1
|
Cizmecioglu A, Eryavuz Onmaz D, Senturk S, Askin D, Unlu A, Korkmaz H, Gungor G. Kynurenine Pathway Dysregulation and Pain Perception in Acute Pancreatitis: Has the Connection Unraveled? Neurosci Lett 2024; 837:137902. [PMID: 39029612 DOI: 10.1016/j.neulet.2024.137902] [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: 01/02/2024] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 07/21/2024]
Abstract
AIM Tryptophan (TRP), an essential amino acid, undergoes catabolism through various pathways. Notably, the kynurenine pathway (KP), constituting one of these pathways, exhibits a unidirectional impact on immune response and energy metabolism. Nonetheless, its influence on pain sensation is characterized by biphasic dynamics. This study aims to scrutinize the influence of the KP pathway on pain sensation, particularly within the context of pancreatic inflammation. METHODS Our prospective case-control study involved individuals diagnosed with acute pancreatitis and a control group matched for gender and age. The patient cohort was subsequently subdivided into severe and non-severe subgroups. To assess metabolites within KP, two blood samples were collected from the patient cohort, one at the time of diagnosis and another during the recovery phase. Furthermore, for pain quantification, daily pain scores utilizing the Visual Analog Scale (VAS) were extracted from the patients' medical records. RESULTS The study incorporated 30 patients along with an equivalent number of controls. A noticeable distinction was evident between the patient and control groups, characterized by an increase in kynurenine levels and a decrease in the tryptophan/kynurenine ratio. Throughout the process of disease recovery, a uniform decrease was observed in all KP metabolites, excluding 3-Hydroxykynurenine. Elevated levels of Kynurenic acid (KYNA) were correlated with increased pain scores. Critically, no apparent distinctions in KP metabolites were discerned concerning pain severity in patients with comorbidities characterized by neural involvement. CONCLUSION Based on our results, the kynurenine pathway (KP) is activated in instances of acute pancreatitis. Elevated levels of KYNA were found to be associated with heightened pain scores. The operative stages within the KP responsible for pain modulation are impaired in cases characterized by neuropathy-induced pain sensation.
Collapse
Affiliation(s)
- Ahmet Cizmecioglu
- Department of Internal Medicine, Faculty of Medicine, Selcuk University, Konya, Turkiye.
| | - Duygu Eryavuz Onmaz
- Department of Biochemistry, Faculty of Medicine, Selcuk University, Konya, Turkiye.
| | - Suleyman Senturk
- Division of Gastroenterology, Department of Internal Medicine, Faculty of Medicine, Selcuk University, Konya, Turkiye.
| | - Dudu Askin
- Department of Internal Medicine, Alanya Sifa Private Medical Center, Antalya, Turkiye.
| | - Ali Unlu
- Department of Biochemistry, Faculty of Medicine, Selcuk University, Konya, Turkiye.
| | - Huseyin Korkmaz
- Division of Gastroenterology, Department of Internal Medicine, Faculty of Medicine, Selcuk University, Konya, Turkiye.
| | - Gokhan Gungor
- Division of Gastroenterology, Department of Internal Medicine, Faculty of Medicine, Selcuk University, Konya, Turkiye.
| |
Collapse
|
2
|
Hazrati E, Eftekhar SP, Mosaed R, Shiralizadeh Dini S, Namazi M. Understanding the kynurenine pathway: A narrative review on its impact across chronic pain conditions. Mol Pain 2024; 20:17448069241275097. [PMID: 39093627 PMCID: PMC11331475 DOI: 10.1177/17448069241275097] [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: 05/07/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024] Open
Abstract
Chronic pain is a debilitating symptom with a significant negative impact on the quality of life and socioeconomic status, particularly among adults and the elderly. Major Depressive Disorder (MDD) stands out as one of the most important comorbid disorders accompanying chronic pain. The kynurenine pathway serves as the primary route for tryptophan degradation and holds critical significance in various biological processes, including the regulation of neurotransmitters, immune responses, cancer development, metabolism, and inflammation. This review encompasses key research studies related to the kynurenine pathway in the context of headache, neuropathic pain, gastrointestinal disorders, fibromyalgia, chronic fatigue syndrome, and MDD. Various metabolites produced in the kynurenine pathway, such as kynurenic acid and quinolinic acid, exhibit neuroprotective and neurotoxic effects, respectively. Recent studies have highlighted the significant involvement of kynurenine and its metabolites in the pathophysiology of pain. Moreover, pharmacological interventions targeting the regulation of the kynurenine pathway have shown therapeutic promise in pain management. Understanding the underlying mechanisms of this pathway presents an opportunity for developing personalized, innovative, and non-opioid approaches to pain treatment. Therefore, this narrative review explores the role of the kynurenine pathway in various chronic pain disorders and its association with depression and chronic pain.
Collapse
Affiliation(s)
- Ebrahim Hazrati
- Trauma and Surgery Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Seyed Parsa Eftekhar
- Trauma and Surgery Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Reza Mosaed
- Trauma and Surgery Research Center, AJA University of Medical Sciences, Tehran, Iran
| | | | - Mehrshad Namazi
- Trauma and Surgery Research Center, AJA University of Medical Sciences, Tehran, Iran
- Clinical Biomechanics and Ergonomics Research Center, AJA University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Shen J, Zhao W, Cheng J, Cheng J, Zhao L, Dai C, Fu Y, Li B, Chen Z, Shi D, Li H, Deng Y. Lipopolysaccharide accelerates tryptophan degradation in the ovary and the derivative kynurenine disturbs hormone biosynthesis and reproductive performance. JOURNAL OF HAZARDOUS MATERIALS 2023; 458:131988. [PMID: 37418963 DOI: 10.1016/j.jhazmat.2023.131988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 07/01/2023] [Accepted: 07/01/2023] [Indexed: 07/09/2023]
Abstract
Lipopolysaccharide (LPS), also known as endotoxin, is a component of the outer membrane of gram-negative bacteria. LPS is released into the surrounding environment during bacterial death and lysis. Due to its chemical and thermal stability, LPS can be detected anywhere and easily exposed to humans and animals. Previous studies have shown that LPS causes hormonal imbalances, ovarian failure, and infertility in mammals. However, the potential mechanisms remain unclear. In this study, we investigated the effects and mechanisms of LPS on tryptophan degradation, both in vivo and in vitro. The effects of kynurenine, a tryptophan derivative, on granulosa cell function and reproductive performance were explored. Results showed that p38, NF-κB, and JNK signaling pathways were involved in LPS-induced Ido1 expressions and kynurenine accumulation. Furthermore, the kynurenine decreased estradiol production, but increased granulosa cell proliferation. In vivo, experiments showed that kynurenine decreased estradiol and FSH production and inhibited ovulation and corpus luteum formation. Additionally, pregnancy and offspring survival rates decreased considerably after kynurenine treatment. Our findings suggest that kynurenine accumulation disrupts hormone secretion, ovulation, corpus luteal formation, and reproductive performance in mammals.
Collapse
Affiliation(s)
- Jie Shen
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi Key Laboratory of Animal Breeding and Disease Control, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Weimin Zhao
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Juanru Cheng
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi Key Laboratory of Animal Breeding and Disease Control, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Jinhua Cheng
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Lei Zhao
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi Key Laboratory of Animal Breeding and Disease Control, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Chaohui Dai
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Yanfeng Fu
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Bixia Li
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Zhe Chen
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Deshun Shi
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi Key Laboratory of Animal Breeding and Disease Control, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Hui Li
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China.
| | - Yanfei Deng
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi Key Laboratory of Animal Breeding and Disease Control, College of Animal Science and Technology, Guangxi University, Nanning 530004, China.
| |
Collapse
|
4
|
Ciapała K, Pawlik K, Ciechanowska A, Mika J, Rojewska E. Effect of pharmacological modulation of the kynurenine pathway on pain-related behavior and opioid analgesia in a mouse model of neuropathic pain. Toxicol Appl Pharmacol 2023; 461:116382. [PMID: 36681127 DOI: 10.1016/j.taap.2023.116382] [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: 05/01/2022] [Revised: 10/26/2022] [Accepted: 01/14/2023] [Indexed: 01/19/2023]
Abstract
Dysfunction of the central nervous system are accompanied by changes in tryptophan metabolism, with the kynurenine pathway (KP) being the main route of its catabolism. Recently, KP metabolites, which are collectively called kynurenines, have become an area of intense research due to their ability to directly and indirectly affect a variety of classic neurotransmitter systems. However, the significance of KP in neuropathic pain is still poorly understood. Therefore, we designed several experiments to verify changes in the mRNA levels of KP enzymes in parallel with other factors related to this metabolic route after chronic constriction injury of the sciatic nerve (CCI model) in mice. The analysis revealed an increase in, Kmo, Kynu and Haoo mRNA levels in the spinal cord on the 7th day after CCI, while Kat1, Kat2, Tdo2, Ido2 and Qprt mRNA levels remain unchanged. Subsequent pharmacological studies provided evidence that modulation of KP by single intrathecal administration of 1-D-MT, UPF468 or L-kynurenine attenuates mechanical and thermal hypersensitivity and increases the effectiveness of selected opioids in mice as measured on day 7 after CCI. Moreover, our results provide the first evidence that the injection of L-kynurenine preceded by UPF468 (KMO inhibitor) is more effective at reducing hypersensitivity in animals with neuropathic pain. Importantly, L-kynurenine also exerts an analgesic effect after intravenous injections, which is enhanced by the administration of minocycline, an inhibitor of microglial activation. Additionally, L-kynurenine administered intrathecally and intravenously enhances analgesia evoked by all tested opioids (morphine, buprenorphine and oxycodone). Overall, our results indicate that the modulation of KP at different levels might be a new pharmacological tool in neuropathy management.
Collapse
Affiliation(s)
- Katarzyna Ciapała
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, Krakow, Poland
| | - Katarzyna Pawlik
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, Krakow, Poland
| | - Agata Ciechanowska
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, Krakow, Poland
| | - Joanna Mika
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, Krakow, Poland
| | - Ewelina Rojewska
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, Krakow, Poland.
| |
Collapse
|
5
|
Stone TW, Clanchy FIL, Huang YS, Chiang NY, Darlington LG, Williams RO. An integrated cytokine and kynurenine network as the basis of neuroimmune communication. Front Neurosci 2022; 16:1002004. [PMID: 36507331 PMCID: PMC9729788 DOI: 10.3389/fnins.2022.1002004] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 10/31/2022] [Indexed: 11/25/2022] Open
Abstract
Two of the molecular families closely associated with mediating communication between the brain and immune system are cytokines and the kynurenine metabolites of tryptophan. Both groups regulate neuron and glial activity in the central nervous system (CNS) and leukocyte function in the immune system, although neither group alone completely explains neuroimmune function, disease occurrence or severity. This essay suggests that the two families perform complementary functions generating an integrated network. The kynurenine pathway determines overall neuronal excitability and plasticity by modulating glutamate receptors and GPR35 activity across the CNS, and regulates general features of immune cell status, surveillance and tolerance which often involves the Aryl Hydrocarbon Receptor (AHR). Equally, cytokines and chemokines define and regulate specific populations of neurons, glia or immune system leukocytes, generating more specific responses within restricted CNS regions or leukocyte populations. In addition, as there is a much larger variety of these compounds, their homing properties enable the superimposition of dynamic variations of cell activity upon local, spatially limited, cell populations. This would in principle allow the targeting of potential treatments to restricted regions of the CNS. The proposed synergistic interface of 'tonic' kynurenine pathway affecting baseline activity and the superimposed 'phasic' cytokine system would constitute an integrated network explaining some features of neuroimmune communication. The concept would broaden the scope for the development of new treatments for disorders involving both the CNS and immune systems, with safer and more effective agents targeted to specific CNS regions.
Collapse
Affiliation(s)
- Trevor W. Stone
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom,*Correspondence: Trevor W. Stone,
| | - Felix I. L. Clanchy
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - Yi-Shu Huang
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - Nien-Yi Chiang
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - L. Gail Darlington
- Department of Internal Medicine, Ashtead Hospital, Ashtead, United Kingdom
| | - Richard O. Williams
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
6
|
Simpson S, Mclellan R, Wellmeyer E, Matalon F, George O. Drugs and Bugs: The Gut-Brain Axis and Substance Use Disorders. J Neuroimmune Pharmacol 2022; 17:33-61. [PMID: 34694571 PMCID: PMC9074906 DOI: 10.1007/s11481-021-10022-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 09/06/2021] [Indexed: 02/07/2023]
Abstract
Substance use disorders (SUDs) represent a significant public health crisis. Worldwide, 5.4% of the global disease burden is attributed to SUDs and alcohol use, and many more use psychoactive substances recreationally. Often associated with comorbidities, SUDs result in changes to both brain function and physiological responses. Mounting evidence calls for a precision approach for the treatment and diagnosis of SUDs, and the gut microbiome is emerging as a contributor to such disorders. Over the last few centuries, modern lifestyles, diets, and medical care have altered the health of the microbes that live in and on our bodies; as we develop, our diets and lifestyle dictate which microbes flourish and which microbes vanish. An increase in antibiotic treatments, with many antibiotic interventions occurring early in life during the microbiome's normal development, transforms developing microbial communities. Links have been made between the microbiome and SUDs, and the microbiome and conditions that are often comorbid with SUDs such as anxiety, depression, pain, and stress. A better understanding of the mechanisms influencing behavioral changes and drug use is critical in developing novel treatments for SUDSs. Targeting the microbiome as a therapeutic and diagnostic tool is a promising avenue of exploration. This review will provide an overview of the role of the gut-brain axis in a wide range of SUDs, discuss host and microbe pathways that mediate changes in the brain's response to drugs, and the microbes and related metabolites that impact behavior and health within the gut-brain axis.
Collapse
Affiliation(s)
- Sierra Simpson
- Department of Psychiatry, University of California San Diego, La Jolla, San Diego, CA, 92093, US.
| | - Rio Mclellan
- Department of Psychiatry, University of California San Diego, La Jolla, San Diego, CA, 92093, US
| | - Emma Wellmeyer
- Department of Psychiatry, University of California San Diego, La Jolla, San Diego, CA, 92093, US
| | - Frederic Matalon
- Department of Psychiatry, University of California San Diego, La Jolla, San Diego, CA, 92093, US
| | - Olivier George
- Department of Psychiatry, University of California San Diego, La Jolla, San Diego, CA, 92093, US
| |
Collapse
|
7
|
Qi Q, Wang XX, Li JL, Chen YQ, Chang JR, Xi J, Lü HZ, Zhang YX. Neuroprotective Effects of the Pannexin-1 Channel Inhibitor: Probenecid on Spinal Cord Injury in Rats. Front Mol Neurosci 2022; 15:848185. [PMID: 35663270 PMCID: PMC9162172 DOI: 10.3389/fnmol.2022.848185] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 04/01/2022] [Indexed: 11/13/2022] Open
Abstract
Proinflammatory immune cell subsets constitute the majority in the local microenvironment after spinal cord injury (SCI), leading to secondary pathological injury. Previous studies have demonstrated that inflammasomes act as an important part of the inflammatory process after SCI. Probenecid, an inhibitor of the Pannexin-1 channel, can inhibit the activation of inflammasomes. This article focuses on the effects of probenecid on the local immune microenvironment, histopathology, and behavior of SCI. Our data show that probenecid inhibited the expression and activation of nucleotide-binding oligomerization domain receptor pyrindomain-containing 1 (NLRP1), apoptosis-associated speck-like protein containing a CARD (ASC) and caspase-1, interleukin-1β (IL-1β), and caspase-3 proteins associated with inflammasomes, thereby suppressing the proportion of M1 cells. And consequently, probenecid reduced the lesion area and demyelination in SCI. Moreover, the drug increased the survival of motor neurons, which resulted in tissue repair and improved locomotor function in the injured SC. Altogether, existing studies indicated that probenecid can alleviate inflammation by blocking Pannexin-1 channels to inhibit the expression of caspase-1 and IL-1β, which in turn restores the balance of immune cell subsets and exerts neuroprotective effects in rats with SCI.
Collapse
Affiliation(s)
- Qi Qi
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
- School of Basic Medicine, Bengbu Medical College, Bengbu, China
| | - Xiao-Xuan Wang
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
- School of Laboratory Medicine, Bengbu Medical College, Bengbu, China
| | - Jing-Lu Li
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Yu-Qing Chen
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
- School of Laboratory Medicine, Bengbu Medical College, Bengbu, China
| | - Jian-Rong Chang
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Jin Xi
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - He-Zuo Lü
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
- School of Laboratory Medicine, Bengbu Medical College, Bengbu, China
- *Correspondence: He-Zuo Lü,
| | - Yu-Xin Zhang
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
- School of Laboratory Medicine, Bengbu Medical College, Bengbu, China
- Yu-Xin Zhang,
| |
Collapse
|
8
|
Gusso D, Cruz FF, Fritsch PM, da Silva Gobbo MO, Morrone FB, Bonan CD. Pannexin channel 1, P2X7 receptors, and Dimethyl Sulfoxide mediate pain responses in zebrafish. Behav Brain Res 2022; 423:113786. [DOI: 10.1016/j.bbr.2022.113786] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/19/2022] [Accepted: 02/01/2022] [Indexed: 12/15/2022]
|
9
|
Mechanisms Underlying the Selective Therapeutic Efficacy of Carbamazepine for Attenuation of Trigeminal Nerve Injury Pain. J Neurosci 2021; 41:8991-9007. [PMID: 34446571 DOI: 10.1523/jneurosci.0547-21.2021] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 11/21/2022] Open
Abstract
Different peripheral nerve injuries cause neuropathic pain through distinct mechanisms. Even the site of injury may impact underlying mechanisms, as indicated by the clinical finding that the antiseizure drug carbamazepine (CBZ) relieves pain because of compression injuries of trigeminal but not somatic nerves. We leveraged this observation in the present study hypothesizing that because CBZ blocks voltage-gated sodium channels (VGSCs), its therapeutic selectivity reflects differences between trigeminal and somatic nerves with respect to injury-induced changes in VGSCs. CBZ diminished ongoing and evoked pain behavior in rats with chronic constriction injury (CCI) to the infraorbital nerve (ION) but had minimal effect in rats with sciatic nerve CCI. This difference in behavior was associated with a selective increase in the potency of CBZ-induced inhibition of compound action potentials in the ION, an effect mirrored in human trigeminal versus somatic nerves. The increase in potency was associated with a selective increase in the efficacy of the NaV1.1 channel blocker ICA-121431 and NaV1.1 protein in the ION, but no change in NaV1.1 mRNA in trigeminal ganglia. Importantly, local ICA-121431 administration reversed ION CCI-induced hypersensitivity. Our results suggest a novel therapeutic target for the treatment of trigeminal neuropathic pain.SIGNIFICANCE STATEMENT This study is based on evidence of differences in pain and its treatment depending on whether the pain is above (trigeminal) or below (somatic) the neck, as well as evidence that voltage-gated sodium channels (VGSCs) may contribute to these differences. The focus of the present study was on channels underlying action potential propagation in peripheral nerves. There were differences between somatic and trigeminal nerves in VGSC subtypes underlying action potential propagation both in the absence and presence of injury. Importantly, because the local block of NaV1.1 in the trigeminal nerve reverses nerve injury-induced mechanical hypersensitivity, the selective upregulation of NaV1.1 in trigeminal nerves suggests a novel therapeutic target for the treatment of pain associated with trigeminal nerve injury.
Collapse
|
10
|
The Kynurenine Pathway as a Potential Target for Neuropathic Pain Therapy Design: From Basic Research to Clinical Perspectives. Int J Mol Sci 2021; 22:ijms222011055. [PMID: 34681715 PMCID: PMC8537209 DOI: 10.3390/ijms222011055] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/08/2021] [Accepted: 10/09/2021] [Indexed: 12/20/2022] Open
Abstract
Accumulating evidence suggests the key role of the kynurenine pathway (KP) of the tryptophan metabolism in the pathogenesis of several diseases. Despite extensive research aimed at clarifying the mechanisms underlying the development and maintenance of neuropathic pain, the roles of KP metabolites in this process are still not fully known. Although the function of the peripheral KP has been known for several years, it has only recently been acknowledged that its metabolites within the central nervous system have remarkable consequences related to physiology and behavior. Both the products and metabolites of the KP are involved in the pathogenesis of pain conditions. Apart from the neuroactive properties of kynurenines, the KP regulates several neurotransmitter systems in direct or indirect ways. Some neuroactive metabolites are known to have neuroprotective properties (kynurenic acid, nicotinamide adenine dinucleotide cofactor), while others are toxic (3-hydroxykynurenine, quinolinic acid). Numerous animal models show that modulation of the KP may turn out to be a viable target for the treatment of diseases. Importantly, some compounds that affect KP enzymes are currently described to possess analgesic properties. Additionally, kynurenine metabolites may be useful for assessing response to therapy or as biomarkers in therapeutic monitoring. The following review describes the molecular site of action and changes in the levels of metabolites of the kynurenine pathway in the pathogenesis of various conditions, with a particular emphasis on their involvement in neuropathy. Moreover, the potential clinical implications of KP modulation in chronic pain therapy as well as the directions of new research initiatives are discussed.
Collapse
|
11
|
Tanaka M, Török N, Tóth F, Szabó Á, Vécsei L. Co-Players in Chronic Pain: Neuroinflammation and the Tryptophan-Kynurenine Metabolic Pathway. Biomedicines 2021; 9:biomedicines9080897. [PMID: 34440101 PMCID: PMC8389666 DOI: 10.3390/biomedicines9080897] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/18/2021] [Accepted: 07/19/2021] [Indexed: 01/09/2023] Open
Abstract
Chronic pain is an unpleasant sensory and emotional experience that persists or recurs more than three months and may extend beyond the expected time of healing. Recently, nociplastic pain has been introduced as a descriptor of the mechanism of pain, which is due to the disturbance of neural processing without actual or potential tissue damage, appearing to replace a concept of psychogenic pain. An interdisciplinary task force of the International Association for the Study of Pain (IASP) compiled a systematic classification of clinical conditions associated with chronic pain, which was published in 2018 and will officially come into effect in 2022 in the 11th revision of the International Statistical Classification of Diseases and Related Health Problems (ICD-11) by the World Health Organization. ICD-11 offers the option for recording the presence of psychological or social factors in chronic pain; however, cognitive, emotional, and social dimensions in the pathogenesis of chronic pain are missing. Earlier pain disorder was defined as a condition with chronic pain associated with psychological factors, but it was replaced with somatic symptom disorder with predominant pain in the Diagnostic and Statistical Manual of Mental Disorders, 5th Edition (DSM-5) in 2013. Recently clinical nosology is trending toward highlighting neurological pathology of chronic pain, discounting psychological or social factors in the pathogenesis of pain. This review article discusses components of the pain pathway, the component-based mechanisms of pain, central and peripheral sensitization, roles of chronic inflammation, and the involvement of tryptophan-kynurenine pathway metabolites, exploring the participation of psychosocial and behavioral factors in central sensitization of diseases progressing into the development of chronic pain, comorbid diseases that commonly present a symptom of chronic pain, and psychiatric disorders that manifest chronic pain without obvious actual or potential tissue damage.
Collapse
Affiliation(s)
- Masaru Tanaka
- MTA-SZTE, Neuroscience Research Group, Semmelweis u. 6, H-6725 Szeged, Hungary; (M.T.); (N.T.); (F.T.)
- Interdisciplinary Excellence Centre, Department of Neurology, Faculty of Medicine, University of Szeged, H-6725 Szeged, Hungary;
| | - Nóra Török
- MTA-SZTE, Neuroscience Research Group, Semmelweis u. 6, H-6725 Szeged, Hungary; (M.T.); (N.T.); (F.T.)
- Interdisciplinary Excellence Centre, Department of Neurology, Faculty of Medicine, University of Szeged, H-6725 Szeged, Hungary;
| | - Fanni Tóth
- MTA-SZTE, Neuroscience Research Group, Semmelweis u. 6, H-6725 Szeged, Hungary; (M.T.); (N.T.); (F.T.)
| | - Ágnes Szabó
- Interdisciplinary Excellence Centre, Department of Neurology, Faculty of Medicine, University of Szeged, H-6725 Szeged, Hungary;
| | - László Vécsei
- MTA-SZTE, Neuroscience Research Group, Semmelweis u. 6, H-6725 Szeged, Hungary; (M.T.); (N.T.); (F.T.)
- Interdisciplinary Excellence Centre, Department of Neurology, Faculty of Medicine, University of Szeged, H-6725 Szeged, Hungary;
- Correspondence: ; Tel.: +36-62-545-351
| |
Collapse
|
12
|
Spekker E, Laborc KF, Bohár Z, Nagy-Grócz G, Fejes-Szabó A, Szűcs M, Vécsei L, Párdutz Á. Effect of dural inflammatory soup application on activation and sensitization markers in the caudal trigeminal nucleus of the rat and the modulatory effects of sumatriptan and kynurenic acid. J Headache Pain 2021; 22:17. [PMID: 33789568 PMCID: PMC8011387 DOI: 10.1186/s10194-021-01229-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 03/15/2021] [Indexed: 01/12/2023] Open
Abstract
Background The topical inflammatory soup can model the inflammation of the dura mater causing hypersensitivity and activation of the trigeminal system, a phenomenon present in migraineurs. Calcitonin gene-related peptide, transient receptor potential vanilloid-1 receptor, and neuronal nitric oxide synthase are important in the sensitization process there. 5-HT1B/1D receptor agonists, triptans are used as a treatment of migraine. Kynurenic acid an NMDA antagonist can act on structures involved in trigeminal activation. Aim We investigated the effect of inflammatory soup induced dural inflammation on the calcitonin gene-related peptide, transient receptor potential vanilloid-1 receptor, and neuronal nitric oxide synthase levels in the caudal trigeminal nucleus. We also tested whether pretreatment with a well-known antimigraine drug, such as sumatriptan and kynurenic acid, a compound with a different mechanism of action, can affect these changes and if their modulatory effects are comparable. Material and methods After subcutaneous sumatriptan or intraperitoneal kynurenic acid the dura mater of adult male Sprague-Dawley rats (n = 72) was treated with inflammatory soup or its vehicle (synthetic interstitial fluid). Two and a half or four hours later perfusion was performed and the caudal trigeminal nucleus was removed for immunohistochemistry. Results and conclusion Inflammatory soup increased calcitonin gene-related peptide, transient receptor potential vanilloid-1 receptor, and neuronal nitric oxide synthase in the caudal trigeminal nucleus compared to placebo, which was attenuated by sumatriptan and kynurenic acid. This suggests the involvement of 5-HT1B/1D and NMDA receptors in neurogenic inflammation development of the dura and thus in migraine attacks.
Collapse
Affiliation(s)
- Eleonóra Spekker
- Department of Neurology, Interdisciplinary Excellence Center, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis utca 6, Szeged, H-6725, Hungary
| | - Klaudia Flóra Laborc
- Department of Neurology, Interdisciplinary Excellence Center, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis utca 6, Szeged, H-6725, Hungary
| | - Zsuzsanna Bohár
- Department of Neurology, Interdisciplinary Excellence Center, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis utca 6, Szeged, H-6725, Hungary.,MTA-SZTE Neuroscience Research Group, Szeged, Hungary
| | - Gábor Nagy-Grócz
- Department of Neurology, Interdisciplinary Excellence Center, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis utca 6, Szeged, H-6725, Hungary.,Faculty of Health Sciences and Social Studies, University of Szeged, Szeged, Hungary
| | | | - Mónika Szűcs
- Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
| | - László Vécsei
- Department of Neurology, Interdisciplinary Excellence Center, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis utca 6, Szeged, H-6725, Hungary. .,MTA-SZTE Neuroscience Research Group, Szeged, Hungary.
| | - Árpád Párdutz
- Department of Neurology, Interdisciplinary Excellence Center, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis utca 6, Szeged, H-6725, Hungary
| |
Collapse
|
13
|
Morales-Puerto N, Giménez-Gómez P, Pérez-Hernández M, Abuin-Martínez C, Gil de Biedma-Elduayen L, Vidal R, Gutiérrez-López MD, O'Shea E, Colado MI. Addiction and the kynurenine pathway: A new dancing couple? Pharmacol Ther 2021; 223:107807. [PMID: 33476641 DOI: 10.1016/j.pharmthera.2021.107807] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/05/2021] [Indexed: 12/15/2022]
Abstract
Drug use poses a serious threat to health systems throughout the world and the number of consumers rises relentlessly every year. The kynurenine pathway, main pathway of tryptophan degradation, has drawn interest in this field due to its relationship with addictive behaviour. Recently it has been confirmed that modulation of kynurenine metabolism at certain stages of the pathway can reduce, prevent or abolish drug seeking-like behaviours in studies with several different drugs. In this review, we present an up-to-date summary of the evidences of a relationship between drug use and the kynurenine pathway, both the alterations of the pathway due to drug use as well as modulation of the pathway as a potential approach to treat drug addiction. The review discusses ethanol, nicotine, cannabis, amphetamines, cocaine and opioids and new prospects in the drug research field are proposed.
Collapse
Affiliation(s)
- Nuria Morales-Puerto
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain; Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain; Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Pablo Giménez-Gómez
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain; Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain; Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Mercedes Pérez-Hernández
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain; Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain; Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Cristina Abuin-Martínez
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain; Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain; Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Leticia Gil de Biedma-Elduayen
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain; Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain; Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Rebeca Vidal
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain; Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain; Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - María Dolores Gutiérrez-López
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain; Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain; Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Esther O'Shea
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain; Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain; Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain.
| | - María Isabel Colado
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain; Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain; Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain.
| |
Collapse
|
14
|
The Role of the Kynurenine Signaling Pathway in Different Chronic Pain Conditions and Potential Use of Therapeutic Agents. Int J Mol Sci 2020; 21:ijms21176045. [PMID: 32842609 PMCID: PMC7503462 DOI: 10.3390/ijms21176045] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/11/2020] [Accepted: 08/19/2020] [Indexed: 12/11/2022] Open
Abstract
Tryptophan (TRP) is an essential, aromatic amino acid catabolized by indoleamine 2,3-dioxygenase (IDO) and tryptophan 2,3-dioxygenase (TDO) enzymes into kynurenine. The IDO enzyme is expressed in peripheral tissues and the central nervous system. Another enzyme of interest in the kynurenine signaling pathway is kynurenine 3-monooxygenase (KMO). The purpose of this review is to discuss the role of TRP and the kynurenine signaling pathway in different chronic pain patients. The IDO-1, IDO-2, and KMO enzymes and the kynurenine metabolite have been shown to be involved in the pathogenesis of neuropathic pain and other painful conditions (migraine, cluster headache, etc.) as well as depressive behavior. We highlighted the analgesic potential of novel agents targeting the enzymes of the kynurenine signaling pathway to explore their efficacy in both future basic science and transitional studies. Upcoming studies conducted on animal models will need to take into consideration the differences in TRP metabolism between human and non-human species. Since chronic painful conditions and depression have common pathophysiological patterns, and the kynurenine signaling pathway is involved in both of them, future clinical studies should aim to have outcomes targeting not only pain, but also functionality, mood changes, and quality of life.
Collapse
|
15
|
Zhang YX, Wang SN, Chen J, Hu JG, Lü HZ. A transcriptomic study of probenecid on injured spinal cords in mice. PeerJ 2020; 8:e8367. [PMID: 31921518 PMCID: PMC6944129 DOI: 10.7717/peerj.8367] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 12/06/2019] [Indexed: 11/20/2022] Open
Abstract
Background Recent studies have found that probenecid has neuroprotective and reparative effects on central nervous system injuries. However, its effect on genome-wide transcription in acute spinal cord injury (SCI) remains unknown. In the present study, RNA sequencing (RNA-Seq) is used to analyze the effect of probenecid on the local expression of gene transcription 8 h after spinal injury. Methods An Infinite Horizon impactor was used to perform contusive SCI in mice. The SCI model was made by using a rod (1.3 mm diameter) with a force of 50 Kdynes. Sham-operated mice only received a laminectomy without contusive injury. The injured mice were randomly assigned into either the control (SCI_C) or probenecid injection (SCI_P) group. In the latter group, the probenecid drug was intraperitoneally injected (0.5 mg/kg) immediately following injury. Eight hours after the injury or laminectomy, the spinal cords were removed from the mice in both groups. The total RNAs were extracted and purified for library preparation and transcriptome sequencing. Differential gene expressions (DEGs) of the three groups-sham, SCI_C and SCI_P-were analyzed using a DESeq software. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis of DEGs were performed using a GOseq R package and KOBAS software. Real-time quantitative reverse-transcriptase polymerase chain reaction was used to validate RNA-Seq results. Results RNA-Seq showed that, compared to the SCI_C group, the number of DEGs was 641 in the SCI_P group (286 upregulated and 355 downregulated). According to GO analysis, DEGs were most enriched in extracellular matrix (ECM), collagen trimer, protein bounding and sequence specific DNA binding. KEGG analysis showed that the most enriched pathways included: cell adhesion molecules, Leukocyte transendothelial migration, ECM-receptor interactions, PI3K-Akt signaling pathways, hematopoietic cell lineages, focal adhesions, the Rap1 signaling pathway, etc. The sequence data have been deposited into the Sequence Read Archive (https://www.ncbi.nlm.nih.gov/sra/PRJNA554464).
Collapse
Affiliation(s)
- Yu-Xin Zhang
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Department of Biochemistry and Molecular Biology, Bengbu Medical College, Bengbu, China
| | - Sai-Nan Wang
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Jing Chen
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Jian-Guo Hu
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - He-Zuo Lü
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| |
Collapse
|
16
|
Rojewska E, Ciapała K, Mika J. Kynurenic acid and zaprinast diminished CXCL17-evoked pain-related behaviour and enhanced morphine analgesia in a mouse neuropathic pain model. Pharmacol Rep 2019; 71:139-148. [DOI: 10.1016/j.pharep.2018.10.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 09/03/2018] [Accepted: 10/02/2018] [Indexed: 12/23/2022]
|
17
|
Hagos FT, Empey PE, Wang P, Ma X, Poloyac SM, Bayır H, Kochanek PM, Bell MJ, Clark RSB. Exploratory Application of Neuropharmacometabolomics in Severe Childhood Traumatic Brain Injury. Crit Care Med 2018; 46:1471-1479. [PMID: 29742587 PMCID: PMC6095742 DOI: 10.1097/ccm.0000000000003203] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVES To employ metabolomics-based pathway and network analyses to evaluate the cerebrospinal fluid metabolome after severe traumatic brain injury in children and the capacity of combination therapy with probenecid and N-acetylcysteine to impact glutathione-related and other pathways and networks, relative to placebo treatment. DESIGN Analysis of cerebrospinal fluid obtained from children enrolled in an Institutional Review Board-approved, randomized, placebo-controlled trial of a combination of probenecid and N-acetylcysteine after severe traumatic brain injury (Trial Registration NCT01322009). SETTING Thirty-six-bed PICU in a university-affiliated children's hospital. PATIENTS AND SUBJECTS Twelve children 2-18 years old after severe traumatic brain injury and five age-matched control subjects. INTERVENTION Probenecid (25 mg/kg) and N-acetylcysteine (140 mg/kg) or placebo administered via naso/orogastric tube. MEASUREMENTS AND MAIN RESULTS The cerebrospinal fluid metabolome was analyzed in samples from traumatic brain injury patients 24 hours after the first dose of drugs or placebo and control subjects. Feature detection, retention time, alignment, annotation, and principal component analysis and statistical analysis were conducted using XCMS-online. The software "mummichog" was used for pathway and network analyses. A two-component principal component analysis revealed clustering of each of the groups, with distinct metabolomics signatures. Several novel pathways with plausible mechanistic involvement in traumatic brain injury were identified. A combination of metabolomics and pathway/network analyses showed that seven glutathione-centered pathways and two networks were enriched in the cerebrospinal fluid of traumatic brain injury patients treated with probenecid and N-acetylcysteine versus placebo-treated patients. Several additional pathways/networks consisting of components that are known substrates of probenecid-inhibitable transporters were also identified, providing additional mechanistic validation. CONCLUSIONS This proof-of-concept neuropharmacometabolomics assessment reveals alterations in known and previously unidentified metabolic pathways and supports therapeutic target engagement of the combination of probenecid and N-acetylcysteine treatment after severe traumatic brain injury in children.
Collapse
Affiliation(s)
- Fanuel T. Hagos
- Center for Clinical Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA
| | - Philip E. Empey
- Center for Clinical Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA
- Department of Pharmacy and Therapeutics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA
- Clinical and Translational Science Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Pengcheng Wang
- Center for Pharmacogenetics, University of Pittsburgh, Pittsburgh, PA
| | - Xiaochao Ma
- Center for Pharmacogenetics, University of Pittsburgh, Pittsburgh, PA
| | - Samuel M. Poloyac
- Center for Clinical Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA
- Department of Pharmacy and Therapeutics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA
- Clinical and Translational Science Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Hülya Bayır
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA
- Brain Care Institute, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, PA
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA
| | - Patrick M. Kochanek
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA
- Brain Care Institute, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, PA
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Michael J. Bell
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA
- Brain Care Institute, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, PA
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Pediatrics, Children’s National Health System, Washington, DC
| | - Robert S. B. Clark
- Clinical and Translational Science Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA
- Brain Care Institute, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, PA
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
18
|
Samavati R, Zádor F, Szűcs E, Tuka B, Martos D, Veres G, Gáspár R, Mándity IM, Fülöp F, Vécsei L, Benyhe S, Borsodi A. Kynurenic acid and its analogue can alter the opioid receptor G-protein signaling after acute treatment via NMDA receptor in rat cortex and striatum. J Neurol Sci 2017; 376:63-70. [PMID: 28431630 DOI: 10.1016/j.jns.2017.02.053] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 02/21/2017] [Accepted: 02/23/2017] [Indexed: 01/15/2023]
Abstract
Previously, we have shown that the N-methyl d-aspartate (NMDA)-receptor antagonist kynurenic acid (KYNA) and its analogue KYNA1 do not bind directly to mu, kappa and delta opioid receptors in vitro. On the other hand, chronic administration of KYNA and KYNA1 resulted in region (cortex vs striatum) and opioid receptor-type specific alterations in G-protein activation of mouse brain homogenates. Here we describe for the first time the acute effect of KYNA and KYNA1 on opioid receptor function with the possible involvement of the NMDA receptor. The acute 30minute in vivo KYNA1 and KYNA treatments altered opioid receptor G-protein signaling or ligand potency depending on the opioid receptor type and brain region (rat cortex vs striatum) using [35S]GTPγS binding assays. Pretreatment with the NMDA receptor antagonist MK-801 impaired or reversed the effects of KYNA1 and KYNA. These results suggest an NMDA receptor mediated effect. After acute 30minute treatment HPLC measurements revealed a similar KYNA1 and a higher KYNA plasma concentration compared to cerebrospinal fluid concentrations. Finally, KYNA, KYNA1 and MK-801 showed comparable results in opioid receptor G-protein activity and ligand potency with acute in vivo treatments when they were administered in vitro for 30min on isolated cortex and striatum slices. We previously demonstrated that KYNA1 and KYNA acutely altered opioid receptor function in vivo and in vitro through the NMDA receptor depending on the opioid receptor type and brain region. This study may lead to a new, indirect approach to influence opioid receptor signaling.
Collapse
Affiliation(s)
- Reza Samavati
- Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary; Institute of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, Eötvös u. 6, 6720 Szeged, Hungary
| | - Ferenc Zádor
- Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary.
| | - Edina Szűcs
- Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary
| | - Bernadett Tuka
- Department of Neurology, Faculty of Medicine, University of Szeged, Semmelweis utca 6, H-6725 Szeged, Hungary; MTA-SZTE Neuroscience Research Group, University of Szeged, Semmelweis utca 6, H-6725 Szeged, Hungary
| | - Diána Martos
- Department of Neurology, Faculty of Medicine, University of Szeged, Semmelweis utca 6, H-6725 Szeged, Hungary
| | - Gábor Veres
- Department of Neurology, Faculty of Medicine, University of Szeged, Semmelweis utca 6, H-6725 Szeged, Hungary; MTA-SZTE Neuroscience Research Group, University of Szeged, Semmelweis utca 6, H-6725 Szeged, Hungary
| | - Róbert Gáspár
- Institute of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, Eötvös u. 6, 6720 Szeged, Hungary
| | - István M Mándity
- Institute of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Szeged, Eötvös u. 6, 6720 Szeged, Hungary
| | - Ferenc Fülöp
- Institute of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Szeged, Eötvös u. 6, 6720 Szeged, Hungary
| | - László Vécsei
- Department of Neurology, Faculty of Medicine, University of Szeged, Semmelweis utca 6, H-6725 Szeged, Hungary; MTA-SZTE Neuroscience Research Group, University of Szeged, Semmelweis utca 6, H-6725 Szeged, Hungary
| | - Sándor Benyhe
- Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary
| | - Anna Borsodi
- Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary; Bio-Targeting Ltd., Vitez u. 1, 6722 Szeged, Hungary
| |
Collapse
|
19
|
Majláth Z, Török N, Toldi J, Vécsei L. Memantine and Kynurenic Acid: Current Neuropharmacological Aspects. Curr Neuropharmacol 2016; 14:200-9. [PMID: 26564141 PMCID: PMC4825950 DOI: 10.2174/1570159x14666151113123221] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 04/22/2015] [Accepted: 12/03/2015] [Indexed: 12/31/2022] Open
Abstract
Glutamatergic neurotransmission, of special importance in the human brain, is implicated in key brain functions such as synaptic plasticity and memory. The excessive activation of N-methyl- D-aspartate (NMDA) receptors may result in excitotoxic neuronal damage; this process has been implicated in the pathomechanism of different neurodegenerative disorders, such as Alzheimer’s disease (AD). Memantine is an uncompetitive antagonist of NMDA receptors with a favorable pharmacokinetic profile, and is therefore clinically well tolerated. Memantine is approved for the treatment of AD, but may additionally be beneficial for other dementia forms and pain conditions. Kynurenic acid
(KYNA) is an endogenous antagonist of NMDA receptors which has been demonstrated under experimental conditions to be neuroprotective. The development of a well-tolerated NMDA antagonist may offer a novel therapeutic option for the treatment of neurodegenerative disease and pain syndromes. KYNA may be a valuable candidate for future drug development.
Collapse
Affiliation(s)
| | | | | | - László Vécsei
- Department of Neurology, Faculty of Medicine, University of Szeged, Albert Szent-Györgyi Clinical Center, Semmelweis u. 6. H-6725 Szeged, Hungary.
| |
Collapse
|
20
|
Forrest CM, McNair K, Pisar M, Khalil OS, Darlington LG, Stone TW. Altered hippocampal plasticity by prenatal kynurenine administration, kynurenine-3-monoxygenase (KMO) deletion or galantamine. Neuroscience 2015; 310:91-105. [PMID: 26365611 PMCID: PMC4642643 DOI: 10.1016/j.neuroscience.2015.09.022] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 08/11/2015] [Accepted: 09/07/2015] [Indexed: 11/16/2022]
Abstract
Glutamate receptors sensitive to N-methyl-D-aspartate (NMDA) are involved in embryonic brain development but their activity may be modulated by the kynurenine pathway of tryptophan metabolism which includes an agonist (quinolinic acid) and an antagonist (kynurenic acid) at these receptors. Our previous work has shown that prenatal inhibition of the pathway produces abnormalities of brain development. In the present study kynurenine and probenecid (both 100mg/kg, doses known to increase kynurenic acid levels in the brain) were administered to female Wistar rats on embryonic days E14, E16 and E18 of gestation and the litter was allowed to develop to post-natal day P60. Western blotting revealed no changes in hippocampal expression of several proteins previously found to be altered by inhibition of the kynurenine pathway including the NMDA receptor subunits GluN1, GluN2A and GluN2B, as well as doublecortin, Proliferating Cell Nuclear Antigen (PCNA), sonic hedgehog and unco-ordinated (unc)-5H1 and 5H3. Mice lacking the enzyme kynurenine-3-monoxygenase (KMO) also showed no changes in hippocampal expression of several of these proteins or the 70-kDa and 100-kDa variants of Disrupted in Schizophrenia-1 (DISC1). Electrical excitability of pyramidal neurons in the CA1 region of hippocampal slices was unchanged, as was paired-pulse facilitation and inhibition. Long-term potentiation was decreased in the kynurenine-treated rats and in the KMO(-/-) mice, but galantamine reversed this effect in the presence of nicotinic receptor antagonists, consistent with evidence that it can potentiate glutamate at NMDA receptors. It is concluded that interference with the kynurenine pathway in utero can have lasting effects on brain function of the offspring, implying that the kynurenine pathway is involved in the regulation of early brain development.
Collapse
Affiliation(s)
- C M Forrest
- Institute of Neuroscience and Psychology, West Medical Building, University of Glasgow, Glasgow G12 8QQ, UK
| | - K McNair
- Institute of Neuroscience and Psychology, West Medical Building, University of Glasgow, Glasgow G12 8QQ, UK
| | - M Pisar
- Institute of Neuroscience and Psychology, West Medical Building, University of Glasgow, Glasgow G12 8QQ, UK
| | - O S Khalil
- Institute of Neuroscience and Psychology, West Medical Building, University of Glasgow, Glasgow G12 8QQ, UK
| | - L G Darlington
- Ashtead Hospital, The Warren, Ashtead, Surrey KT21 2SB, UK
| | - T W Stone
- Institute of Neuroscience and Psychology, West Medical Building, University of Glasgow, Glasgow G12 8QQ, UK.
| |
Collapse
|
21
|
Pineda-Farias JB, Barragán-Iglesias P, Loeza-Alcocer E, Torres-López JE, Rocha-González HI, Pérez-Severiano F, Delgado-Lezama R, Granados-Soto V. Role of anoctamin-1 and bestrophin-1 in spinal nerve ligation-induced neuropathic pain in rats. Mol Pain 2015; 11:41. [PMID: 26130088 PMCID: PMC4487556 DOI: 10.1186/s12990-015-0042-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 06/11/2015] [Indexed: 01/17/2023] Open
Abstract
Background Calcium-activated chloride channels (CaCCs) activation induces membrane depolarization by increasing chloride efflux in primary sensory neurons that can facilitate action potential generation. Previous studies suggest that CaCCs family members bestrophin-1 and anoctamin-1 are involved in inflammatory pain. However, their role in neuropathic pain is unclear. In this investigation we assessed the involvement of these CaCCs family members in rats subjected to the L5/L6 spinal nerve ligation. In addition, anoctamin-1 and bestrophin-1 mRNA and protein expression in dorsal root ganglion (DRG) and spinal cord was also determined in the presence and absence of selective inhibitors. Results L5/L6 spinal nerve ligation induced mechanical tactile allodynia. Intrathecal administration of non-selective CaCCs inhibitors (NPPB, 9-AC and NFA) dose-dependently reduced tactile allodynia. Intrathecal administration of selective CaCCs inhibitors (T16Ainh-A01 and CaCCinh-A01) also dose-dependently diminished tactile allodynia and thermal hyperalgesia. Anoctamin-1 and bestrophin-1 mRNA and protein were expressed in the dorsal spinal cord and DRG of naïve, sham and neuropathic rats. L5/L6 spinal nerve ligation rose mRNA and protein expression of anoctamin-1, but not bestrophin-1, in the dorsal spinal cord and DRG from day 1 to day 14 after nerve ligation. In addition, repeated administration of CaCCs inhibitors (T16Ainh-A01, CaCCinh-A01 or NFA) or anti-anoctamin-1 antibody prevented spinal nerve ligation-induced rises in anoctamin-1 mRNA and protein expression. Following spinal nerve ligation, the compound action potential generation of putative C fibers increased while selective CaCCs inhibitors (T16Ainh-A01 and CaCCinh-A01) attenuated such increase. Conclusions There is functional anoctamin-1 and bestrophin-1 expression in rats at sites related to nociceptive processing. Blockade of these CaCCs suppresses compound action potential generation in putative C fibers and lessens established tactile allodynia. As CaCCs activity contributes to neuropathic pain maintenance, selective inhibition of their activity may function as a tool to generate analgesia in nerve injury pain states. Electronic supplementary material The online version of this article (doi:10.1186/s12990-015-0042-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jorge Baruch Pineda-Farias
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados (Cinvestav), Sede Sur, Calzada de los Tenorios 235, Colonia Granjas Coapa, 14330, México, D.F., México.
| | - Paulino Barragán-Iglesias
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados (Cinvestav), Sede Sur, Calzada de los Tenorios 235, Colonia Granjas Coapa, 14330, México, D.F., México.
| | - Emanuel Loeza-Alcocer
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados (Cinvestav), Zacatenco, México, D.F., México.
| | - Jorge E Torres-López
- Laboratorio de Mecanismos de Dolor, División Académica de Ciencias de la Salud, Universidad Juárez Autónoma de Tabasco, Villahermosa, Tabasco, México. .,Hospital Regional de Alta Especialidad "Dr. Juan Graham Casasús", Villahermosa, Tabasco, México.
| | - Héctor Isaac Rocha-González
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, México, D.F., México.
| | - Francisca Pérez-Severiano
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", México, D.F., México.
| | - Rodolfo Delgado-Lezama
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados (Cinvestav), Zacatenco, México, D.F., México.
| | - Vinicio Granados-Soto
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados (Cinvestav), Sede Sur, Calzada de los Tenorios 235, Colonia Granjas Coapa, 14330, México, D.F., México.
| |
Collapse
|
22
|
García-Lara L, Pérez-Severiano F, González-Esquivel D, Elizondo G, Segovia J. Absence of aryl hydrocarbon receptors increases endogenous kynurenic acid levels and protects mouse brain against excitotoxic insult and oxidative stress. J Neurosci Res 2015; 93:1423-33. [DOI: 10.1002/jnr.23595] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 03/27/2015] [Accepted: 04/06/2015] [Indexed: 01/08/2023]
Affiliation(s)
- Lucia García-Lara
- Departamento de Fisiología; Biofísica; y Neurociencias; Centro de Investigación y de Estudios Avanzados del IPN; México D.F. México
| | - Francisca Pérez-Severiano
- Departamento de Neuroquímica; Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez; México D.F. México
| | - Dinora González-Esquivel
- Departamento de Neuroquímica; Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez; México D.F. México
| | - Guillermo Elizondo
- Departamento de Biología Celular; Centro de Investigación y de Estudios Avanzados del IPN; México D.F. México
| | - José Segovia
- Departamento de Fisiología; Biofísica; y Neurociencias; Centro de Investigación y de Estudios Avanzados del IPN; México D.F. México
| |
Collapse
|
23
|
Zhou W, Dantzer R, Budac DP, Walker AK, Mao-Ying QL, Lee AW, Heijnen CJ, Kavelaars A. Peripheral indoleamine 2,3-dioxygenase 1 is required for comorbid depression-like behavior but does not contribute to neuropathic pain in mice. Brain Behav Immun 2015; 46:147-53. [PMID: 25637485 PMCID: PMC4414738 DOI: 10.1016/j.bbi.2015.01.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 01/19/2015] [Accepted: 01/19/2015] [Indexed: 01/04/2023] Open
Abstract
Chronic pain frequently co-occurs with major depressive disorder but the mechanisms are poorly understood. We investigated the contribution of indoleamine 2,3-dioxygenase-1 (IDO1), a rate-limiting enzyme in the conversion of tryptophan to neurotoxic metabolites, to this comorbidity using the spared nerve injury (SNI) model of neuropathic pain in mice. SNI resulted in unilateral mechanical allodynia, reduced social interaction, and increased immobility in the forced swim test without changes in locomotor activity. These findings indicate SNI-induced pain and comorbid depression-like behavior. These behavioral responses were accompanied by increases in plasma kynurenine/tryptophan ratios and increased expression of Ido1 and Il1b mRNA in the liver. Interestingly, SNI did not induce detectable changes in spinal cord or brain Ido1 mRNA levels. SNI was associated with spinal cord inflammatory activity as evidenced by increased Il1b mRNA expression. The SNI-induced increase of liver Ido1and Il1b mRNA was abrogated by intrathecal administration of the IL-1 inhibitor IL-1RA. Intrathecal IL-1RA also inhibited both mechanical allodynia and depression-like behavior. We also show that Ido1 is required for the development of depression-like behavior because Ido1(-/-) mice do not develop increased immobility in the forced swim test or decreased social exploration in response to SNI. Mechanical allodynia was similar in WT and Ido1(-/-) mice. In conclusion, our findings show for the first time that neuropathic pain is associated with an increase of Ido1 in liver, but not brain, downstream of spinal cord IL-1β signaling and that Ido1 mediates comorbid depression. Moreover, comorbidity of neuropathic pain and depression are only partially mediated by a common mechanism because mechanical hyperalgesia develops independently of Ido1.
Collapse
Affiliation(s)
- Wenjun Zhou
- Laboratory of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Robert Dantzer
- Laboratory of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - David P Budac
- Bioanalysis and Physiology, Lundbeck Research, Paramus, NJ 07652, USA
| | - Adam K Walker
- Laboratory of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Qi-Liang Mao-Ying
- Laboratory of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institute of Acupuncture Research, Fudan University, Shanghai 200032, China
| | - Anna W Lee
- Neuroinflammation Disease Biology Unit, Lundbeck Research USA, Paramus, NJ 07652, USA
| | - Cobi J Heijnen
- Laboratory of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Annemieke Kavelaars
- Laboratory of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
24
|
Rocha-González HI, Ramírez-Aguilar M, Granados-Soto V, Reyes-García JG, Torres-López JE, Huerta-Cruz JC, Navarrete A. Antineuropathic effect of 7-hydroxy-3,4-dihydrocadalin in streptozotocin-induced diabetic rodents. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 14:129. [PMID: 24708659 PMCID: PMC3984493 DOI: 10.1186/1472-6882-14-129] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 03/31/2014] [Indexed: 01/03/2023]
Abstract
Background Painful neuropathy is the most common and debilitating complication of diabetes and results in hyperalgesia and allodynia. Hyperglycemia clearly plays a key role in the development and progression of diabetic neuropathy. Current therapeutic approaches are only partially successful and they are only thought to reduce the pain associated with peripheral neuropathy. Some natural products offer combined antioxidant, anti-inflammatory and antinociceptive properties that may help to treat in a more integrative manner this condition. In this regard, the purpose of this study was to investigate the antineuropathic effect of 7-hydroxy-3,4-dihydrocadalin in streptozotocin-induced diabetic rats and mice without glucose control as well as the possible mechanism of action involved in this effect. Methods Rats and mice were injected with 50 or 200 mg/kg streptozotocin, respectively, to produce hyperglycemia. The formalin test and von Frey filaments were used to assess the nociceptive activity. Rota-rod was utilized to measure motor activity and malondialdehyde assay to determine anti-oxidative properties. Results After 3 weeks of diabetes induction, chemical hyperalgesia was observed in streptozotocin-injected rats. Oral acute administration of 7-hydroxy-3,4-dihydrocadalin (0.3–30 mg/kg) decreased in a dose-dependent manner formalin-evoked hyperalgesia in diabetic rats. In addition, methiothepin (non-selective 5-HT receptor antagonist, 1 mg/kg, i.p.) and ODQ (guanylyl cyclase inhibitor, 2 mg/kg, i.p.), but not naltrexone (opioid receptor antagonist, 1 mg/kg, s.c.), prevented 7-hydroxy-3,4-dihydrocadalin-induced antihyperalgesic effect. The anti-hyperalgesic effect of 7-hydroxy-3,4-dihydrocadalin was similar to that produced by pregabalin (10 mg/kg, p.o.). Furthermore, oral acute administration of 7-hydroxy-3,4-dihydrocadalin (30 mg/kg) reduced streptozotocin-induced changes in malondialdehyde concentration from plasma samples. Unlike pregabalin, 7-hydroxy-3,4-dihydrocadalin did not affect motor activity. Six weeks after diabetes induction, tactile allodynia was observed in the streptozotocin-injected rats. At this time, oral administration of 7-hydroxy-3,4-dihydrocadalin (30 mg/kg) or pregabalin (10 mg/kg) reduced in a similar way tactile allodynia in diabetic rats. Finally, chronic oral administration of 7-hydroxy-3,4-dihydrocadalin (30-300 mg/kg, 3 times/week, during 6 weeks), significantly prevented the development of mechanical hyperalgesia and allodynia in streptozotocin-induced diabetic mice. Conclusions Data suggests that 7-hydroxy-3,4-dihydrocadalin has acute and chronic effects in painful diabetic neuropathy. This effect seems to involve antioxidant properties as well as activation of 5-HT receptors and inhibition of guanylyl cyclase enzyme.
Collapse
|
25
|
Khalil OS, Pisar M, Forrest CM, Vincenten MCJ, Darlington LG, Stone TW. Prenatal inhibition of the kynurenine pathway leads to structural changes in the hippocampus of adult rat offspring. Eur J Neurosci 2014; 39:1558-71. [PMID: 24646396 PMCID: PMC4368408 DOI: 10.1111/ejn.12535] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 01/28/2014] [Accepted: 01/30/2014] [Indexed: 12/13/2022]
Abstract
Glutamate receptors for N-methyl-d-aspartate (NMDA) are involved in early brain development. The kynurenine pathway of tryptophan metabolism includes the NMDA receptor agonist quinolinic acid and the antagonist kynurenic acid. We now report that prenatal inhibition of the pathway in rats with 3,4-dimethoxy-N-[4-(3-nitrophenyl)thiazol-2-yl]benzenesulphonamide (Ro61-8048) produces marked changes in hippocampal neuron morphology, spine density and the immunocytochemical localisation of developmental proteins in the offspring at postnatal day 60. Golgi–Cox silver staining revealed decreased overall numbers and lengths of CA1 basal dendrites and secondary basal dendrites, together with fewer basal dendritic spines and less overall dendritic complexity in the basal arbour. Fewer dendrites and less complexity were also noted in the dentate gyrus granule cells. More neurons containing the nuclear marker NeuN and the developmental protein sonic hedgehog were detected in the CA1 region and dentate gyrus. Staining for doublecortin revealed fewer newly generated granule cells bearing extended dendritic processes. The number of neuron terminals staining for vesicular glutamate transporter (VGLUT)-1 and VGLUT-2 was increased by Ro61-8048, with no change in expression of vesicular GABA transporter or its co-localisation with vesicle-associated membrane protein-1. These data support the view that constitutive kynurenine metabolism normally plays a role in early embryonic brain development, and that interfering with it has profound consequences for neuronal structure and morphology, lasting into adulthood.
Collapse
Affiliation(s)
- Omari S Khalil
- Institute of Neuroscience and Psychology, West Medical Building, University of Glasgow, Glasgow, G12 8QQ, UK
| | | | | | | | | | | |
Collapse
|