrolein after ASCI.
Collapse
|
29
|
Pradipta AR, Saigitbatalova E, Takamatsu M, Kurbangalieva A, Tanaka K. Progress in the Development of Reaction-Based Sensors for Detection of Acrolein in Biological Samples. BIONANOSCIENCE 2016. [DOI: 10.1007/s12668-016-0266-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
30
|
Chen WY, Zhang J, Ghare S, Barve S, McClain C, Joshi-Barve S. Acrolein Is a Pathogenic Mediator of Alcoholic Liver Disease and the Scavenger Hydralazine Is Protective in Mice. Cell Mol Gastroenterol Hepatol 2016; 2:685-700. [PMID: 28119953 PMCID: PMC5042858 DOI: 10.1016/j.jcmgh.2016.05.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 05/17/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Alcoholic liver disease (ALD) remains a major cause of morbidity and mortality, with no Food and Drug Administration-approved therapy. Chronic alcohol consumption causes a pro-oxidant environment and increases hepatic lipid peroxidation, with acrolein being the most reactive/toxic by-product. This study investigated the pathogenic role of acrolein in hepatic endoplasmic reticulum (ER) stress, steatosis, and injury in experimental ALD, and tested acrolein elimination/scavenging (using hydralazine) as a potential therapy in ALD. METHODS In vitro (rat hepatoma H4IIEC cells) and in vivo (chronic+binge alcohol feeding in C57Bl/6 mice) models were used to examine alcohol-induced acrolein accumulation and consequent hepatic ER stress, apoptosis, and injury. In addition, the potential protective effects of the acrolein scavenger, hydralazine, were examined both in vitro and in vivo. RESULTS Alcohol consumption/metabolism resulted in hepatic accumulation of acrolein-protein adducts, by up-regulation of cytochrome P4502E1 and alcohol dehydrogenase, and down-regulation of glutathione-s-transferase-P, which metabolizes/detoxifies acrolein. Alcohol-induced acrolein adduct accumulation led to hepatic ER stress, proapoptotic signaling, steatosis, apoptosis, and liver injury; however, ER-protective/adaptive responses were not induced. Notably, direct exposure to acrolein in vitro mimicked the in vivo effects of alcohol, indicating that acrolein mediates the adverse effects of alcohol. Importantly, hydralazine, a known acrolein scavenger, protected against alcohol-induced ER stress and liver injury, both in vitro and in mice. CONCLUSIONS Our study shows the following: (1) alcohol consumption triggers pathologic ER stress without ER adaptation/protection; (2) alcohol-induced acrolein is a potential therapeutic target and pathogenic mediator of hepatic ER stress, cell death, and injury; and (3) removal/clearance of acrolein by scavengers may have therapeutic potential in ALD.
Collapse
Key Words
- ADH, alcohol dehydrogenase
- ALD, alcoholic liver disease
- ALDH, aldehyde dehydrogenase
- ALT, alanine aminotransferase
- AST, aspartate aminotransferase
- ATF, activating transcription factor
- Apoptosis
- CHOP
- CHOP, CCAAT/enhancer-binding protein homologous protein
- CYP2E1, cytochrome P4502E1
- ER, endoplasmic reticulum
- FDP-lysine, Nε-(3-formyl-3,4-dehydropiperidino)lysine
- GRP, glucose regulated protein
- GSTP, glutathione-s-transferase-Pi
- IRE1, inositol-requiring enzyme 1
- JNK, cJun N-terminal kinase
- LPO, lipid peroxidation
- Lipid Peroxidation
- NIAAA, National Institute on Alcohol Abuse and Alcoholism
- PERK, protein kinase RNA-like endoplasmic reticulum kinase
- PUFA, polyunsaturated fatty acids
- TRAF, TNF receptor-associated factor
- TUNEL, terminal deoxynucleotidyl transferase–mediated deoxyuridine triphosphate nick-end labeling
- Therapeutic
- UPR, unfolded protein response
- XBP1, X-box binding protein-1
- mRNA, messenger RNA
- siRNA, small interfering RNA
Collapse
Affiliation(s)
- Wei-Yang Chen
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky
- Alcohol Research Center, University of Louisville, Louisville, Kentucky
| | - Jingwen Zhang
- Alcohol Research Center, University of Louisville, Louisville, Kentucky
- Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Smita Ghare
- Alcohol Research Center, University of Louisville, Louisville, Kentucky
- Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Shirish Barve
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky
- Alcohol Research Center, University of Louisville, Louisville, Kentucky
- Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Craig McClain
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky
- Alcohol Research Center, University of Louisville, Louisville, Kentucky
- Department of Medicine, University of Louisville, Louisville, Kentucky
- Robley Rex Veterans Affairs Medical Center, University of Louisville, Louisville, Kentucky
| | - Swati Joshi-Barve
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky
- Alcohol Research Center, University of Louisville, Louisville, Kentucky
- Department of Medicine, University of Louisville, Louisville, Kentucky
- Correspondence Address correspondence to: Swati Joshi-Barve, PhD, Departments of Medicine, and Pharmacology and Toxicology, University of Louisville, 505 South Hancock Street, Room 505 Clinical Translational Research Building, Louisville, Kentucky 40202. fax: (502) 852-8927.Departments of Medicine, and Pharmacology and ToxicologyUniversity of Louisville505 South Hancock StreetRoom 505 Clinical Translational Research BuildingLouisvilleKentucky 40202
| |
Collapse
|
31
|
Chen Z, Park J, Butler B, Acosta G, Vega-Alvarez S, Zheng L, Tang J, McCain R, Zhang W, Ouyang Z, Cao P, Shi R. Mitigation of sensory and motor deficits by acrolein scavenger phenelzine in a rat model of spinal cord contusive injury. J Neurochem 2016; 138:328-38. [PMID: 27060873 DOI: 10.1111/jnc.13639] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 03/15/2016] [Accepted: 04/07/2016] [Indexed: 12/13/2022]
Abstract
Currently there are no effective therapies available for the excruciating neuropathic pain that develops after spinal cord injuries (SCI). As such, a great deal of effort is being put into the investigation of novel therapeutic targets that can alleviate this pain. One such target is acrolein, a highly reactive aldehyde produced as a byproduct of oxidative stress and inflammation that is capable of activating the transient receptor potential ankyrin 1 (TRPA1) cation channel, known to be involved in the transmission and propagation of chronic neuropathic pain. One anti-acrolein agent, hydralazine, has already been shown to reduce neuropathic pain behaviors and offer neuroprotection after SCI. This study investigates another acrolein scavenger, phenelzine, for its possible role of alleviating sensory hypersensitivity through acrolein suppression. The results show that phenelzine is indeed capable of attenuating neuropathic pain behaviors in acute, delayed, and chronic administration schedules after injury in a rat model of SCI. In addition, upon the comparison of hydralazine to phenelzine, both acrolein scavengers displayed a dose-dependent response in the reduction of acrolein in vivo. Finally, phenelzine proved capable of providing locomotor function recovery and neuroprotection of spinal cord tissue when administered immediately after injury for 2 weeks. These results indicate that phenelzine may be an effective treatment for neuropathic pain after SCI and likely a viable alternative to hydralazine. We have shown that phenelzine can attenuate neuropathic pain behavior in acute, delayed, and chronic administration in post-SCI rats. This was accompanied by a dose-dependent reduction in an acrolein metabolite in urine and an acrolein adduct in spinal cord tissue, and the suppression of TRPA1 over-expression in central and peripheral locations post-trauma. Acrolein scavenging might be a novel therapeutic strategy to reduce post-SCI neuropathic pain.
Collapse
Affiliation(s)
- Zhe Chen
- Department of Orthopedics, Rui-Jin Hospital, School of Medicine, Shanghai Jiao-tong University, Institute of Trauma and Orthopedics, Shanghai, China.,Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopedics, Rui-Jin Hospital, Shanghai Jiao-tong University School of Medicine, Shanghai, China
| | - Jonghyuck Park
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA.,Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Breanne Butler
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Glen Acosta
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - Sasha Vega-Alvarez
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - Lingxing Zheng
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA.,Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Jonathan Tang
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA.,Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Robyn McCain
- Purdue Translational Pharmacology, Bindley Bioscience Center, Purdue University, West Lafayette, IN, USA
| | - Wenpeng Zhang
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Zheng Ouyang
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA.,Department of Chemistry, Purdue University, West Lafayette, IN, USA
| | - Peng Cao
- Department of Orthopedics, Rui-Jin Hospital, School of Medicine, Shanghai Jiao-tong University, Institute of Trauma and Orthopedics, Shanghai, China.,Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopedics, Rui-Jin Hospital, Shanghai Jiao-tong University School of Medicine, Shanghai, China
| | - Riyi Shi
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA.,Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
32
|
Yan R, Page JC, Shi R. Acrolein-mediated conduction loss is partially restored by K⁺ channel blockers. J Neurophysiol 2015; 115:701-10. [PMID: 26581866 DOI: 10.1152/jn.00467.2015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 11/18/2015] [Indexed: 11/22/2022] Open
Abstract
Acrolein-mediated myelin damage is thought to be a critical mechanism leading to conduction failure following neurotrauma and neurodegenerative diseases. The exposure and activation of juxtaparanodal voltage-gated K(+) channels due to myelin damage leads to conduction block, and K(+) channel blockers have long been studied as a means for restoring axonal conduction in spinal cord injury (SCI) and multiple sclerosis (MS). In this study, we have found that 100 μM K(+) channel blockers 4-aminopyridine-3-methanol (4-AP-3-MeOH), and to a lesser degree 4-aminopyridine (4-AP), can significantly restore compound action potential (CAP) conduction in spinal cord tissue following acrolein-mediated myelin damage using a well-established ex vivo SCI model. In addition, 4-AP-3-MeOH can effectively restore CAP conduction in acrolein-damaged axons with a range of concentrations from 0.1 to 100 μM. We have also shown that while both compounds at 100 μM showed no preference of small- and large-caliber axons when restoring CAP conduction, 4-AP-3-MeOH, unlike 4-AP, is able to augment CAP amplitude while causing little change in axonal responsiveness measured in refractory periods and response to repetitive stimuli. In a prior study, we show that 4-AP-3-MeOH was able to functionally rescue mechanically injured axons. In this investigation, we conclude that 4-AP-3-MeOH is an effective K(+) channel blocker in restoring axonal conduction following both primary (physical) and secondary (chemical) insults. These findings also suggest that 4-AP-3-MeOH is a viable alternative of 4-AP for treating myelin damage and improving function following central nervous system trauma and neurodegenerative diseases.
Collapse
Affiliation(s)
- Rui Yan
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana; and
| | - Jessica C Page
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana; and
| | - Riyi Shi
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana; and Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana
| |
Collapse
|
33
|
Park J, Zheng L, Acosta G, Vega-Alvarez S, Chen Z, Muratori B, Cao P, Shi R. Acrolein contributes to TRPA1 up-regulation in peripheral and central sensory hypersensitivity following spinal cord injury. J Neurochem 2015; 135:987-97. [PMID: 26365991 DOI: 10.1111/jnc.13352] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Revised: 07/27/2015] [Accepted: 09/04/2015] [Indexed: 11/30/2022]
Abstract
Acrolein, an endogenous aldehyde, has been shown to be involved in sensory hypersensitivity after rat spinal cord injury (SCI), for which the pathogenesis is unclear. Acrolein can directly activate a pro-algesic transient receptor protein ankyrin 1 (TRPA1) channel that exists in sensory neurons. Both acrolein and TRPA1 mRNA are elevated post SCI, which contributes to the activation of TRPA1 by acrolein and consequently, neuropathic pain. In the current study, we further showed that, post-SCI elevation of TRPA1 mRNA exists not only in dorsal root ganglias but also in both peripheral (paw skin) and central endings of primary afferent nerves (dorsal horn of spinal cord). This is the first indication that pain signaling can be over-amplified in the peripheral skin by elevated expressions of TRPA1 following SCI, in addition over-amplification previously seen in the spinal cord and dorsal root ganglia. Furthermore, we show that acrolein alone, in the absence of physical trauma, could lead to the elevation of TRPA1 mRNA at various locations when injected to the spinal cord. In addition, post-SCI elevation of TRPA1 mRNA could be mitigated using acrolein scavengers. Both of these attributes support the critical role of acrolein in elevating TRPA1 expression through gene regulation. Taken together, these data indicate that acrolein is likely a critical causal factor in heightening pain sensation post-SCI, through both the direct binding of TRPA1 receptor, and also by boosting the expression of TRPA1. Finally, our data also further support the notion that acrolein scavenging may be an effective therapeutic approach to alleviate neuropathic pain after SCI. We propose that the trauma-mediated elevation of acrolein causes neuropathic pain through at least two mechanisms: acrolein stimulates the production of transient receptor protein ankyrin 1 (TRPA1) in both central and peripheral locations, and it activates TRPA1 channels directly. Therefore, acrolein appears to be a critical factor in the pathogenesis of post-SCI sensory hypersensitivity, becoming a novel therapeutic target to relieve both acute and chronic post-SCI neuropathic pain.
Collapse
Affiliation(s)
- Jonghyuck Park
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana, USA.,Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Lingxing Zheng
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana, USA.,Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Glen Acosta
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana, USA
| | - Sasha Vega-Alvarez
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana, USA
| | - Zhe Chen
- Department of Orthopedics, Rui-Jin Hospital, School of Medicine, Shanghai Jiao-tong University, Institute of Trauma and Orthopedics, Shanghai, China
| | - Breanne Muratori
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Peng Cao
- Department of Orthopedics, Rui-Jin Hospital, School of Medicine, Shanghai Jiao-tong University, Institute of Trauma and Orthopedics, Shanghai, China
| | - Riyi Shi
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana, USA.,Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
34
|
Moghe A, Ghare S, Lamoreau B, Mohammad M, Barve S, McClain C, Joshi-Barve S. Molecular mechanisms of acrolein toxicity: relevance to human disease. Toxicol Sci 2015; 143:242-55. [PMID: 25628402 DOI: 10.1093/toxsci/kfu233] [Citation(s) in RCA: 341] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Acrolein, a highly reactive unsaturated aldehyde, is a ubiquitous environmental pollutant and its potential as a serious environmental health threat is beginning to be recognized. Humans are exposed to acrolein per oral (food and water), respiratory (cigarette smoke, automobile exhaust, and biocide use) and dermal routes, in addition to endogenous generation (metabolism and lipid peroxidation). Acrolein has been suggested to play a role in several disease states including spinal cord injury, multiple sclerosis, Alzheimer's disease, cardiovascular disease, diabetes mellitus, and neuro-, hepato-, and nephro-toxicity. On the cellular level, acrolein exposure has diverse toxic effects, including DNA and protein adduction, oxidative stress, mitochondrial disruption, membrane damage, endoplasmic reticulum stress, and immune dysfunction. This review addresses our current understanding of each pathogenic mechanism of acrolein toxicity, with emphasis on the known and anticipated contribution to clinical disease, and potential therapies.
Collapse
Affiliation(s)
- Akshata Moghe
- *Department of Pharmacology and Toxicology, Department of Medicine and Robley Rex VAMC, Louisville, Kentucky 40202
| | - Smita Ghare
- *Department of Pharmacology and Toxicology, Department of Medicine and Robley Rex VAMC, Louisville, Kentucky 40202
| | - Bryan Lamoreau
- *Department of Pharmacology and Toxicology, Department of Medicine and Robley Rex VAMC, Louisville, Kentucky 40202
| | - Mohammad Mohammad
- *Department of Pharmacology and Toxicology, Department of Medicine and Robley Rex VAMC, Louisville, Kentucky 40202
| | - Shirish Barve
- *Department of Pharmacology and Toxicology, Department of Medicine and Robley Rex VAMC, Louisville, Kentucky 40202 *Department of Pharmacology and Toxicology, Department of Medicine and Robley Rex VAMC, Louisville, Kentucky 40202
| | - Craig McClain
- *Department of Pharmacology and Toxicology, Department of Medicine and Robley Rex VAMC, Louisville, Kentucky 40202 *Department of Pharmacology and Toxicology, Department of Medicine and Robley Rex VAMC, Louisville, Kentucky 40202 *Department of Pharmacology and Toxicology, Department of Medicine and Robley Rex VAMC, Louisville, Kentucky 40202
| | - Swati Joshi-Barve
- *Department of Pharmacology and Toxicology, Department of Medicine and Robley Rex VAMC, Louisville, Kentucky 40202 *Department of Pharmacology and Toxicology, Department of Medicine and Robley Rex VAMC, Louisville, Kentucky 40202
| |
Collapse
|
35
|
Walls MK, Race N, Zheng L, Vega-Alvarez SM, Acosta G, Park J, Shi R. Structural and biochemical abnormalities in the absence of acute deficits in mild primary blast-induced head trauma. J Neurosurg 2015; 124:675-86. [PMID: 26295915 DOI: 10.3171/2015.1.jns141571] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
OBJECTIVE Blast-induced neurotrauma (BINT), if not fatal, is nonetheless potentially crippling. It can produce a wide array of acute symptoms in moderate-to-severe exposures, but mild BINT (mBINT) is characterized by the distinct absence of acute clinical abnormalities. The lack of observable indications for mBINT is particularly alarming, as these injuries have been linked to severe long-term psychiatric and degenerative neurological dysfunction. Although the long-term sequelae of BINT are extensively documented, the underlying mechanisms of injury remain poorly understood, impeding the development of diagnostic and treatment strategies. The primary goal of this research was to recapitulate primary mBINT in rodents in order to facilitate well-controlled, long-term investigations of blast-induced pathological neurological sequelae and identify potential mechanisms by which ongoing damage may occur postinjury. METHODS A validated, open-ended shock tube model was used to deliver blast overpressure (150 kPa) to anesthetized rats with body shielding and head fixation, simulating the protective effects of military-grade body armor and isolating a shock wave injury from confounding systemic injury responses, head acceleration, and other elements of explosive events. Evans Blue-labeled albumin was used to visualize blood-brain barrier (BBB) compromise at 4 hours postinjury. Iba1 staining was used to visualize activated microglia and infiltrating macrophages in areas of peak BBB compromise. Acrolein, a potent posttraumatic neurotoxin, was quantified in brain tissue by immunoblotting and in urine through liquid chromatography with tandem mass spectrometry at 1, 2, 3, and 5 days postinjury. Locomotor behavior, motor performance, and short-term memory were assessed with open field, rotarod, and novel object recognition (NOR) paradigms at 24 and 48 hours after the blast. RESULTS Average speed, maximum speed, and distance traveled in an open-field exploration paradigm did not show significant differences in performance between sham-injured and mBINT rats. Likewise, rats with mBINT did not exhibit deficits in maximum revolutions per minute or total run time in a rotarod paradigm. Short-term memory was also unaffected by mBINT in an NOR paradigm. Despite lacking observable motor or cognitive deficits in the acute term, blast-injured rats displayed brain acrolein levels that were significantly elevated for at least 5 days, and acrolein's glutathione-reduced metabolite, 3-HPMA, was present in urine for 2 days after injury. Additionally, mBINT brain tissue demonstrated BBB damage 4 hours postinjury and colocalized neuroinflammatory changes 24 hours postinjury. CONCLUSIONS This model highlights mBINT's potential for underlying detrimental physical and biochemical alterations despite the lack of apparent acute symptoms and, by recapitulating the human condition, represents an avenue for further examining the pathophysiology of mBINT. The sustained upregulation of acrolein for days after injury suggests that acrolein may be an upstream player potentiating ongoing postinjury damage and neuroinflammation. Ultimately, continued research with this model may lead to diagnostic and treatment mechanisms capable of preventing or reducing the severity of long-term neurological dysfunction following mBINT.
Collapse
Affiliation(s)
- Michael K Walls
- Department of Basic Medical Sciences, College of Veterinary Medicine; and
| | - Nicholas Race
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana
| | - Lingxing Zheng
- Department of Basic Medical Sciences, College of Veterinary Medicine; and.,Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana
| | | | - Glen Acosta
- Department of Basic Medical Sciences, College of Veterinary Medicine; and
| | - Jonghyuck Park
- Department of Basic Medical Sciences, College of Veterinary Medicine; and.,Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana
| | - Riyi Shi
- Department of Basic Medical Sciences, College of Veterinary Medicine; and.,Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana
| |
Collapse
|
36
|
Shi R, Page JC, Tully M. Molecular mechanisms of acrolein-mediated myelin destruction in CNS trauma and disease. Free Radic Res 2015; 49:888-95. [PMID: 25879847 DOI: 10.3109/10715762.2015.1021696] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Myelin is a critical component of the nervous system facilitating efficient propagation of electrical signals and thus communication between the central and peripheral nervous systems and the organ systems that they innervate throughout the body. In instances of neurotrauma and neurodegenerative disease, injury to myelin is a prominent pathological feature responsible for conduction deficits, and leaves axons vulnerable to damage from noxious compounds. Although the pathological mechanisms underlying myelin loss have yet to be fully characterized, oxidative stress (OS) appears to play a prominent role. Specifically, acrolein, a neurotoxic aldehyde that is both a product and an instigator of OS, has been observed in studies to elicit demyelination through calcium-independent and -dependent mechanisms and also by affecting glutamate uptake and promoting excitotoxicity. Furthermore, pharmacological scavenging of acrolein has demonstrated a neuroprotective effect in animal disease models, by conserving myelin's structural integrity and alleviating functional deficits. This evidence indicates that acrolein may be a key culprit of myelin damage while acrolein scavenging could potentially be a promising therapeutic approach for patients suffering from nervous system trauma and disease.
Collapse
Affiliation(s)
- R Shi
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University , West Lafayette, IN , USA
| | | | | |
Collapse
|
37
|
Tully M, Zheng L, Acosta G, Tian R, Shi R. Acute systemic accumulation of acrolein in mice by inhalation at a concentration similar to that in cigarette smoke. Neurosci Bull 2014; 30:1017-1024. [PMID: 25446876 DOI: 10.1007/s12264-014-1480-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Accepted: 06/18/2014] [Indexed: 11/28/2022] Open
Abstract
Cigarette smoke is an important environmental factor associated with a wide array of public health concerns. Acrolein, a component of tobacco smoke and a known toxin to various cell types, may be a key pathological factor mediating the adverse effects linked with tobacco smoke. Although acrolein is known to accumulate in the respiratory system after acute nasal exposure, it is not clear if it accumulates systemically, and less is known in the nervous system. The aim of this study was to assess the degree of acrolein accumulation in the circulation and in the spinal cord following acute acrolein inhalation in mice. Using a laboratory-fabricated inhalation chamber, we found elevated urinary 3-HPMA, an acrolein metabolite, and increased acrolein adducts in the spinal cord after weeks of nasal exposure to acrolein at a concentration similar to that in tobacco smoke. The data indicated that acrolein is absorbed into the circulatory system and some enters the nervous system. It is expected that these findings may facilitate further studies to probe the pathological role of acrolein in the nervous system resulting from smoke and other external sources.
Collapse
Affiliation(s)
- Melissa Tully
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA.,Indiana University School of Medicine, Indianapolis, IN, USA
| | - Lingxing Zheng
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA.,Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Glen Acosta
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - Ran Tian
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA.,Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Riyi Shi
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA. .,Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
38
|
Toxicological assessment of kretek cigarettes Part 3: kretek and American-blended cigarettes, inhalation toxicity. Regul Toxicol Pharmacol 2014; 70 Suppl 1:S26-40. [PMID: 25455226 DOI: 10.1016/j.yrtph.2014.09.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 09/29/2014] [Indexed: 11/20/2022]
Abstract
A typical Indonesian kretek cigarette brand and an experimental kretek reference cigarette were compared to the reference cigarette 2R4F in two 90-day inhalation studies. Male and female rats were exposed nose-only to mainstream smoke for 6 hours daily, for 90 consecutive days. Biological endpoints were assessed according to OECD guideline 413, with special emphasis on respiratory tract histopathology and on lung inflammation (broncho-alveolar lavage fluid levels of neutrophils, macrophages and lymphocytes). Histopathological alterations included: in the nose, hyperplasia and squamous metaplasia of the respiratory epithelium and squamous metaplasia and atrophy of the olfactory epithelium; in the larynx, epithelial squamous metaplasia and hyperplasia; in the lungs, accumulation of macrophages in alveoli and goblet cell hyperplasia in bronchial epithelium. The findings were qualitatively consistent with observations from previous similar studies on conventional cigarettes. Compared to 2R4F cigarette, however, kretek smoke exposure was associated with a pronounced attenuation of pulmonary inflammation and less severe histopathological changes in the respiratory tract. Neutrophilic inflammation was also significantly lower (>70%). These results are consistent with the observations made on smoke chemistry and in vitro toxicology. They do not support any increased toxicity of the smoke of kretek cigarettes compared to conventional American-blended cigarettes.
Collapse
|
39
|
Park J, Muratori B, Shi R. Acrolein as a novel therapeutic target for motor and sensory deficits in spinal cord injury. Neural Regen Res 2014; 9:677-83. [PMID: 25206871 PMCID: PMC4146266 DOI: 10.4103/1673-5374.131564] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2014] [Indexed: 12/18/2022] Open
Abstract
IN THE HOURS TO WEEKS FOLLOWING TRAUMATIC SPINAL CORD INJURIES (SCI), BIOCHEMICAL PROCESSES ARE INITIATED THAT FURTHER DAMAGE THE TISSUE WITHIN AND SURROUNDING THE INITIAL INJURY SITE: a process termed secondary injury. Acrolein, a highly reactive unsaturated aldehyde, has been shown to play a major role in the secondary injury by contributing significantly to both motor and sensory deficits. In particular, efforts have been made to elucidate the mechanisms of acrolein-mediated damage at the cellular level and the resulting paralysis and neuropathic pain. In this review, we will highlight the recent developments in the understanding of the mechanisms of acrolein in motor and sensory dysfunction in animal models of SCI. We will also discuss the therapeutic benefits of using acrolein scavengers to attenuate acrolein-mediated neuronal damage following SCI.
Collapse
Affiliation(s)
- Jonghyuck Park
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA ; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Breanne Muratori
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Riyi Shi
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA ; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
40
|
Tully M, Zheng L, Shi R. Acrolein detection: potential theranostic utility in multiple sclerosis and spinal cord injury. Expert Rev Neurother 2014; 14:679-85. [PMID: 24831349 DOI: 10.1586/14737175.2014.918849] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Oxidative stress has been implicated as a major pathological process underlying CNS disease and trauma. More specifically, acrolein, an unsaturated aldehyde, produced by way of lipid peroxidation, has been shown to play a crucial role in initiating and perpetuating detrimental effects associated with multiple sclerosis and spinal cord injury. In light of these findings, quantification of acrolein levels both systemically and locally could allow for the use of acrolein as a biomarker to aid in diagnosis and guide treatment regimens. The three main approaches currently available are acrolein derivatization followed by LC/GC-MS, application of an acrolein antibody and subsequent immunoblotting, and the 3-hydroxypropylmercapturic acid-based method. Of these three strategies, the 3-hydroxypropylmercapturic acid-based method is the least invasive allowing for rapid translation of acrolein detection into a clinical setting.
Collapse
Affiliation(s)
- Melissa Tully
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | | | | |
Collapse
|
41
|
Park J, Zheng L, Marquis A, Walls M, Duerstock B, Pond A, Vega-Alvarez S, Wang H, Ouyang Z, Shi R. Neuroprotective role of hydralazine in rat spinal cord injury-attenuation of acrolein-mediated damage. J Neurochem 2013; 129:339-49. [PMID: 24286176 DOI: 10.1111/jnc.12628] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 11/25/2013] [Accepted: 11/26/2013] [Indexed: 12/20/2022]
Abstract
Acrolein, an α,β-unsaturated aldehyde and a reactive product of lipid peroxidation, has been suggested as a key factor in neural post-traumatic secondary injury in spinal cord injury (SCI), mainly based on in vitro and ex vivo evidence. Here, we demonstrate an increase of acrolein up to 300%; the elevation lasted at least 2 weeks in a rat SCI model. More importantly, hydralazine, a known acrolein scavenger can provide neuroprotection when applied systemically. Besides effectively reducing acrolein, hydralazine treatment also resulted in significant amelioration of tissue damage, motor deficits, and neuropathic pain. This effect was further supported by demonstrating the ability of hydralazine to reach spinal cord tissue at a therapeutic level following intraperitoneal application. This suggests that hydralazine is an effective neuroprotective agent not only in vitro, but in a live animal model of SCI as well. Finally, the role of acrolein in SCI was further validated by the fact that acrolein injection into the spinal cord caused significant SCI-like tissue damage and motor deficits. Taken together, available evidence strongly suggests a critical causal role of acrolein in the pathogenesis of spinal cord trauma. Since acrolein has been linked to a variety of illness and conditions, we believe that acrolein-scavenging measures have the potential to be expanded significantly ensuring a broad impact on human health.
Collapse
Affiliation(s)
- Jonghyuck Park
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana, USA; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|