1
|
Alkanli N, Ay A, Cevik G. Investigation of the relationships between eNOS T786C, G894T, intron 4 VNTR (4a/b) gene variations and prostate cancer development and progression. Nitric Oxide 2024; 152:69-77. [PMID: 39322022 DOI: 10.1016/j.niox.2024.09.008] [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: 04/07/2024] [Revised: 08/09/2024] [Accepted: 09/20/2024] [Indexed: 09/27/2024]
Abstract
BACKGROUND This study aimed to investigate the relationships between eNOS T786C, G894T, intron 4 VNTR (4a/b) gene variations and prostate cancer development and progression. MATERIALS AND METHODS This study included 88 patients diagnosed with prostate cancer and 91 healthy controls. Polymerase chain reaction (PCR) and restriction fragment length polymorphism (RFLP) methods were used to determine the genotype distributions of eNOS T786C, G894T, intron 4 VNTR (4a/b) gene variations. RESULTS In our study, the CC homozygous genotype of eNOS T786C gene variation was determined to be significantly higher in the prostate cancer patient group compared to the healthy control group (OR: 2.343, 95%Cl: 0.990-5.544, p = 0.026), while the CT heterozygous genotype was found to be significantly higher in the healthy control group compared to the prostate cancer patient group was found to be significantly higher (OR: 0.589, 95%Cl: 0.325-1.068, p = 0.041). In addition, while the TT homozygous genotype of the eNOS G894T gene variation was found to be significantly higher in the prostate cancer patient group compared to the healthy control group (OR: 9.068, 95%Cl: 4.396-18.777, p < 0.001), the GT heterozygous genotype was found to be significantly higher in the healthy control group compared to the prostate cancer patient group was determined significantly higher (OR: 0.227, 95%Cl: 0.121-0.427, p < 0.001). For eNOS (4VNTR (4a/b) - G894T) gene variations, aa-TT (p = 0.042) and bb-TT (p < 0.001) haplotype frequencies were significantly higher in the prostate cancer patient group, while aa-GT (p = 0.017), bb-GG (p = 0.049) and bb-GT (p < 0.001) haplotype frequencies were found to be significantly higher in the healthy control group. For eNOS (4VNTR (4a/b) - T786C) gene variations, the bb-CC haplotype frequency was determined to be significantly higher in the patient group (p = 0.049), while the bb-CT haplotype frequency was determined to be significantly higher in the control group (p = 0.008). For eNOS (T786C -G894T) gene variations, TT-TT (p < 0.001) and CC-TT (p = 0.025) haplotype frequencies were found to be significantly higher in the patient group. On the other hand, TT-GT (p = 0.002) and CT-GT (p < 0.001) haplotype frequencies were determined to be significantly higher in the control group. The aa genotype of the intron 4 VNTR (4a/b) gene variation was determined to be significantly higher at Gleason score ≥7 compared to Gleason score <7 (OR: 0.184, 95%Cl: 0.050-0.677, p = 0.005). PSA levels were determined significantly higher in patients with Gleason score 7 and above (p = 0.008). The risk of developing prostate cancer was found to be significantly higher in patients carrying the CC homozygous mutant genotype of the eNOS T786C gene variation (p = 0.024) and in patients carrying the TT homozygous genotype of the G894T gene variation (p = 0.021). CONCLUSIONS In our study, the CC homozygous genotype of the eNOS T786C gene variation was determined as a genetic risk factor for the development of prostate cancer, while the CT heterozygous genotype was determined as a protective factor against prostate cancer. For the eNOS G894T gene variation, the TT homozygous genotype was determined as a genetic risk factor for the development of prostate cancer, while the GT heterozygous genotype was determined as a protective factor against prostate cancer. Additionally, for eNOS (4VNTR (4a/b) - G894T) gene variations, aa-TT and bb-TT haplotypes have been identified as genetic risk factors for the development of prostate cancer, while aa-GT, bb-GG and bb-GT haplotypes have been identified as protective factors against the disease has been determined. For eNOS (4VNTR (4a/b) - T786C) gene variations, the bb-CC haplotype was determined as a genetic risk factor in the development of prostate cancer, while the bb-CT haplotype was determined as a protective factor against the disease. TT-TT and CC-TT haplotypes for eNOS (T786C -G894T) gene variations have been identified as genetic risk factors for the development of prostate cancer. In contrast, TT-GT and CT-GT haplotypes were found to be protective factors against the disease. The aa genotype of the intron 4 VNTR (4a/b) gene variation has also been identified as an important genetic risk factor in prostate cancer progression. Significantly increased PSA levels in patients with Gleason score 7 and above, and significantly increased PSA levels in patients carrying the CC and TT homozygous mutant genotype for T786C and G894T gene variations were determined as important risk factors. It is thought that the genetic biomarkers in our study may play a role as personalized therapeutic agents in slowing down the development of prostate cancer, increasing the effectiveness of treatment in prostate cancer, affecting the responses to drugs that regulate NO signaling, predetermining genetic predisposition to prostate cancer, and risk assessment in patients with prostate cancer.
Collapse
Affiliation(s)
- Nevra Alkanli
- Department of Biophysics, Faculty of Medicine, Haliç University, Istanbul, Turkey, 34060.
| | - Arzu Ay
- Department of Biophysics, Faculty of Medicine, Trakya University, Edirne, Turkey, 22030.
| | - Gokhan Cevik
- Department of Urology, Faculty of Medicine, Trakya University, Edirne, Turkey, 22030.
| |
Collapse
|
2
|
Ko EJ, Kim EJ, Cho HJ, Oh J, Park HS, Ryu CS, Kim JO, Jun HH, Chong SY, Kim JW, Kim NK. Prognostic significance of three endothelial nitric oxide synthase (eNOS) polymorphisms and metabolic syndrome (MetS) in patients with colorectal cancer. Genes Genomics 2022; 44:659-670. [PMID: 35377131 DOI: 10.1007/s13258-022-01246-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 03/12/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Polymorphisms of endothelial nitric oxide synthases (eNOS) have been associated with cancer susceptibility. Also, metabolic syndrome is associated with cancer malignancy. However, the effect of eNOS polymorphisms and metabolic syndrome on colorectal cancer (CRC) prognosis remains unclear. OBJECTIVE To investigated whether three genetic polymorphisms (- 786 T > C rs2070744, 4a4b rs869109213, and 894G > T rs1799983) in the eNOS and metabolic syndrome (MetS) were associated with CRC patient survival. METHODS We genotyped three polymorphisms of eNOS (- 786 T > C, 4a4b, and 894G > T) in 312 CRC cases from the Korean population by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) analysis. RESULTS Although the three eNOS polymorphisms were not causative of MetS, the TT genotype of the 894G > T polymorphism was associated with a worse survival rate compared with the GG genotype in the CRC group with MetS than in the CRC group without MetS (5-years survival; adjusted HR = 54.777; 95% CI 5.073-591.487 and RFS; adjusted HR = 14.909; 95% CI 1.571-141.528). CONCLUSIONS The eNOS polymorphisms were not associated with metabolic syndrome prevalence in CRC patients. However, our findings suggest that the eNOS 894G > T polymorphism with MetS was associated with poor clinical outcomes.
Collapse
Affiliation(s)
- Eun Ju Ko
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, South Korea
| | - Eo Jin Kim
- Division of Hematology/Oncology, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hye Jung Cho
- Department of Surgery, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Jisu Oh
- Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, South Korea.,Division of Hematology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Han Sung Park
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, South Korea
| | - Chang Soo Ryu
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, South Korea
| | - Jung Oh Kim
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, South Korea
| | - Hak Hoon Jun
- Department of Surgery, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - So Young Chong
- Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Jong Woo Kim
- Department of Surgery, CHA Bundang Medical Center, CHA University, Seongnam, South Korea.
| | - Nam Keun Kim
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, South Korea.
| |
Collapse
|
3
|
Ghasemi A, Jeddi S, Kashfi K. The laboratory rat: Age and body weight matter. EXCLI JOURNAL 2021; 20:1431-1445. [PMID: 34737685 PMCID: PMC8564917 DOI: 10.17179/excli2021-4072] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 08/31/2021] [Indexed: 12/20/2022]
Abstract
Animal experimentation helps us to understand human biology. Rodents and, in particular, rats are among the most common animals used in animal experiments. Reporting data on animal age, animal body weight, and animal postnatal developmental stages is not consistent, which can cause the failure to translate animal data to humans. This review summarizes age-related postnatal developmental stages in rats by addressing age-related changes in their body weights. The age and body weight of animals can affect drug metabolism, gene expression, metabolic parameters, and other dependent variables measured in animal studies. In addition, considering the age and the body weight of the animals is of particular importance in animal modeling of human diseases. Appropriate reporting of age, body weight, and the developmental stage of animals used in studies can improve animal to human translation.
Collapse
Affiliation(s)
- Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajad Jeddi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, USA
| |
Collapse
|
4
|
da Silva GM, da Silva MC, Nascimento DVG, Lima Silva EM, Gouvêa FFF, de França Lopes LG, Araújo AV, Ferraz Pereira KN, de Queiroz TM. Nitric Oxide as a Central Molecule in Hypertension: Focus on the Vasorelaxant Activity of New Nitric Oxide Donors. BIOLOGY 2021; 10:1041. [PMID: 34681140 PMCID: PMC8533285 DOI: 10.3390/biology10101041] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/08/2021] [Accepted: 10/09/2021] [Indexed: 12/15/2022]
Abstract
Cardiovascular diseases include all types of disorders related to the heart or blood vessels. High blood pressure is an important risk factor for cardiac complications and pathological disorders. An increase in circulating angiotensin-II is a potent stimulus for the expression of reactive oxygen species and pro-inflammatory cytokines that activate oxidative stress, perpetuating a deleterious effect in hypertension. Studies demonstrate the capacity of NO to prevent platelet or leukocyte activation and adhesion and inhibition of proliferation, as well as to modulate inflammatory or anti-inflammatory reactions and migration of vascular smooth muscle cells. However, in conditions of low availability of NO, such as during hypertension, these processes are impaired. Currently, there is great interest in the development of compounds capable of releasing NO in a modulated and stable way. Accordingly, compounds containing metal ions coupled to NO are being investigated and are widely recognized as having great relevance in the treatment of different diseases. Therefore, the exogenous administration of NO is an attractive and pharmacological alternative in the study and treatment of hypertension. The present review summarizes the role of nitric oxide in hypertension, focusing on the role of new NO donors, particularly the metal-based drugs and their protagonist activity in vascular function.
Collapse
Affiliation(s)
- Gabriela Maria da Silva
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão 55.608-680, PE, Brazil; (G.M.d.S.); (M.C.d.S.); (D.V.G.N.); (E.M.L.S.); (A.V.A.); (K.N.F.P.)
| | - Mirelly Cunha da Silva
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão 55.608-680, PE, Brazil; (G.M.d.S.); (M.C.d.S.); (D.V.G.N.); (E.M.L.S.); (A.V.A.); (K.N.F.P.)
| | - Déborah Victória Gomes Nascimento
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão 55.608-680, PE, Brazil; (G.M.d.S.); (M.C.d.S.); (D.V.G.N.); (E.M.L.S.); (A.V.A.); (K.N.F.P.)
| | - Ellen Mayara Lima Silva
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão 55.608-680, PE, Brazil; (G.M.d.S.); (M.C.d.S.); (D.V.G.N.); (E.M.L.S.); (A.V.A.); (K.N.F.P.)
| | - Fabíola Furtado Fialho Gouvêa
- School of Technical Health, Health Sciences Center, Federal University of Paraíba, João Pessoa 58.051-900, PB, Brazil;
| | - Luiz Gonzaga de França Lopes
- Laboratory of Bioinorganic Chemistry, Department of Organic and Inorganic Chemistry, Federal University of Ceará, Fortaleza 60.020-181, CE, Brazil;
| | - Alice Valença Araújo
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão 55.608-680, PE, Brazil; (G.M.d.S.); (M.C.d.S.); (D.V.G.N.); (E.M.L.S.); (A.V.A.); (K.N.F.P.)
| | - Kelli Nogueira Ferraz Pereira
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão 55.608-680, PE, Brazil; (G.M.d.S.); (M.C.d.S.); (D.V.G.N.); (E.M.L.S.); (A.V.A.); (K.N.F.P.)
| | - Thyago Moreira de Queiroz
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão 55.608-680, PE, Brazil; (G.M.d.S.); (M.C.d.S.); (D.V.G.N.); (E.M.L.S.); (A.V.A.); (K.N.F.P.)
| |
Collapse
|
5
|
Barberio L, Paulesu L, Canesi L, Grasselli E, Mandalà M. Bisphenol a Interferes with Uterine Artery Features and Impairs Rat Feto-Placental Growth. Int J Mol Sci 2021; 22:ijms22136912. [PMID: 34199136 PMCID: PMC8268965 DOI: 10.3390/ijms22136912] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/15/2021] [Accepted: 06/22/2021] [Indexed: 12/11/2022] Open
Abstract
Bisphenol A (BPA) is a widespread environmental contaminant, found in human fluids and tissues. Maternal BPA exposure is associated with alterations in pregnancy outcomes. Because maternal uterine circulation plays a crucial role in normal placenta and fetal growth, we hypothesized that BPA compromises the function of uterine arteries (UAs) and fetoplacental development. Female rats were orally administered with BPA (2.5, 25 and 250 µg/kg/day) or with its vehicle (ethanol) for 30 days before pregnancy and during the first 20 days of pregnancy. To compare the effect of BPA in the reproductive vs. systemic circulation, it was tested on UAs and mesenteric arteries (MAs). Arteries were isolated and examined by pressure myography. Moreover, fetuses and placentas were weighed to provide an index of reproductive performance. In UAs of BPA-treated rats, lumen diameter, acetylcholine-relaxation and expressions of endothelial nitric oxide synthase 3 (NOS3), estrogen receptor α (ERα) and peroxisome proliferator-activated receptor ɣ (PPARɣ) were reduced. Conversely, no changes were observed in MAs. BPA treatment also reduced placental weights, while fetal weights were increased. For the first time, our results indicate that UAs represent a specific target of BPA during pregnancy and provide insight into the molecular mechanisms that underlie its negative effects on pregnancy outcomes.
Collapse
Affiliation(s)
- Laura Barberio
- Department of Biology, Ecology & Earth Sciences, University of Calabria, 87036 Rende, Italy;
| | - Luana Paulesu
- Department of Life Sciences, University of Siena, 53100 Siena, Italy;
| | - Laura Canesi
- Department of Earth, Environment and Life Sciences (DISTAV), University of Genova, 16132 Genova, Italy; (L.C.); (E.G.)
| | - Elena Grasselli
- Department of Earth, Environment and Life Sciences (DISTAV), University of Genova, 16132 Genova, Italy; (L.C.); (E.G.)
| | - Maurizio Mandalà
- Department of Biology, Ecology & Earth Sciences, University of Calabria, 87036 Rende, Italy;
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Vermont, Burlington, VT 05405, USA
- Correspondence:
| |
Collapse
|
6
|
Tang X, Yan K, Wang Y, Wang Y, Chen H, Xu J, Lu Y, Wang X, Liang J, Zhang X. Activation of PPAR-β/δ Attenuates Brain Injury by Suppressing Inflammation and Apoptosis in a Collagenase-Induced Intracerebral Hemorrhage Mouse Model. Neurochem Res 2020; 45:837-850. [PMID: 31939088 PMCID: PMC7078151 DOI: 10.1007/s11064-020-02956-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 12/02/2019] [Accepted: 01/07/2020] [Indexed: 01/01/2023]
Abstract
Brain injury has been proposed as the major cause of the poor outcomes associated with intracerebral hemorrhage (ICH). Emerging evidence indicates that the nuclear receptor, peroxisome proliferator-activated receptor β/δ (PPAR-β/δ), plays a crucial role in the pathological process of central nervous impairment. The present study was undertaken to evaluate the protective effects of PPAR-β/δ activation using a selective PPAR-β/δ agonist, GW0742, against brain injury after ICH in a mouse model. ICH was induced by intravenous injection of collagenase into the right caudate putamen. To examine the protective effect of PPAR-β/δ activation against ICH-induced brain injury, mice were either intraperitoneally injected with GW0742 (3 mg/kg, body weight) or saline (control group) 30 min before inducing ICH. Behavioral dysfunction was evaluated 24 and 72 h after injury. Then, all mice were killed to assess hematoma volume, brain water content, and blood-brain barrier (BBB) permeability. TUNEL and Nissl staining were performed to quantify the brain injury. The expression of PPAR-β/δ, interleukin (IL)-1β, tumor necrosis factor (TNF)-α, Bcl-2-related X-protein (Bax), and B-cell lymphoma 2 (Bcl-2) in the perihematomal area was examined by immunohistochemistry and western blotting analysis. Mice treated with GW0742 showed significantly less severe behavioral deficits compared to the control group, accompanied by increased expression of PPAR-β/δ and Bcl-2, and increased expression of IL-1β, TNF-α, and Bax decreased simultaneously in the GW0742-treated group. Furthermore, the GW0742-pretreated group showed significantly less brain edema and BBB leakage. Neuronal loss was attenuated, and the number of apoptotic neuronal cells in perihematomal tissues reduced, in the GW0742-pretreated group compared to the control group. However, the hematoma volume did not decrease significantly on day 3 after ICH. These results suggest that the activation of PPAR-β/δ exerts a neuroprotective effect on ICH-induced brain injury, possibly through anti-inflammatory and anti-apoptotic pathways.
Collapse
Affiliation(s)
- Xiangming Tang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
| | - Kunning Yan
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
| | - Yingge Wang
- Department of Neurology, Affiliated Hospital of Yangzhou University, Yangzhou, 225001, China
| | - Yaping Wang
- Department of Electrocardiogram, Affiliated WuTaiShan Hospital of Medical College of Yangzhou University, Yangzhou Mental Health Centre, Yangzhou, 225000, China
| | - Hongmei Chen
- School of Nursing, Yangzhou University, Yangzhou, 225009, China
| | - Jiang Xu
- General Hospital of Xuzhou Mining Group, Xuzhou, 221006, China
| | - Yaoyao Lu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
| | - Xiaohong Wang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
| | - Jingyan Liang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China. .,Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China. .,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, China.
| | - Xinjiang Zhang
- Department of Neurology, Affiliated Hospital of Yangzhou University, Yangzhou, 225001, China.
| |
Collapse
|
7
|
Qasem H, Al-Ayadhi L, Bjørklund G, Chirumbolo S, El-Ansary A. Impaired lipid metabolism markers to assess the risk of neuroinflammation in autism spectrum disorder. Metab Brain Dis 2018; 33:1141-1153. [PMID: 29569150 DOI: 10.1007/s11011-018-0206-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 02/19/2018] [Indexed: 01/19/2023]
Abstract
Autism spectrum disorder (ASD) is a multifactorial disorder caused by an interaction between environmental risk factors and a genetic background. It is characterized by impairment in communication, social interaction, repetitive behavior, and sensory processing. The etiology of ASD is still not fully understood, and the role of neuroinflammation in autism behaviors needs to be further investigated. The aim of the present study was to test the possible association between prostaglandin E2 (PGE2), cyclooxygenase-2 (COX-2), microsomal prostaglandin E synthase-1 (mPGES-1), prostaglandin PGE2 EP2 receptors and nuclear kappa B (NF-κB) and the severity of cognitive disorders, social impairment, and sensory dysfunction. PGE2, COX-2, mPGES-1, PGE2-EP2 receptors and NF-κB as biochemical parameters related to neuroinflammation were determined in the plasma of 47 Saudi male patients with ASD, categorized as mild to moderate and severe as indicated by the Childhood Autism Rating Scale (CARS) or the Social Responsiveness Scale (SRS) or the Short Sensory Profile (SSP) and compared to 46 neurotypical controls. The data indicated that ASD patients have remarkably higher levels of the measured parameters compared to neurotypical controls, except for EP2 receptors that showed an opposite trend. While the measured parameter did not correlate with the severity of social and cognitive dysfunction, PGE2, COX-2, and mPGES-1 were remarkably associated with the dysfunction in sensory processing. NF-κB was significantly increased in relation to age. Based on the discussed data, the positive correlation between PGE2, COX-2, and mPGES-1 confirm the role of PGE2 pathway and neuroinflammation in the etiology of ASD, and the possibility of using PGE2, COX-2 and mPGES-1 as biomarkers of autism severity. NF-κB as inflammatory inducer showed an elevated level in plasma of ASD individuals. Receiver operating characteristic analysis together with predictiveness diagrams proved that the measured parameters could be used as predictive biomarkers of biochemical correlates to ASD.
Collapse
Affiliation(s)
- Hanan Qasem
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Laila Al-Ayadhi
- Autism Research and Treatment Center, Riyadh, Saudi Arabia
- Shaik AL-Amodi Autism Research Chair, King Saud University, Riyadh, Saudi Arabia
- Physiology Department, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Mo i Rana, Norway
| | - Salvatore Chirumbolo
- Department of Neurological and Movement Sciences, University of Verona, Verona, Italy
| | - Afaf El-Ansary
- Autism Research and Treatment Center, Riyadh, Saudi Arabia.
- Shaik AL-Amodi Autism Research Chair, King Saud University, Riyadh, Saudi Arabia.
- Central laboratory, Female Centre for Scientific and Medical Studies, King Saud University, Riyadh, Saudi Arabia.
- Therapeutic Chemistry Department, National Research Center, Dokki, Guiza, Egypt.
| |
Collapse
|
8
|
Gheibi S, Jeddi S, Kashfi K, Ghasemi A. Regulation of vascular tone homeostasis by NO and H 2S: Implications in hypertension. Biochem Pharmacol 2018; 149:42-59. [PMID: 29330066 PMCID: PMC5866223 DOI: 10.1016/j.bcp.2018.01.017] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 01/05/2018] [Indexed: 02/09/2023]
Abstract
Nitric oxide (NO) and hydrogen sulfide (H2S) are two gasotransmitters that are produced in the vasculature and contribute to the regulation of vascular tone. NO and H2S are synthesized in both vascular smooth muscle and endothelial cells; NO functions primarily through the sGC/cGMP pathway, and H2S mainly through activation of the ATP-dependent potassium channels; both leading to relaxation of vascular smooth muscle cells. A deficit in the NO/H2S homeostasis is involved in the pathogenesis of various cardiovascular diseases, especially hypertension. It is now becoming increasingly clear that there are important interactions between NO and H2S and that have a profound impact on vascular tone and this may provide insights into the new therapeutic interventions. The aim of this review is to provide a better understanding of individual and interactive roles of NO and H2S in vascular biology. Overall, available data indicate that both NO and H2S contribute to vascular (patho)physiology and in regulating blood pressure. In addition, boosting NO and H2S using various dietary sources or donors could be a hopeful therapeutic strategy in the management of hypertension.
Collapse
Affiliation(s)
- Sevda Gheibi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Neurophysiology Research Center and Department of Physiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajad Jeddi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, NY, USA
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Duran CL, Howell DW, Dave JM, Smith RL, Torrie ME, Essner JJ, Bayless KJ. Molecular Regulation of Sprouting Angiogenesis. Compr Physiol 2017; 8:153-235. [PMID: 29357127 DOI: 10.1002/cphy.c160048] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The term angiogenesis arose in the 18th century. Several studies over the next 100 years laid the groundwork for initial studies performed by the Folkman laboratory, which were at first met with some opposition. Once overcome, the angiogenesis field has flourished due to studies on tumor angiogenesis and various developmental models that can be genetically manipulated, including mice and zebrafish. In addition, new discoveries have been aided by the ability to isolate primary endothelial cells, which has allowed dissection of various steps within angiogenesis. This review will summarize the molecular events that control angiogenesis downstream of biochemical factors such as growth factors, cytokines, chemokines, hypoxia-inducible factors (HIFs), and lipids. These and other stimuli have been linked to regulation of junctional molecules and cell surface receptors. In addition, the contribution of cytoskeletal elements and regulatory proteins has revealed an intricate role for mobilization of actin, microtubules, and intermediate filaments in response to cues that activate the endothelium. Activating stimuli also affect various focal adhesion proteins, scaffold proteins, intracellular kinases, and second messengers. Finally, metalloproteinases, which facilitate matrix degradation and the formation of new blood vessels, are discussed, along with our knowledge of crosstalk between the various subclasses of these molecules throughout the text. Compr Physiol 8:153-235, 2018.
Collapse
Affiliation(s)
- Camille L Duran
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - David W Howell
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Jui M Dave
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Rebecca L Smith
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Melanie E Torrie
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Jeffrey J Essner
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Kayla J Bayless
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| |
Collapse
|
10
|
Turrin NP, Rivest S. Unraveling the Molecular Details Involved in the Intimate Link between the Immune and Neuroendocrine Systems. Exp Biol Med (Maywood) 2016; 229:996-1006. [PMID: 15522835 DOI: 10.1177/153537020422901003] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
During systemic infections, the immune system can signal the brain and act on different neuronal circuits via soluble molecules, such as proinflammatory cytokines, that act on the cells forming the blood-brain barrier and the circumventricular organs. These activated cells release prostaglandin of the E2 type (PGE2), which is the endogenous ligand that triggers the pathways involved in the control of autonomic functions necessary to restore homeostasis and provide inhibitory feedback to innate immunity. Among these neurophysiological functions, activation of the circuits that control the plasma release of glucocorticoids is probably the most critical to the survival of the host in the presence of pathogens. This review revisits this issue and describes in depth the molecular details (including the emerging role of Toll-like receptors during inflammation) underlying the influence of circulating inflammatory molecules on the cerebral tissue, focusing on their contribution in the synthesis and action PGE2 in the brain. We also provide an innovative view supporting the concept of “fast and delayed response” involving the same ligands but different groups of cells, signal transduction pathways, and target genes.
Collapse
Affiliation(s)
- Nicolas P Turrin
- Laboratory of Molecular Endocrinology, CHUL Research Center and Department of Anatomy and Physiology, Laval University, 2705 Boulevard Laurier, Québec G1V 4G2, Canada
| | | |
Collapse
|
11
|
Meroni PL, Tincani A, Sepp N, Raschi E, Testoni C, Corsini E, Cavazzana I, Pellegrini S, Salmaggi A. Endothelium and the brain in CNS lupus. Lupus 2016; 12:919-28. [PMID: 14714912 DOI: 10.1191/0961203303lu503oa] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Central nervous system (CNS) involvement in systemic lupus erythematosus (SLE) is common and results in different clinical manifestations. Several pathogenic mechanisms have been suggested to play a rolein determiningsuch a varietyof clinicalsymptoms.The thrombophilicstateassociatedto the presence of antiphospholipidantibodies has been suggested to be responsible for a noninflammatory vasculopathywhichcauses clear ischaemiceventsas well as alterationsof the cerebralmicrocirculation that are likely associated to seizures, cognitive dysfunction or psychosis. Although less frequent, a true vasculitic process affecting cerebral circulation has also been reported. In both cases, brain endothelium does represent the target of the pathogenic mechanisms. Brain endothelial cells display peculiar functional and phenotypical characteristics in comparison with endothelial cells from other anatomical districts, raising the possibility that this might be the reason for its susceptibility in lupus disease. We review and present data suggesting that a higher and firmer expression of beta 2 glycoprotein I on endothelialcell membranes can be responsiblefor a selective damage/activation by circulating anti-beta 2 glycoprotein I, and that antiendothelial cell antibodies crossreact with brain endothelium and in some cases, specifically bind brain endothelial cells only in lupus patients with central nervous involvement.
Collapse
Affiliation(s)
- P L Meroni
- Allergy and Clinical Immunology Unit, Department of Internal Medicine, University of Milan, IRCCS Istituto Auxologico Italiano, Milan, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Inflammatory transcription factors as activation markers and functional readouts in immune-to-brain communication. Brain Behav Immun 2016; 54:1-14. [PMID: 26348582 DOI: 10.1016/j.bbi.2015.09.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 08/31/2015] [Accepted: 09/04/2015] [Indexed: 02/06/2023] Open
Abstract
Immune-to-brain communication pathways involve humoral mediators, including cytokines, central modulation by neuronal afferents and immune cell trafficking to the brain. During systemic inflammation these pathways contribute to mediating brain-controlled sickness symptoms including fever. Experimentally, activation of these signaling pathways can be mimicked and studied when injecting animals with pathogen associated molecular patterns (PAMPS). One central component of the brain inflammatory response, which leads, for example, to fever induction, is transcriptional activation of brain cells via cytokines and PAMPS. We and others have studied the spatiotemporal activation and the physiological significance of transcription factors for the induction of inflammation within the brain and the manifestation of fever. Evidence has revealed a role of nuclear factor (NF)κB in the initiation, signal transducer and activator of transcription (STAT)3 in the maintenance and NF-interleukin (IL)6 in the maintenance or even termination of brain-inflammation and fever. Moreover, psychological stressors, such as exposure to a novel environment, leads to increased body core temperature and genomic NF-IL6-activation, suggesting a potential use of NF-IL6-immunohistochemistry as a multimodal brain cell activation marker and a role for NF-IL6 for differential brain activity. In addition, the nutritional status, as reflected by circulating levels of the cytokine-like hormone leptin, influence immune-to-brain communication and age-dependent changes in LPS-induced fever. Overall, transcription factors remain therapeutically important targets for the treatment of brain-inflammation and fever induction during infectious/non-infectious inflammatory and psychological stress. However, the exact physiological role and significance of these transcription factors requires to be further investigated.
Collapse
|
13
|
Recent advances in stress research: Focus on nitric oxide. Eur J Pharmacol 2015; 765:406-14. [DOI: 10.1016/j.ejphar.2015.08.055] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 08/27/2015] [Accepted: 08/28/2015] [Indexed: 12/29/2022]
|
14
|
Murphy EJ. Blood-brain barrier and brain fatty acid uptake: Role of arachidonic acid and PGE2. J Neurochem 2015; 135:845-8. [PMID: 26383055 DOI: 10.1111/jnc.13289] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 08/13/2015] [Indexed: 02/02/2023]
Abstract
How do fatty acids enter the brain and what role, if any, do membrane and cytosolic fatty acid binding proteins have on facilitating this process? This is a fundamental question that many lipid neurochemists will freely admit they cannot answer in any kind of definitive manner. A study by Dalvi and colleagues in this issue of the Journal of Neurochemistry now adds to our knowledge in this field. Among other important observations, their experiments demonstrate that a physiological level of arachidonic acid (ARA), that could be associated with many different physiological and pathophysiological states, increases permeability in a model of the human blood brain barrier (BBB) in the absence of cytokines. This last point is very important as it suggests increases in BBB permeability may occur in situations other than those associated with increases in tumor necrosis factor a (TNFα) and interleukin1b (IL1β), giving additional options for developing drugs impacting BBB permeability.
Collapse
Affiliation(s)
- Eric J Murphy
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, USA
| |
Collapse
|
15
|
Pathologic role of glial nitric oxide in adult and pediatric neuroinflammatory diseases. Neurosci Biobehav Rev 2014; 45:168-82. [DOI: 10.1016/j.neubiorev.2014.06.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2013] [Revised: 05/28/2014] [Accepted: 06/05/2014] [Indexed: 01/22/2023]
|
16
|
Lukewich MK, Rogers RC, Lomax AE. Divergent neuroendocrine responses to localized and systemic inflammation. Semin Immunol 2014; 26:402-8. [PMID: 24486057 DOI: 10.1016/j.smim.2014.01.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 01/09/2014] [Indexed: 12/19/2022]
Abstract
The sympathetic nervous system (SNS) is part of an integrative network that functions to restore homeostasis following injury and infection. The SNS can provide negative feedback control over inflammation through the secretion of catecholamines from postganglionic sympathetic neurons and adrenal chromaffin cells (ACCs). Central autonomic structures receive information regarding the inflammatory status of the body and reflexively modulate SNS activity. However, inflammation and infection can also directly regulate SNS function by peripheral actions on postganglionic cells. The present review discusses how inflammation activates autonomic reflex pathways and compares the effect of localized and systemic inflammation on ACCs and postganglionic sympathetic neurons. Systemic inflammation significantly enhanced catecholamine secretion through an increase in Ca(2+) release from the endoplasmic reticulum. In contrast, acute and chronic GI inflammation reduced voltage-gated Ca(2+) current. Thus it appears that the mechanisms underlying the effects of peripheral and systemic inflammation neuroendocrine function converge on the modulation of intracellular Ca(2+) signaling.
Collapse
Affiliation(s)
- Mark K Lukewich
- Department of Biological Sciences, Brock University, St. Catharines, Ontario, Canada
| | - Richard C Rogers
- Laboratory for Autonomic Neuroscience, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Alan E Lomax
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada; Department of Medicine, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
17
|
Rao JS, Kim HW, Harry GJ, Rapoport SI, Reese EA. RETRACTED: Increased neuroinflammatory and arachidonic acid cascade markers, and reduced synaptic proteins, in the postmortem frontal cortex from schizophrenia patients. Schizophr Res 2013; 147:24-31. [PMID: 23566496 PMCID: PMC3812915 DOI: 10.1016/j.schres.2013.02.017] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 02/12/2013] [Accepted: 02/19/2013] [Indexed: 12/22/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal). This article has been retracted at the request of the Editors. The National Institutes of Health has found that Dr. Jagadeesh S. Rao engaged in research misconduct by falsifying data. Data in Figures 1A, 1E, 3E and 3F were falsified. Dr. Rao was solely responsible for the falsification. None of the other authors are implicated in any way.
Collapse
Affiliation(s)
- Jagadeesh Sridhara Rao
- Brain Physiology and Metabolism Section, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA.
| | - Hyung-Wook Kim
- Brain Physiology and Metabolism Section, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Gaylia Jean Harry
- Laboratory of Toxicology and Pharmacology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Stanley Isaac Rapoport
- Brain Physiology and Metabolism Section, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Edmund Arthur Reese
- Brain Physiology and Metabolism Section, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
18
|
Wang H, Zuo X, Wang Q, Yu Y, Xie L, Wang H, Wu H, Xie W. Nicorandil inhibits hypoxia-induced apoptosis in human pulmonary artery endothelial cells through activation of mitoKATP and regulation of eNOS and the NF-κB pathway. Int J Mol Med 2013; 32:187-94. [PMID: 23670355 DOI: 10.3892/ijmm.2013.1379] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Accepted: 03/12/2013] [Indexed: 11/05/2022] Open
Abstract
Apoptosis of human pulmonary artery endothelial cells (HPAECs) is the initial step and triggering event for pulmonary hypertension (PH). However, little is known about the actions of nicorandil on HPAECs in vitro. In the present study, we investigated the anti-apoptotic effect of nicorandil on HPAECs exposed to hypoxia, and explored the underlying mechanism(s) of action. Cell viability was assessed using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT). Annexin V and propidium iodide staining, and Hoechst 33342 staining assay were employed to detect apoptosis. In addition, the protein expression of Bax, Bcl-2, caspase-9 and -3, endothelial nitric oxide synthase (eNOS), nuclear factor-κB (NF-κB) and IκBα were determined by western blotting to investigate the possible mechanisms. We found that exposure to hypoxia for 24 h significantly decreased cell viability and increased cell apoptosis. Pretreatment with nicorandil (100 µM) effectively abolished the influence of hypoxia on HPAECs. However, these protective effects of nicorandil were significantly inhibited by an antagonist of mitochondrial adenosine triphosphate-sensitive potassium (mitoKATP) channels, 5-hydroxydecanoate (5-HD, 500 µM), and by an eNOS inhibitor, NG-nitro-L-arginine methyl ester (L-NAME, 300 µM). We further observed that nicorandil could upregulate the decreased protein expression of eNOS and IκBα, and downregulate the increased protein expression of NF-κB, induced by hypoxia. In addition, nicorandil inhibited the enhancement of caspase-3 and -9 expression, and the increase in the Bax/Bcl-2 expression ratio, induced by hypoxia. However, these effects were also abolished by 5-HD and L-NAME. Collectively, these findings suggest that nicorandil inhibits hypoxia-induced apoptosis of HPAECs through activation of mitoKATP channels and increased eNOS expression, which in turn inhibits the NF-κB pathway and the mitochondrial apoptotic pathway.
Collapse
Affiliation(s)
- Hui Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Nitric oxide donors as neuroprotective agents after an ischemic stroke-related inflammatory reaction. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:297357. [PMID: 23691263 PMCID: PMC3649699 DOI: 10.1155/2013/297357] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 02/26/2013] [Accepted: 02/27/2013] [Indexed: 12/17/2022]
Abstract
Cerebral ischemia initiates a cascade of detrimental events including glutamate-associated excitotoxicity, intracellular calcium accumulation, formation of Reactive oxygen species (ROS), membrane lipid degradation, and DNA damage, which lead to the disruption of cellular homeostasis and structural damage of ischemic brain tissue. Cerebral ischemia also triggers acute inflammation, which exacerbates primary brain damage. Therefore, reducing oxidative stress (OS) and downregulating the inflammatory response are options that merit consideration as potential therapeutic targets for ischemic stroke. Consequently, agents capable of modulating both elements will constitute promising therapeutic solutions because clinically effective neuroprotectants have not yet been discovered and no specific therapy for stroke is available to date. Because of their ability to modulate both oxidative stress and the inflammatory response, much attention has been focused on the role of nitric oxide donors (NOD) as neuroprotective agents in the pathophysiology of cerebral ischemia-reperfusion injury. Given their short therapeutic window, NOD appears to be appropriate for use during neurosurgical procedures involving transient arterial occlusions, or in very early treatment of acute ischemic stroke, and also possibly as complementary treatment for neurodegenerative diseases such as Parkinson or Alzheimer, where oxidative stress is an important promoter of damage. In the present paper, we focus on the role of NOD as possible neuroprotective therapeutic agents for ischemia/reperfusion treatment.
Collapse
|
20
|
Keleshian VL, Modi HR, Rapoport SI, Rao JS. Aging is associated with altered inflammatory, arachidonic acid cascade, and synaptic markers, influenced by epigenetic modifications, in the human frontal cortex. J Neurochem 2013; 125:63-73. [PMID: 23336521 PMCID: PMC3606672 DOI: 10.1111/jnc.12153] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Revised: 12/17/2012] [Accepted: 12/18/2012] [Indexed: 01/12/2023]
Abstract
Aging is a risk factor for Alzheimer's disease (AD) and is associated with cognitive decline. However, underlying molecular mechanisms of brain aging are not clear. Recent studies suggest epigenetic influences on gene expression in AD, as DNA methylation levels influence protein and mRNA expression in postmortem AD brain. We hypothesized that some of these changes occur with normal aging. To test this hypothesis, we measured markers of the arachidonic acid (AA) cascade, neuroinflammation, pro- and anti-apoptosis factors, and gene specific epigenetic modifications in postmortem frontal cortex from nine middle-aged [41 ± 1 (SEM) years] and 10 aged subjects (70 ± 3 years). The aged compared with middle-aged brain showed elevated levels of neuroinflammatory and AA cascade markers, altered pro and anti-apoptosis factors and loss of synaptophysin. Some of these changes correlated with promoter hypermethylation of brain derived neurotrophic factor (BDNF), cyclic AMP responsive element binding protein (CREB), and synaptophysin and hypomethylation of BCL-2 associated X protein (BAX). These molecular alterations in aging are different from or more subtle than changes associated with AD pathology. The degree to which they are related to changes in cognition or behavior during normal aging remains to be evaluated.
Collapse
Affiliation(s)
- Vasken L. Keleshian
- Brain Physiology and Metabolism Section, Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Hiren R. Modi
- Brain Physiology and Metabolism Section, Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Stanley I. Rapoport
- Brain Physiology and Metabolism Section, Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Jagadeesh S. Rao
- Brain Physiology and Metabolism Section, Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
21
|
Guo S, Zhou Y, Xing C, Lok J, Som AT, Ning M, Ji X, Lo EH. The vasculome of the mouse brain. PLoS One 2012; 7:e52665. [PMID: 23285140 PMCID: PMC3527566 DOI: 10.1371/journal.pone.0052665] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 11/20/2012] [Indexed: 01/08/2023] Open
Abstract
The blood vessel is no longer viewed as passive plumbing for the brain. Increasingly, experimental and clinical findings suggest that cerebral endothelium may possess endocrine and paracrine properties – actively releasing signals into and receiving signals from the neuronal parenchyma. Hence, metabolically perturbed microvessels may contribute to central nervous system (CNS) injury and disease. Furthermore, cerebral endothelium can serve as sensors and integrators of CNS dysfunction, releasing measurable biomarkers into the circulating bloodstream. Here, we define and analyze the concept of a brain vasculome, i.e. a database of gene expression patterns in cerebral endothelium that can be linked to other databases and systems of CNS mediators and markers. Endothelial cells were purified from mouse brain, heart and kidney glomeruli. Total RNA were extracted and profiled on Affymetrix mouse 430 2.0 micro-arrays. Gene expression analysis confirmed that these brain, heart and glomerular preparations were not contaminated by brain cells (astrocytes, oligodendrocytes, or neurons), cardiomyocytes or kidney tubular cells respectively. Comparison of the vasculome between brain, heart and kidney glomeruli showed that endothelial gene expression patterns were highly organ-dependent. Analysis of the brain vasculome demonstrated that many functionally active networks were present, including cell adhesion, transporter activity, plasma membrane, leukocyte transmigration, Wnt signaling pathways and angiogenesis. Analysis of representative genome-wide-association-studies showed that genes linked with Alzheimer’s disease, Parkinson’s disease and stroke were detected in the brain vasculome. Finally, comparison of our mouse brain vasculome with representative plasma protein databases demonstrated significant overlap, suggesting that the vasculome may be an important source of circulating signals in blood. Perturbations in cerebral endothelial function may profoundly affect CNS homeostasis. Mapping and dissecting the vasculome of the brain in health and disease may provide a novel database for investigating disease mechanisms, assessing therapeutic targets and exploring new biomarkers for the CNS.
Collapse
Affiliation(s)
- Shuzhen Guo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail: (SG); (EHL)
| | - Yiming Zhou
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Broad Institute, Massachusetts Institute of Technology and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Changhong Xing
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Josephine Lok
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Angel T. Som
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - MingMing Ning
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Clinical Proteomics Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Xunming Ji
- Cerebrovascular Research Center, XuanWu Hospital, Capital Medical University, Beijing, Peoples Republic of China
| | - Eng H. Lo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Clinical Proteomics Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail: (SG); (EHL)
| |
Collapse
|
22
|
Rao JS, Kellom M, Kim HW, Rapoport SI, Reese EA. Neuroinflammation and synaptic loss. Neurochem Res 2012; 37:903-10. [PMID: 22311128 PMCID: PMC3478877 DOI: 10.1007/s11064-012-0708-2] [Citation(s) in RCA: 169] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Revised: 01/08/2012] [Accepted: 01/11/2012] [Indexed: 01/14/2023]
Abstract
Neuroinflammation plays a critical role in the progression of many neurodegenerative, neuropsychiatric and viral diseases. In neuroinflammation, activated microglia and astrocytes release cytokines and chemokines as well as nitric oxide, which in turn activate many signal transduction pathways. The cytokines, interleukin-1 beta and tumor necrosis factor alpha, regulate transcription of a number of genes within the brain, which can lead to the formation of pro-inflammatory products of the arachidonic acid cascade. Formation of pro-inflammatory agents and associated cytotoxic products during neuroinflammation can be detrimental to neurons by altering synaptic proteins. Neuroinflammation as well as excitotoxic insults reduce synaptic markers such as synaptophysin and drebrin. Neurodegenerative, neuropsychiatric illnesses and viral infections are accompanied by loss of both pre- and post-synaptic proteins. These synaptic changes may contribute to the progressive cognitive decline and behavioral changes associated with these illnesses.
Collapse
Affiliation(s)
- Jagadeesh S Rao
- Brain Physiology and Metabolism Section, National Institute on Aging, National Institutes of Health, Bldg. 9, Rm. 1S126 MSC 0947, Bethesda, MD 20892-0947, USA.
| | | | | | | | | |
Collapse
|
23
|
Rao JS, Kim HW, Kellom M, Greenstein D, Chen M, Kraft AD, Harry GJ, Rapoport SI, Basselin M. Increased neuroinflammatory and arachidonic acid cascade markers, and reduced synaptic proteins, in brain of HIV-1 transgenic rats. J Neuroinflammation 2011; 8:101. [PMID: 21846384 PMCID: PMC3175175 DOI: 10.1186/1742-2094-8-101] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Accepted: 08/16/2011] [Indexed: 12/23/2022] Open
Abstract
Background Cognitive impairment has been reported in human immune deficiency virus-1- (HIV-1-) infected patients as well as in HIV-1 transgenic (Tg) rats. This impairment has been linked to neuroinflammation, disturbed brain arachidonic acid (AA) metabolism, and synapto-dendritic injury. We recently reported upregulated brain AA metabolism in 7- to 9-month-old HIV-1 Tg rats. We hypothesized that these HIV-1 Tg rats also would show upregulated brain inflammatory and AA cascade markers and a deficit of synaptic proteins. Methods We measured protein and mRNA levels of markers of neuroinflammation and the AA cascade, as well as pro-apoptotic factors and synaptic proteins, in brains from 7- to 9-month-old HIV-1 Tg and control rats. Results Compared with control brain, HIV-1 Tg rat brain showed immunoreactivity to glycoprotein 120 and tat HIV-1 viral proteins, and significantly higher protein and mRNA levels of (1) the inflammatory cytokines interleukin-1β and tumor necrosis factor α, (2) the activated microglial/macrophage marker CD11b, (3) AA cascade enzymes: AA-selective Ca2+-dependent cytosolic phospholipase A2 (cPLA2)-IVA, secretory sPLA2-IIA, cyclooxygenase (COX)-2, membrane prostaglandin E2 synthase, 5-lipoxygenase (LOX) and 15-LOX, cytochrome p450 epoxygenase, and (4) transcription factor NF-κBp50 DNA binding activity. HIV-1 Tg rat brain also exhibited signs of cell injury, including significantly decreased levels of brain-derived neurotrophic factor (BDNF) and drebrin, a marker of post-synaptic excitatory dendritic spines. Expression of Ca2+-independent iPLA2-VIA and COX-1 was unchanged. Conclusions HIV-1 Tg rats show elevated brain markers of neuroinflammation and AA metabolism, with a deficit in several synaptic proteins. These changes are associated with viral proteins and may contribute to cognitive impairment. The HIV-1 Tg rat may be a useful model for understanding progression and treatment of cognitive impairment in HIV-1 patients.
Collapse
Affiliation(s)
- Jagadeesh Sridhara Rao
- Brain Physiology and Metabolism Section, National Institute on Aging, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
IkappaB-alpha expression following transient focal cerebral ischemia is modulated by nitric oxide. Brain Res 2011; 1372:145-51. [DOI: 10.1016/j.brainres.2010.11.071] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2010] [Revised: 11/18/2010] [Accepted: 11/19/2010] [Indexed: 01/29/2023]
|
25
|
Serrats J, Schiltz JC, García-Bueno B, van Rooijen N, Reyes TM, Sawchenko PE. Dual roles for perivascular macrophages in immune-to-brain signaling. Neuron 2010; 65:94-106. [PMID: 20152116 DOI: 10.1016/j.neuron.2009.11.032] [Citation(s) in RCA: 195] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/20/2009] [Indexed: 11/25/2022]
Abstract
Cytokines produced during infection/inflammation activate adaptive central nervous system (CNS) responses, including acute stress responses mediated by the hypothalamo-pituitary-adrenal (HPA) axis. The mechanisms by which cytokines engage HPA control circuitry remain unclear, though stimulated release of prostanoids from neighboring vascular cells has been implicated in this regard. How specific vascular cell types, endothelial cells (ECs) versus perivascular cells (PVCs; a subset of brain-resident macrophages), participate in this response remains unsettled. We exploited the phagocytic activity of PVCs to deplete them in rats by central injection of a liposome-encapsulated proapoptotic drug. This manipulation abrogated CNS and hormonal indices of HPA activation under immune challenge conditions (interleukin-1) that activated prostanoid synthesis only in PVCs, while enhancing these responses to stimuli (lipopolysaccharide) that engaged prostanoid production by ECs as well. Thus, PVCs provide both prostanoid-mediated drive to the HPA axis and an anti-inflammatory action that constrains endothelial and overall CNS responses to inflammatory insults.
Collapse
Affiliation(s)
- Jordi Serrats
- Laboratory of Neuronal Structure and Function, The Salk Institute for Biological Studies and The Clayton Medical Research Foundation, La Jolla, CA 92037, USA
| | | | | | | | | | | |
Collapse
|
26
|
Gaskin FS, Kamada K, Yusof M, Durante W, Gross G, Korthuis RJ. AICAR preconditioning prevents postischemic leukocyte rolling and adhesion: role of K(ATP) channels and heme oxygenase. Microcirculation 2009; 16:167-76. [PMID: 19152177 DOI: 10.1080/10739680802355897] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE We previously demonstrated that pharmacologic activation of AMP-activated protein kinase (AMPK) with 5-aminoimidazole-4-carboxamide 1-beta-D-ribofuranoside (AICAR) 24 hours prior to (AICAR preconditioning; AICAR-PC) ischemia/reperfusion (I/R) prevents postischemic leukocyte-endothelial cell adhesive interactions (LEI) by a mechanism initiated by endothelial nitric oxide synthase (eNOS)-dependent NO production during the period of AICAR-PC. The major aim of this study was to examine the role of ATP-sensitive potassium (K(ATP)) channels and heme oxygenase as mediators of the antiadhesive effects of AICAR-PC during I/R 24 hours later. METHODS Intravital fluorescence microscopy was used to quantify LEI in the small intestine of AICAR-preconditioned C57BL/6J mice treated with K(ATP) channel or heme oxygenase inhibitors during I/R 24 hours after AICAR-PC in separate experiments. RESULTS I/R induced marked increases in LEI relative to sham control mice, proadhesive responses that were prevented by AICAR-PC 24 hours prior to I/R. The effects of AICAR-PC to prevent postischemic LEI were abolished by K(ATP) channel or heme oxygenase inhibition during I/R. DISCUSSION/CONCLUSION Our results indicate that the antiadhesive effects of AICAR-PC are mediated by K(ATP) channel- and heme oxygenase-dependent mechanisms during I/R.
Collapse
Affiliation(s)
- F Spencer Gaskin
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, One Hospital Drive, Columbia, MO 65212, USA
| | | | | | | | | | | |
Collapse
|
27
|
Yang GY, Taboada S, Liao J. Induced nitric oxide synthase as a major player in the oncogenic transformation of inflamed tissue. Methods Mol Biol 2009; 512:119-156. [PMID: 19347276 DOI: 10.1007/978-1-60327-530-9_8] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Nitric oxide (NO) is a free radical that is involved in the inflammatory process and carcinogenesis. There are four nitric oxide synthase enzymes involved in NO production: induced nitric oxide synthase (iNOS), endothelial NO synthase (eNOS), neural NO synthase (nNOS), and mitochondrial NOS. iNOS is an inducible and key enzyme in the inflamed tissue. Recent literatures indicate that NO as well as iNOS and eNOS can modulate cancer-related events including nitro-oxidative stress, apoptosis, cell cycle, angio-genesis, invasion, and metastasis. This chapter focuses on linking NO/iNOS/eNOS to inflammation and carcinogenesis from experimental evidence to potential targets on cancer prevention and treatment.
Collapse
Affiliation(s)
- Guang-Yu Yang
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | | |
Collapse
|
28
|
Hayley S, Mangano E, Strickland M, Anisman H. Lipopolysaccharide and a social stressor influence behaviour, corticosterone and cytokine levels: divergent actions in cyclooxygenase-2 deficient mice and wild type controls. J Neuroimmunol 2008; 197:29-36. [PMID: 18455806 DOI: 10.1016/j.jneuroim.2008.03.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2007] [Revised: 03/11/2008] [Accepted: 03/20/2008] [Indexed: 02/02/2023]
Abstract
Administration of the endotoxin, lipopolysaccharide (LPS) diminished motor activity and increased plasma corticosterone as well as circulating levels of interleukin-1beta (IL-1beta), IL-6, tumor necrosis-factor-alpha (TNF-alpha) and IL-10. Among cyclooxygenase-2 (COX-2) knockout mice the behavioural, corticosterone and cytokine variations promoted by LPS were moderately (home cage activity, corticosterone, TNF-alpha) or largely (IL-6) reduced. However, if mice were exposed to a psychosocial stressor (social disruption associated with grouping mice with novel cage-mates after a period of isolation) coupled with LPS treatment, then the effects of the COX-2 deletion were absent, or there was a synergistic or additive elevation apparent (e.g., in the case of TNF-alpha, IL-6 and corticosterone). Evidently, COX-2 deletion may have either pro- or anti-inflammatory actions, depending upon the psychosocial context in which immune activation occurs.
Collapse
Affiliation(s)
- Shawn Hayley
- Institute of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, Canada K1S 5B6.
| | | | | | | |
Collapse
|
29
|
MyD88 signaling in brain endothelial cells is essential for the neuronal activity and glucocorticoid release during systemic inflammation. Mol Psychiatry 2008; 13:480-97. [PMID: 18180766 DOI: 10.1038/sj.mp.4002122] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Activation of neuronal circuits involved in the control of autonomic responses is critical for the host survival to immune threats. The brain vascular system plays a key role in such immune-CNS communication, but the signaling pathway and exact type of cells within the blood-brain barrier (BBB) mediating these functions have yet to be uncovered. To elucidate this issue we used myeloid differentiation factor 88 (MyD88)-deficient mice, because these animals do not show any responses to the cytokine interleukin-1beta (IL-1beta). We created chimeric mice with competent MyD88 signaling in either the BBB endothelium or perivascular microglia of bone marrow origin and challenged them with IL-1beta. Systemic treatment with the cytokine caused a robust transcriptional activation of genes involved in the prostaglandin E(2) (PGE(2)) production by vascular cells of the brain. Upregulation of these genes is dependent on a functional MyD88 signaling in the endothelium, because MyD88-deficient mice that received bone marrow stem cells from wild-type animals (for example, functional perivascular microglia) exhibited no response to systemic IL-1beta administration. MyD88 competent endothelial cells also mediate neuronal activation and plasma release of glucocorticoids, whereas chimeric mice with MyD88-competent perivascular microglia did not show a significant increase of these functions. Moreover, competent endothelial cells for the gene encoding Toll-like receptor 4 (TLR4) are essential for the release of plasma corticosterone in response to low and high doses of lipopolysaccharide. Therefore, BBB endothelial cells and not perivascular microglia are the main target of circulating inflammatory mediators to activate the brain circuits and key autonomic functions during systemic immune challenges.
Collapse
|
30
|
Brain Response to Endotoxin. ACTA ACUST UNITED AC 2008. [DOI: 10.1016/s1567-7443(07)10021-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
31
|
McCarty MF. Scavenging of peroxynitrite-derived radicals by flavonoids may support endothelial NO synthase activity, contributing to the vascular protection associated with high fruit and vegetable intakes. Med Hypotheses 2008; 70:170-81. [PMID: 17825500 DOI: 10.1016/j.mehy.2005.09.058] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2005] [Accepted: 09/07/2005] [Indexed: 01/24/2023]
Abstract
Ample intakes of fruit and vegetables have been linked epidemiologically with reduced risk for coronary disease, stroke, hypertension, obesity, many types of cancer, chronic pulmonary disease, osteoporosis, and various ocular disorders. The favorable impact of diets rich in fruit and vegetables on coronary risk has been confirmed in meta-analyses, and is thought to be largely attributable to the folk acid and potassium supplied by these foods. Although high intakes of vitamin C appear to confer some cardiovascular protection, the amounts supplied by typical diets may be too low to be of much benefit in this regard. High flavonoid intakes emerge as protective in some epidemiological studies, albeit the dose-response pattern observed is often L-shaped - seemingly more consistent with low intakes being harmful, than with high intakes being protective. Nonetheless, flavonoids have shown anti-atherogenic activity in rodent models, and both clinical and rodent supplementation studies with foods and food extracts rich in flavonoids demonstrate improvements in endothelium-dependent vasodilation traceable to increased endothelial nitric oxide synthesis. However, flavonoids do not appear to increase the expression of endothelial NO synthase, nor do they modify endothelial superoxide production. A likely explanation is that, even in nanomolar concentrations achievable in vivo, flavonoids can act as efficient scavengers of peroxynitrite-derived radicals, thereby protecting the cofactor tetrahydrobiopterin, crucial for NO synthase activity. Studies with cultured endothelial cells should be useful for evaluating this possibility. It would also be appropriate to assess the effects of flavonoids on prostacylin synthetase activity, on endothelial catabolism of asymmetric dimethylarginine, and on signaling mechanisms that activate NO synthase. Since peroxynitrite can induce mutagenic damage to DNA, it is conceivable that scavenging of peroxynitrite-derived radicals contributes to the reduction in mutagenesis associated with high intakes of fruits and vegetables. Carotenoids also have the potential to prevent peroxynitrite-mediated damage, although, as contrasted with flavonoids, there is comparatively little evidence that these compounds are anti-atherogenic or beneficial for endothelial function; a recent meta-analysis of epidemiological studies suggests that high lutein intakes may modestly reduce coronary risk.
Collapse
Affiliation(s)
- Mark F McCarty
- Natural Alternatives International, 1185 Linda Vista Dr., San Marcos, CA 92078, United States.
| |
Collapse
|
32
|
Delahaye NF, Coltel N, Puthier D, Barbier M, Benech P, Joly F, Iraqi FA, Grau GE, Nguyen C, Rihet P. Gene expression analysis reveals early changes in several molecular pathways in cerebral malaria-susceptible mice versus cerebral malaria-resistant mice. BMC Genomics 2007; 8:452. [PMID: 18062806 PMCID: PMC2246131 DOI: 10.1186/1471-2164-8-452] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2007] [Accepted: 12/06/2007] [Indexed: 11/10/2022] Open
Abstract
Background Microarray analyses allow the identification and assessment of molecular signatures in whole tissues undergoing pathological processes. To better understand cerebral malaria pathogenesis, we investigated intra-cerebral gene-expression profiles in well-defined genetically cerebral malaria-resistant (CM-R) and CM-susceptible (CM-S) mice, upon infection by Plasmodium berghei ANKA (PbA). We investigated mouse transcriptional responses at early and late stages of infection by use of cDNA microarrays. Results Through a rigorous statistical approach with multiple testing corrections, we showed that PbA significantly altered brain gene expression in CM-R (BALB/c), and in CM-S (CBA/J and C57BL/6) mice, and that 327 genes discriminated between early and late infection stages, between mouse strains, and between CM-R and CM-S mice. We further identified 104, 56, 84 genes with significant differential expression between CM-R and CM-S mice on days 2, 5, and 7 respectively. The analysis of their functional annotation indicates that genes involved in metabolic energy pathways, the inflammatory response, and the neuroprotection/neurotoxicity balance play a major role in cerebral malaria pathogenesis. In addition, our data suggest that cerebral malaria and Alzheimer's disease may share some common mechanisms of pathogenesis, as illustrated by the accumulation of β-amyloid proteins in brains of CM-S mice, but not of CM-R mice. Conclusion Our microarray analysis highlighted marked changes in several molecular pathways in CM-S compared to CM-R mice, particularly at early stages of infection. This study revealed some promising areas for exploration that may both provide new insight into the knowledge of CM pathogenesis and the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Nicolas F Delahaye
- Laboratoire de Pharmacogénétique des maladies parasitaires-EA864, Université de la Méditerranée, IFR48, Marseille, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Bivol LM, Berge RK, Iversen BM. Tetradecylthioacetic acid prevents the inflammatory response in two-kidney, one-clip hypertension. Am J Physiol Regul Integr Comp Physiol 2007; 294:R438-47. [PMID: 18032469 DOI: 10.1152/ajpregu.00590.2007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
ANG II promotes inflammation through nuclear factor-kappaB (NF-kappaB)-mediated induction of cytokines and reactive oxygen species (ROS). The aim of the present study was to examine the effect of tetradecylthioacetic acid (TTA), a modified fatty acid, on NF-kappaB, proinflammatory markers, ROS, and nitric oxide (NO) production in two-kidney, one-clip (2K1C) hypertension. The 2K1C TTA-treated group had lower blood pressure (128 +/- 3 mmHg) compared with 2K1C nontreated (178 +/- 5 mmHg, P < 0.001). The p50 and p65 subunits of NF-kappaB were higher in the clipped kidney (0.44 +/- 0.01 and 0.22 +/- 0.01, respectively) compared with controls (0.25 +/- 0.03 and 0.12 +/- 0.02, respectively, P < 0.001). In the 2K1C TTA-treated group, these values were similar to control levels. The same pattern of response was seen in the nonclipped kidney. In 2K1C hypertension, cytokines plasma were higher than in control: TNF-alpha was 13.5 +/- 2 pg/ml (P < 0.03), IL-1beta was 58.8 +/- 10 pg/ml (P = 0.003), IL-6 was 210 +/- 33 pg/ml (P < 0.001), and monocyte chemoattractant protein-1 was 429 +/- 21 pg/ml (P = 0.04). In the 2K1C TTA-treated group, these values were similar to controls, and the same pattern was seen in the clipped kidney. Clipping increased 8-iso-PGF-2alpha (P < 0.01) and decreased NO production (P < 0.01 vs. control) in the urine. TTA treatment normalized these values. NO production was also lower in clipped and nonclipped kidney (P < 0.001). After TTA treatment, these values were similar to controls. The results indicate that TTA has a potent anti-inflammatory effect in 2K1C by inhibition of p50/p65 NF-kappaB subunit activation, reduction of cytokines production and ROS, and enhanced NO production.
Collapse
Affiliation(s)
- Liliana M Bivol
- Renal Research Group, Institute of Medicine, Haukeland Hospital, Bergen, Norway.
| | | | | |
Collapse
|
34
|
Ying L, Hofseth LJ. An emerging role for endothelial nitric oxide synthase in chronic inflammation and cancer. Cancer Res 2007; 67:1407-10. [PMID: 17308075 DOI: 10.1158/0008-5472.can-06-2149] [Citation(s) in RCA: 191] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Nitric oxide (NO) is a free radical that is involved in carcinogenesis. Recent literature indicates that endothelial NO synthase (eNOS) can modulate cancer-related events (angiogenesis, apoptosis, cell cycle, invasion, and metastasis). We review the literature linking eNOS to carcinogenesis to encourage future research assessing the role of eNOS in cancer prevention and treatment.
Collapse
Affiliation(s)
- Lei Ying
- Department of Basic Pharmaceutical Sciences, South Carolina College of Pharmacy, University of South Carolina, 770 Sumter Street, Columbia, SC 29208, USA
| | | |
Collapse
|
35
|
Gaskin FS, Kamada K, Yusof M, Korthuis RJ. 5'-AMP-activated protein kinase activation prevents postischemic leukocyte-endothelial cell adhesive interactions. Am J Physiol Heart Circ Physiol 2006; 292:H326-32. [PMID: 16935999 DOI: 10.1152/ajpheart.00744.2006] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Preconditioning (PC) with nitric oxide (NO) donors or agents that increase endothelial NO synthase (eNOS) activity 24 h before ischemia-reperfusion (I/R) prevents postischemic leukocyte rolling (LR) and stationary leukocyte adhesion (LA). Since 5'-AMP-activated protein kinase (AMPK) phosphorylates eNOS at Ser1177, resulting in activation, we postulated that AMPK activation may trigger the development of a preconditioned anti-inflammatory phenotype similar to that induced by NO donors. Wild-type (WT) C57BL/6J and eNOS(-/-) mice were treated with the AMPK agonist 5-aminoimidazole-4-carboxamide 1-beta-d-furanoside (AICAR) 30 min (early AICAR PC) or 24 h (late AICAR PC) before I/R; LR and LA were quantified in single postcapillary venules in the jejunum using intravital microscopy. I/R induced comparable marked increases in LR and LA in WT and eNOS(-/-) mice relative to sham-operated (no ischemia) animals. Late AICAR PC prevented postischemic LR and LA, whereas early AICAR PC prevented LA in WT mice. Late AICAR PC was ineffective in preventing I/R-induced LR but not LA in the eNOS(-/-) mice, and the same pattern was seen in WT animals treated with the NOS inhibitor N(omega)-nitro-l-arginine. Early AICAR PC remained effective in preventing LA in eNOS(-/-) mice. Our results indicate that both early and late PC with an AMPK agonist produces an anti-inflammatory phenotype in postcapillary venules. Since the protection afforded by late AICAR PC on postischemic LR was prevented by NOS inhibition in WT mice and absent in eNOS-deficient mice, it appears that eNOS triggers this protective effect. In stark contrast, antecedent AMPK activation prevented I/R-induced LA by an eNOS-independent mechanism.
Collapse
Affiliation(s)
- F Spencer Gaskin
- Dept. of Medical Pharmacology and Physiology, Univ. of Missouri-Columbia, 1 Hospital Dr., Columbia, MO 65212, USA.
| | | | | | | |
Collapse
|
36
|
Kaminski A, Kasch C, Zhang L, Kumar S, Sponholz C, Choi YH, Ma N, Liebold A, Ladilov Y, Steinhoff G, Stamm C. Endothelial nitric oxide synthase mediates protective effects of hypoxic preconditioning in lungs. Respir Physiol Neurobiol 2006; 155:280-5. [PMID: 16916627 DOI: 10.1016/j.resp.2006.06.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2005] [Revised: 06/16/2006] [Accepted: 06/19/2006] [Indexed: 02/01/2023]
Abstract
To elucidate the protective mechanism of whole-body hypoxic preconditioning (WHPC) on pulmonary ischemia-reperfusion injury focussing on nitric oxide synthases (NOS), mice were placed in a hypoxic chamber (FIO(2)=0.1) for 4h followed by 12h of normoxia. Then, pulmonary ischemia for 1h followed by 5h of reperfusion was performed by clamping the left hilum in vivo (I/R). WHPC protected WT mice from pulmonary leukocyte infiltration as assessed by myeloperoxidase (MPO) activity, associated with a mild further increase in endothelial permeability (Evans Blue extravasation). When all NOS isoforms were inhibited during WHPC by L-NAME, mortality and MPO activity after I/R markedly increased. To determine the responsible NOS isoform, quantitative RT-PCR was performed for eNOS and iNOS mRNA, showing that only eNOS was upregulated in response to WHPC. While eNOS total protein expression remained unchanged, the amount of phosphorylated eNOS also increased. The WHPC/IR experiments were then repeated with eNOS knockout mice. Here, we found that the protective effect of WHPC on pulmonary leukocyte sequestration was abrogated, and endothelial leakage was further exacerbated. We conclude that WHPC limits neutrophil sequestration via an eNOS-dependent mechanism, and that eNOS helps preserve endothelial permeability during hypoxia and I/R.
Collapse
Affiliation(s)
- A Kaminski
- Department of Cardiac Surgery, University of Rostock, Schillingallee 35, 18055 Rostock, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Blais V, Turrin NP, Rivest S. Cyclooxygenase 2 (COX-2) inhibition increases the inflammatory response in the brain during systemic immune stimuli. J Neurochem 2005; 95:1563-74. [PMID: 16277613 DOI: 10.1111/j.1471-4159.2005.03480.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) and inhibitors of the cyclooxygenase (COX) pathways are currently recommended for the prevention and treatment of several inflammatory diseases, including neurodegenerative disorders. However non-selective blockade of COX was found to have pro-inflammatory properties, because they have the ability to alter the plasma glucocorticoid levels that play a critical role in the control of the innate immune response. The present study investigated the role of non-selective (ketorolac or indomethacin) or specific inhibitors of COX-1 (SC-560) and COX-2 (NS-398) in these effects. Mice challenged systemically with the endotoxin lipopolysaccharide (LPS) exhibited a robust hybridization signal for numerous inflammatory genes in vascular-associated cells of the brain and microglia across the cerebral tissue. Ketorolac, indomethacin and NS-398 significantly increased the ability of LPS to trigger such an innate immune response at time 3 h post challenge, whereas SC-560 failed to change gene expression in the brain of animals treated with the endotoxin. These data together with the crucial role of COX-2-derived prostaglandin E2 (PGE2) in the increase of glucocorticoids during systemic immune stimuli provide evidence that inhibition of this pathway results in an exacerbated early innate immune reaction. This may have a major impact on the use of these drugs in diseases where inflammation is believed to be a contributing and detrimental factor.
Collapse
Affiliation(s)
- Véronique Blais
- Laboratory of Molecular Endocrinology, CHUL Research Center and Department of Anatomy and Physiology, Laval University, Québec, Canada
| | | | | |
Collapse
|
38
|
Trickler WJ, Mayhan WG, Miller DW. Brain microvessel endothelial cell responses to tumor necrosis factor-alpha involve a nuclear factor kappa B (NF-κB) signal transduction pathway. Brain Res 2005; 1048:24-31. [PMID: 15916752 DOI: 10.1016/j.brainres.2005.04.028] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2004] [Revised: 04/01/2005] [Accepted: 04/13/2005] [Indexed: 01/13/2023]
Abstract
The involvement of nuclear factor kappa B (NF-kappaB) in TNF-induced increases in cerebral microvascular permeability was evaluated both in vitro, using primary cultured bovine brain microvessel endothelial cells (BBMEC), and in vivo, using the rat cranial window model. In primary cultured BBMEC, TNF exposure resulted in an increased appearance of the Rel A subunit of NF-kappaB in immunoblots of cell lysates. Increases in the Rel A subunit of NF-kappaB were observed as early as 30-min after administration of TNF. The increased permeability and the secretion of prostaglandin E2 in response to TNF exposure in BBMEC monolayers were significantly reduced by several different NF-kappaB inhibitors, including PDTC, CAPE, BAY 11-7085, and lactacystin. Similar results were also obtained in the rat cranial window model where treatment with the COX-2 inhibitor, NS-398 (0.1 microM), or the NF-kappaB inhibitor, PDTC (10 microM), significantly reduced the permeability increases produced by TNF. These studies suggest that the increases in BBB permeability following TNF exposure are attributable to activation of an NF-kappaB-mediated signaling pathway in the cerebral microvasculature.
Collapse
Affiliation(s)
- W J Trickler
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, 986025 Nebraska Medical Center, Omaha, NE 68198-6025, USA
| | | | | |
Collapse
|
39
|
Akahoshi T, Tanigawa T, Sarfeh IJ, Chiou SK, Hashizume M, Maehara Y, Jones MK. Selective cyclooxygenase (COX) inhibition causes damage to portal hypertensive gastric mucosa: roles of nitric oxide and NF-kappaB. FASEB J 2005; 19:1163-5. [PMID: 15845610 DOI: 10.1096/fj.04-3325fje] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Portal hypertension (PHT) is associated with increased susceptibility of the gastric mucosa to injury by a variety of factors, including nonsteroidal anti-inflammatory drugs (NSAIDs) that nonselectively inhibit both isoforms of cyclooxygenase (COX-1 and -2). PHT gastric mucosa also has excessive nitric oxide (NO) production that contributes to the general increased susceptibility to injury. Using a rat model of PHT, we studied whether selective COX inhibition, which does not damage normal (normotensive) gastric mucosa, is sufficient to cause PHT gastric damage and, if so, whether and how excessive NO is involved. Indomethacin, a nonselective NSAID, caused 2.4-fold more gastric injury to PHT vs. normotensive sham-operated (SO) control rats. Neither NS-398 nor celecoxib, selective COX-2 inhibitors, caused gastric damage in either SO or PHT rats. SC-560, a selective COX-1 inhibitor, did not cause gastric damage in SO rats but dose-dependently caused gastric damage in PHT rats. There was a compensatory increase in COX-2 expression and activity in SC-560-treated SO rats but not SC-560-treated PHT rats. Partial inhibition of NO production restored gastric COX-2 expression and activity levels in SC-560-treated PHT rats to those of SC-560-treated SO rats, by a mechanism consistent with induction of NF-kappaB, and significantly reduced gastric damage. These studies indicate that, in contrast to normotensive gastric mucosa, inhibition of COX-1 alone is sufficient to cause PHT gastric damage as a result of excessive NO that prevents the induction of NF-kappaB and the compensatory increase in COX-2.
Collapse
Affiliation(s)
- Tomohiko Akahoshi
- Department of Medicine, Department of Veterans Affairs Medical Center, Long Beach, California, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Lancel S, Tissier S, Mordon S, Marechal X, Depontieu F, Scherpereel A, Chopin C, Neviere R. Peroxynitrite decomposition catalysts prevent myocardial dysfunction and inflammation in endotoxemic rats. J Am Coll Cardiol 2004; 43:2348-58. [PMID: 15193704 DOI: 10.1016/j.jacc.2004.01.047] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2003] [Revised: 01/08/2004] [Accepted: 01/12/2004] [Indexed: 11/23/2022]
Abstract
OBJECTIVES The aim of this study was to test whether peroxynitrite neutralizers would reduce peroxynitrite accumulation and improve myocardial contractile dysfunction and inflammation in endotoxin-treated rats. BACKGROUND Release of endogenous proinflammatory cytokines such as tumor necrosis factor (TNF)-alpha in response to endotoxin is responsible for the production of large amounts of nitric oxide (NO), which may exert detrimental effects on the myocardium in animal models, isolated hearts, and isolated cardiac myocytes. Recent studies have indicated that many of the deleterious effects of NO are mediated by peroxynitrite, a powerful oxidant generated from a fast diffusion-limited reaction of NO and superoxide anion. METHODS We studied the effects of peroxynitrite neutralizers, such as mercaptoethylguanidine (MEG) sodium succinate (10 mg/kg) and 5,10,15,20-tetrakis(4-sulfonatophenyl)-porphyrinato iron (III) (FeTPPS) (30 mg/kg) on peroxynitrite accumulation, in vivo endothelial cell-leukocyte activation on the mesenteric venule, and myocardial contractile dysfunction and inflammation in a model of sepsis induced by injection of endotoxin (10 mg/kg) in rats. RESULTS Mercaptoethylguanidine sodium succinate and FeTPPS largely prevented the accumulation of peroxynitrite as measured by plasma rhodamine fluorescence and heart nitrotyrosine staining. Interestingly, MEG sodium succinate and FeTPPS improved endotoxin-induced myocardial contractile dysfunction, which was associated with reduced degradation of nuclear factor kappa B inhibitory protein I-kappa-B, plasma TNF-alpha levels, and microvascular endothelial cell-leukocyte activation. CONCLUSIONS These observations suggest that the beneficial effects of MEG and FeTPPS on endotoxin-induced myocardial contractile dysfunction could be related to the unique effects of these compounds on cardiovascular inflammation processes.
Collapse
Affiliation(s)
- Steve Lancel
- EA 2689, Université de Lille 2, Faculté de Médecine, Lille, France
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Sackner MA, Gummels EM, Adams JA. Say NO to fibromyalgia and chronic fatigue syndrome: an alternative and complementary therapy to aerobic exercise. Med Hypotheses 2004; 63:118-23. [PMID: 15193362 DOI: 10.1016/j.mehy.2004.01.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2004] [Accepted: 01/31/2004] [Indexed: 11/24/2022]
Abstract
Increased shear stress to the endothelium increases activity of endothelial nitric oxide synthase (eNOS) with subsequent release of small quantities (nMol) of nitric oxide (NO) into the circulation. It occurs during moderate aerobic exercise mostly as a result of laminar shear stress and with whole body, periodic acceleration as a result of pulsatile shear stress. The latter is administered by means of a new, non-invasive, passive exercise device. Moderate exercise has long been known to alleviate the symptoms of fibromyalgia and chronic fatigue syndrome and in the current study, whole body, periodic acceleration did as well. Since NO through action of eNOS has potent anti-inflammatory properties mainly by suppressing nuclear factor kappabeta activity, it is hypothesized that both diseases have chronic inflammation as their basis. Whole body periodic acceleration can be applied separately or supplementary to aerobic exercise in the treatment of fibromyalgia and chronic fatigue syndrome.
Collapse
Affiliation(s)
- Marvin A Sackner
- Mt. Sinai Medical Center of Greater Miami, Division of Pulmonary Disease and Critical Care Medicine, Miami Beach, FL 33140, USA.
| | | | | |
Collapse
|
42
|
Selley ML. Increased concentrations of homocysteine and asymmetric dimethylarginine and decreased concentrations of nitric oxide in the plasma of patients with Alzheimer's disease. Neurobiol Aging 2003; 24:903-7. [PMID: 12928048 DOI: 10.1016/s0197-4580(03)00007-1] [Citation(s) in RCA: 107] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Vascular risk factors increase the risk of developing Alzheimer's disease. Increased concentrations of circulating homocysteine are associated with an increased risk of both vascular disease and Alzheimer's disease. Asymmetric dimethylarginine (ADMA) is an endogenous inhibitor of nitric oxide synthase. There is an increase in the concentration of ADMA in the circulation in vascular disease. We measured the concentrations of homocysteine, ADMA and nitric oxide (as nitrate and nitrite) in the plasma of 25 patients with Alzheimer's disease and 25 control subjects. There was a highly significant increase in the plasma concentration of homocysteine (P<0.001) and ADMA (P<0.0001) and a highly significant decrease in the plasma concentration of nitric oxide (P<0.0001) among the Alzheimer's patients. In the combined patient and control groups a highly significant positive correlation was found between the plasma concentrations of homocysteine and ADMA (r=0.782, P<0.0001). In addition, significant negative correlations were detected between the plasma concentration of nitric oxide and the plasma concentration of homocysteine (r=-0.592, P<0.0001) and ADMA (r=-0.789, P<0.0001). These significant correlations were found to persist, even when they were restricted to the Alzheimer's patients. The inhibition of endothelial nitric oxide synthesis by ADMA impairs cerebral blood flow, which may contribute to the development of Alzheimer's disease. Endothelial dysfunction is also associated with atherosclerosis and stroke, which are important risk factors for Alzheimer's disease. Inflammation plays an important role in Alzheimer's disease and the inhibition of endothelial nitric oxide by ADMA may increase the concentration of inflammatory mediators in the brain. The inhibition of neuronal nitric oxide synthesis by ADMA may cause cognitive dysfunction in Alzheimer's disease.
Collapse
Affiliation(s)
- M L Selley
- Angiogen Pharmaceuticals Pty. Ltd., P.O. Box 512, Turramurra, NSW 2074, Australia.
| |
Collapse
|
43
|
Zauli G, Pandolfi A, Gonelli A, Di Pietro R, Guarnieri S, Ciabattoni G, Rana R, Vitale M, Secchiero P. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) sequentially upregulates nitric oxide and prostanoid production in primary human endothelial cells. Circ Res 2003; 92:732-40. [PMID: 12649264 DOI: 10.1161/01.res.0000067928.83455.9c] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Endothelial cells express tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) receptors, but the function of TRAIL in endothelial cells is not completely understood. We explored the role of TRAIL in regulation of key intracellular signal pathways in endothelial cells. The addition of TRAIL to primary human endothelial cells increased phosphorylation of endothelial nitric oxide synthase (eNOS), NOS activity, and NO synthesis. Moreover, TRAIL induced cell migration and cytoskeleton reorganization in an NO-dependent manner. TRAIL did not activate the NF-kappaB or COX-2 pathways in endothelial cells. Instead, TRAIL increased prostanoid production (PGE2=PGI2>TXA2), which was preferentially inhibited by the COX-1 inhibitor SC-560. Because NO and prostanoids play a crucial role in the state of blood vessel vasodilatation and angiogenesis, our data suggest that TRAIL might play an important role in endothelial cell function.
Collapse
Affiliation(s)
- Giorgio Zauli
- Department of Normal Human Morphology, University of Trieste, Via Manzoni 16, 34138 Trieste.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Michiels C, Minet E, Mottet D, Raes M. Regulation of gene expression by oxygen: NF-kappaB and HIF-1, two extremes. Free Radic Biol Med 2002; 33:1231-42. [PMID: 12398931 DOI: 10.1016/s0891-5849(02)01045-6] [Citation(s) in RCA: 149] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Aerobic life is dependent on molecular oxygen for ATP regeneration, but only possible in a narrow range of oxygen concentrations. Increased oxygen tension is toxic through the generation of reactive oxygen species (ROS), while a decrease in oxygen concentration impairs energy availability and, hence, cell viability. Cells have developed strategies to respond to changes in oxygen tension: specific systems detect excessive ROS and hypoxia, leading to the activation of specific transcription factors and expression of appropriate target genes. The aim of this review is to describe how hypoxia-inducible factor-1 (HIF-1) and nuclear factor-kappaB (NF-kappaB) are regulated and what could be the sensors to the changes in oxygen levels. Some of the physiological responses initiated by these transcription factors are also mentioned.
Collapse
Affiliation(s)
- Carine Michiels
- Biochemistry and Cellular Biology Laboratory, University of Namur, Belgium.
| | | | | | | |
Collapse
|
45
|
Lugarini F, Hrupka BJ, Schwartz GJ, Plata-Salaman CR, Langhans W. A role for cyclooxygenase-2 in lipopolysaccharide-induced anorexia in rats. Am J Physiol Regul Integr Comp Physiol 2002; 283:R862-8. [PMID: 12228055 DOI: 10.1152/ajpregu.00200.2002] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Because nonselective cycloooxygenase (COX) inhibition attenuated anorexia after lipopolysaccharide (LPS) administration, we tested the ability of resveratrol (2.5, 10, and 40 mg/kg) and NS-398 (2.5, 10, and 40 mg/kg), selective inhibitors of the two COX isoforms COX-1 and -2, respectively, to attenuate LPS (100 microg/kg ip)-induced anorexia. NS-398 (10 and 40 mg/kg) administered with LPS at lights out attenuated LPS-induced anorexia, whereas resveratrol at all doses tested did not. Because prostaglandin (PG) E(2) is considered the major metabolite synthesized by COX, we measured plasma and cerebrospinal fluid (CSF) PGE(2) levels after LPS administration. LPS induced a time-dependent increase of PGE(2) in CSF but not in plasma. NS-398 (5, 10, and 40 mg/kg) blocked the LPS-induced increase in CSF PGE(2), whereas resveratrol (10 mg/kg) did not. These results support a role of COX-2 in mediating the anorectic response to peripheral LPS and point at PGE(2) as a potential neuromodulator involved in this response.
Collapse
Affiliation(s)
- F Lugarini
- Institute of Animal Sciences, Physiology, and Animal Husbandry, Swiss Federal Institute of Technology, 8603 Schwerzenbach, Switzerland.
| | | | | | | | | |
Collapse
|
46
|
Kuruganti PA, Hinojoza JR, Eaton MJ, Ehmann UK, Sobel RA. Interferon-beta counteracts inflammatory mediator-induced effects on brain endothelial cell tight junction molecules-implications for multiple sclerosis. J Neuropathol Exp Neurol 2002; 61:710-24. [PMID: 12152786 DOI: 10.1093/jnen/61.8.710] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
To elucidate mechanisms of endothelial cell (EC) dysfunction in CNS inflammatory responses and beneficial effects of interferon-beta (IFN-gamma) in multiple sclerosis (MS), we analyzed effects of individual and combinations of soluble inflammatory mediators on the intracellular localization of the EC tight junction-associated molecules zonula occludens-1 and -2 (ZO-1 and ZO-2) in human brain ECs. The cytoplasm in the majority of cells in control EC cultures was clear; ZO-1 and ZO-2 were localized peripherally near sites of cell contact and associated with submembranous cytoplasmic filaments. H2O2 induced reversible time- and concentration-dependent translocation of ZO-1 and ZO-2 to a random distribution within EC cytoplasm and retraction of EC borders. For low concentrations, these effects were accompanied by less prominent submembranous filaments but not by evidence of cytotoxicity, increased cell death or altered amounts of ZO-1. Tumor necrosis factor-beta induced similar alterations but interferon-y did not. Co-treatment with either cytokine increased H2O2 effects whereas IFN-beta reversed H2O2-induced effects. In control white matter samples, EC cytoplasm was clear and ZO-1 was located on cell borders. In inflammatory/demyelinating lesions, EC ZO-1 was diffuse, indicating that the alterations induced in vitro mimic those in active MS lesions. These findings suggest that in MS patients, IFN-beta treatment may counteract inflammatory mediator effects on CNS EC tight junction molecules, thereby preserving EC barrier function.
Collapse
Affiliation(s)
- Poonam A Kuruganti
- Department of Pathology, Stanford University School of Medicine, California 94305, USA
| | | | | | | | | |
Collapse
|
47
|
Abstract
Nitric oxide (NO) is a free radical found at higher than normal concentrations within inflammatory multiple sclerosis (MS) lesions. These high concentrations are due to the appearance of the inducible form of nitric oxide synthase (iNOS) in cells such as macrophages and astrocytes. Indeed, the concentrations of markers of NO production (eg, nitrate and nitrite) are raised in the CSF, blood, and urine of patients with MS. Circumstantial evidence suggests that NO has a role in several features of the disease, including disruption of the blood-brain barrier, oligodendrocyte injury and demyelination, axonal degeneration, and that it contributes to the loss of function by impairment of axonal conduction. However, despite these considerations, the net effect of NO production in MS is not necessarily deleterious because it also has several beneficial immunomodulatory effects. These dual effects may help to explain why iNOS inhibition has not provided reliable and encouraging results in animal models of MS, but alternative approaches based on the inhibition of superoxide production, partial sodium-channel blockade, or the replacement of lost immunomodulatory function, may prove beneficial.
Collapse
Affiliation(s)
- Kenneth J Smith
- Neuroinflammation Research Group, Guy's, King's, and St Thomas' School of Medicine, King's College, London SE1 1UL, UK.
| | | |
Collapse
|
48
|
Distinct brain vascular cell types manifest inducible cyclooxygenase expression as a function of the strength and nature of immune insults. J Neurosci 2002. [PMID: 12097512 DOI: 10.1523/jneurosci.22-13-05606.2002] [Citation(s) in RCA: 121] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Induced prostanoid synthesis by cells associated with the cerebral vasculature has been implicated in mediating immune system influences on the CNS, but the cell type(s) involved remain unsettled. To determine whether this might derive from differences in the nature and intensity of the stimuli used to model immune insults, immunochemical and hybridization histochemical methods were used to monitor cyclooxygenase-2 (COX-2) expression alone, or in conjunction with endothelial, perivascular, and glial cell markers, in brains of rats treated with varying doses of interleukin-1 (IL-1) or bacterial lipopolysaccharide (LPS). Vehicle-treated animals displayed weak COX-2 expression in the meninges, choroid plexus, and larger blood vessels. Rats challenged intravenously with IL-1beta (1.87-30 microgram/kg) showed a marked increase in the number of vascular-associated cells displaying COX-2-immunoreactivity (ir). More than 90% stained positively for the ED2 macrophage differentiation antigen, identifying them as perivascular cells, whereas none coexpressed endothelial or glial cell markers. Low doses of LPS (0.1 microgram/kg) elicited a similar response profile, but higher doses (2-100 microgram/kg) provoked COX-2 expression in a progressively greater number of cells exhibiting distinct round or multipolar morphologies, corresponding to cells expressing endothelial (RECA-1) or perivascular (ED2) cell antigens, respectively. Similarly, ultrastructural analysis localized COX-2-ir to the perinuclear region of endothelial cells of LPS-treated but not IL-1-treated rats. We conclude that perivascular cells exhibit the lower threshold to COX-2 expression in response to either IL-1 or endotoxin treatment, and that enzyme expression by endothelial cells requires one or more facets of the more complex immune stimulus presented by LPS.
Collapse
|
49
|
Mendes AF, Caramona MM, de Carvalho AP, Lopes MC. Diacerhein and rhein prevent interleukin-1beta-induced nuclear factor-kappaB activation by inhibiting the degradation of inhibitor kappaB-alpha. PHARMACOLOGY & TOXICOLOGY 2002; 91:22-8. [PMID: 12193257 DOI: 10.1034/j.1600-0773.2002.910104.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Diacerhein and rhein are anthraquinone compounds that ameliorate the course of osteoarthritis. Recent reports also suggest that these compounds may have antiinflammatory properties, but the cellular mechanisms by which they exert antiosteoarthritic and possibly antiinflammatory effects are still incompletely understood. The purpose of this study was to investigate the ability of diacerhein and rhein to inhibit the activation of the transcription factor nuclear factor kappaB, induced by the proinflammatory cytokine interleukin-1beta, in primary monolayer cultures of bovine articular chondrocytes. We also studied the ability of diacerhein and rhein to prevent the expression of the inducible nitric oxide synthase gene, which is driven by nuclear factor-kappaB. We observed that interleukin-1beta induced the degradation of the inhibitor kappaB-alpha protein and the translocation of the protein p65 (a member of the nuclear factor-kappaB family) to the nucleus, which were inhibited by diacerhein and rhein, in a dose-dependent manner. Interleukin-1beta-induced nuclear factor-kappaB binding to a specific (gamma-(32)P)-labelled oligonucleotide probe was also inhibited by treatment of chondrocytes with diacerhein or rhein, as revealed by electrophoretic mobility shift assay. Inducible nitric oxide synthase mRNA and protein synthesis and nitric oxide production were also inhibited by diacerhein and rhein, in a dose-dependent manner. The half-maximal inhibitory concentrations of diacerhein and rhein, relative to nitric oxide production, were 8.2 microM ;and 7.7 microM, respectively. These results suggest that diacerhein and rhein inhibit nuclear factor-kappaB activation and, consequently, the expression of nuclear factor-kappaB-dependent genes, such as the inducible nitric oxide synthase gene, which can explain their antiosteoarthritic and antiinflammatory effects.
Collapse
|