1
|
Sumi MP, Tupta B, Song K, Mavrakis L, Comhair S, Erzurum SC, Liu X, Stuehr DJ, Ghosh A. Expression of soluble guanylate cyclase (sGC) and its ability to form a functional heterodimer are crucial for reviving the NO-sGC signaling in PAH. Free Radic Biol Med 2024; 225:846-855. [PMID: 39515593 DOI: 10.1016/j.freeradbiomed.2024.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 10/28/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
In order to determine the underpinnings of a dysfunctional NO-sGC signal pathway which occurs in pulmonary arterial hypertension (PAH), we investigated pulmonary arterial smooth muscle cells (PASMCs) derived from PAH patients. We found low expression of sGC, a poor sGCα1β1 heterodimer and this correlated with low expression of its facilitator chaperon, hsp90. Treating PASMCs overnight (16 h) with low micromolar doses of a slow release NO donor DETANONOate, reinstated the sGCα1β1 heterodimer and restored its NO-heme dependent activity. Transwell co-culture of HEK cells stably expressing eNOS with PAH PASMCs also restored the sGC heterodimer and its heme-dependent activity with sGC stimulator, BAY 41-2272. To determine whether the dysfunctionality in the NO-sGC pathway stems from a dysfunctional eNOS producing negligible NO, we did transwell co-cultures of pulmonary arterial endothelial cells (PAECs) with PASMCs. Our results indicated that PAECs from both control and PAH samples when activated for eNOS restored both sGC heterodimer and its heme-dependent sGC activity in the corresponding PASMCs, suggesting that PAECs from PAH can also generate NO. In line with these results expression of eNOS, its support chaperon hsp90, its specific kinase Akt, p-Akt or post-translational modifications (PTMs) like OGlcNAc or phospho-tyrosine were unchanged in PAH relative to controls. Additionally there was uniform expression of Hbα/β and Mb in PASMCs or PAECs in PAH or controls and these globins can effectively scavenge the eNOS generated NO, as there was evidence of strong eNOS-Hb/Mb interactions. Our studies suggest that factors such as globin NO scavenging along with vascular remodeling in PAH can cause hampered vasodilation which in the face of poor NO levels as occurs in PAH are additional impediments for effective vasodilation. However importantly our studies suggests that future therapies can use low doses of NO along with sGC stimulators as a potential drug for PAH subjects.
Collapse
Affiliation(s)
- Mamta P Sumi
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, 44196, USA
| | - Blair Tupta
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, 44196, USA
| | - Kevin Song
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, 44196, USA
| | - Lori Mavrakis
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, 44196, USA
| | - Suzy Comhair
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, 44196, USA
| | - Serpil C Erzurum
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, 44196, USA
| | - Xuefeng Liu
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44196, USA
| | - Dennis J Stuehr
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, 44196, USA
| | - Arnab Ghosh
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, 44196, USA.
| |
Collapse
|
2
|
Tao H, Zhu P, Xia W, Chu M, Chen K, Wang Q, Gu Y, Lu X, Bai J, Geng D. The Emerging Role of the Mitochondrial Respiratory Chain in Skeletal Aging. Aging Dis 2024; 15:1784-1812. [PMID: 37815897 PMCID: PMC11272194 DOI: 10.14336/ad.2023.0924] [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: 08/03/2023] [Accepted: 09/24/2023] [Indexed: 10/12/2023] Open
Abstract
Maintenance of mitochondrial homeostasis is crucial for ensuring healthy mitochondria and normal cellular function. This process is primarily responsible for regulating processes that include mitochondrial OXPHOS, which generates ATP, as well as mitochondrial oxidative stress, apoptosis, calcium homeostasis, and mitophagy. Bone mesenchymal stem cells express factors that aid in bone formation and vascular growth. Positive regulation of hematopoietic stem cells in the bone marrow affects the differentiation of osteoclasts. Furthermore, the metabolic regulation of cells that play fundamental roles in various regions of the bone, as well as interactions within the bone microenvironment, actively participates in regulating bone integrity and aging. The maintenance of cellular homeostasis is dependent on the regulation of intracellular organelles, thus understanding the impact of mitochondrial functional changes on overall bone metabolism is crucially important. Recent studies have revealed that mitochondrial homeostasis can lead to morphological and functional abnormalities in senescent cells, particularly in the context of bone diseases. Mitochondrial dysfunction in skeletal diseases results in abnormal metabolism of bone-associated cells and a secondary dysregulated microenvironment within bone tissue. This imbalance in the oxidative system and immune disruption in the bone microenvironment ultimately leads to bone dysplasia. In this review, we examine the latest developments in mitochondrial respiratory chain regulation and its impacts on maintenance of bone health. Specifically, we explored whether enhancing mitochondrial function can reduce the occurrence of bone cell deterioration and improve bone metabolism. These findings offer prospects for developing bone remodeling biology strategies to treat age-related degenerative diseases.
Collapse
Affiliation(s)
- Huaqiang Tao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Jiangsu, China.
| | - Pengfei Zhu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Jiangsu, China.
| | - Wenyu Xia
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Jiangsu, China.
| | - Miao Chu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Jiangsu, China.
| | - Kai Chen
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Jiangsu, China.
| | - Qiufei Wang
- Department of Orthopedics, Changshu Hospital Affiliated to Soochow University, First People’s Hospital of Changshu City, Jiangsu, China.
| | - Ye Gu
- Department of Orthopedics, Changshu Hospital Affiliated to Soochow University, First People’s Hospital of Changshu City, Jiangsu, China.
| | - Xiaomin Lu
- Department of Oncology, Affiliated Haian Hospital of Nantong University, Jiangsu, China.
| | - Jiaxiang Bai
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Jiangsu, China.
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui, China.
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Jiangsu, China.
| |
Collapse
|
3
|
Silva KDA, Nunes JPS, Andrieux P, Brochet P, Almeida RR, Kuramoto Takara ACK, Pereira NB, Abel L, Cobat A, Zaniratto RCF, Levy D, Bydlowski SP, Cecconello I, Seguro FCBDC, Kalil J, Chevillard C, Cunha-Neto E. Chagas Disease Megaesophagus Patients Carrying Variant MRPS18B P260A Display Nitro-Oxidative Stress and Mitochondrial Dysfunction in Response to IFN-γ Stimulus. Biomedicines 2022; 10:2215. [PMID: 36140315 PMCID: PMC9496350 DOI: 10.3390/biomedicines10092215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
Chagas disease (CD), caused by the protozoan parasite Trypanosoma cruzi, affects 8 million people, and around 1/3 develop chronic cardiac (CCC) or digestive disease (megaesophagus/megacolon), while the majority remain asymptomatic, in the indeterminate form of Chagas disease (ASY). Most CCC cases in families with multiple Chagas disease patients carry damaging mutations in mitochondrial genes. We searched for exonic mutations associated to chagasic megaesophagus (CME) in genes essential to mitochondrial processes. We performed whole exome sequencing of 13 CME and 45 ASY patients. We found the damaging variant MRPS18B 688C > G P230A, in five out of the 13 CME patients (one of them being homozygous; 38.4%), while the variant appeared in one out of 45 ASY patients (2.2%). We analyzed the interferon (IFN)-γ-induced nitro-oxidative stress and mitochondrial function of EBV-transformed lymphoblastoid cell lines. We found the CME carriers of the mutation displayed increased levels of nitrite and nitrated proteins; in addition, the homozygous (G/G) CME patient also showed increased mitochondrial superoxide and reduced levels of ATP production. The results suggest that pathogenic mitochondrial mutations may contribute to cytokine-induced nitro-oxidative stress and mitochondrial dysfunction. We hypothesize that, in mutation carriers, IFN-γ produced in the esophageal myenteric plexus might cause nitro-oxidative stress and mitochondrial dysfunction in neurons, contributing to megaesophagus.
Collapse
Affiliation(s)
- Karla Deysiree Alcântara Silva
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-900, Brazil
- Division of Clinical Immunology and Allergy, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil
- Institute for Investigation in Immunology/INCT, São Paulo 05403-900, Brazil
| | - João Paulo Silva Nunes
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-900, Brazil
- Division of Clinical Immunology and Allergy, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil
- Institute for Investigation in Immunology/INCT, São Paulo 05403-900, Brazil
- Institut MarMaRa, INSERM, UMR_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, 13288 Marseille, France
| | - Pauline Andrieux
- Institut MarMaRa, INSERM, UMR_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, 13288 Marseille, France
| | - Pauline Brochet
- Institut MarMaRa, INSERM, UMR_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, 13288 Marseille, France
| | - Rafael Ribeiro Almeida
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-900, Brazil
- Institute for Investigation in Immunology/INCT, São Paulo 05403-900, Brazil
| | - Andréia Cristina Kazue Kuramoto Takara
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-900, Brazil
- Institute for Investigation in Immunology/INCT, São Paulo 05403-900, Brazil
| | - Natalia Bueno Pereira
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-900, Brazil
- Division of Clinical Immunology and Allergy, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil
| | - Laurent Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
- Imagine Institute, University of Paris, 75015 Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
| | - Aurelie Cobat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
- Imagine Institute, University of Paris, 75015 Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
| | - Ricardo Costa Fernandes Zaniratto
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-900, Brazil
| | - Débora Levy
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-900, Brazil
| | - Sergio Paulo Bydlowski
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-900, Brazil
| | - Ivan Cecconello
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-900, Brazil
| | | | - Jorge Kalil
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-900, Brazil
- Division of Clinical Immunology and Allergy, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil
- Institute for Investigation in Immunology/INCT, São Paulo 05403-900, Brazil
| | - Christophe Chevillard
- Institut MarMaRa, INSERM, UMR_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, 13288 Marseille, France
| | - Edecio Cunha-Neto
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-900, Brazil
- Division of Clinical Immunology and Allergy, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil
- Institute for Investigation in Immunology/INCT, São Paulo 05403-900, Brazil
| |
Collapse
|
4
|
Zhu S, Abudupataer M, Yan S, Wang C, Wang L, Zhu K. Construction of a high-throughput aorta smooth muscle-on-a-chip for thoracic aortic aneurysm drug screening. Biosens Bioelectron 2022; 218:114747. [DOI: 10.1016/j.bios.2022.114747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/02/2022] [Accepted: 09/21/2022] [Indexed: 11/27/2022]
|
5
|
Singh RB, Liu L, Anchouche S, Yung A, Mittal SK, Blanco T, Dohlman TH, Yin J, Dana R. Ocular redness - I: Etiology, pathogenesis, and assessment of conjunctival hyperemia. Ocul Surf 2021; 21:134-144. [PMID: 34010701 PMCID: PMC8328962 DOI: 10.1016/j.jtos.2021.05.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 01/01/2023]
Abstract
The translucent appearance of the conjunctiva allows for immediate visualization of changes in the circulation of the conjunctival microvasculature consisting of extensive branching of superficial and deep arterial systems and corresponding drainage pathways, and the translucent appearance of the conjunctiva allows for immediate visualization of changes in the circulation. Conjunctival hyperemia is caused by a pathological vasodilatory response of the microvasculature in response to inflammation due to a myriad of infectious and non-infectious etiologies. It is one of the most common contributors of ocular complaints that prompts visits to medical centers. Our understanding of these neurogenic and immune-mediated pathways has progressed over time and has played a critical role in developing targeted novel therapies. Due to a multitude of underlying etiologies, patients must be accurately diagnosed for efficacious management of conjunctival hyperemia. The diagnostic techniques used for the grading of conjunctival hyperemia have also evolved from descriptive and subjective grading scales to more reliable computer-based objective grading scales.
Collapse
Affiliation(s)
- Rohan Bir Singh
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Lingjia Liu
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Sonia Anchouche
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Ann Yung
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Sharad K Mittal
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Tomas Blanco
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Thomas H Dohlman
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Jia Yin
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Reza Dana
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
6
|
Xu Y, Zhang B, Chen Y, Wang X, Li Y, Wu J, Dong H, Wang S. Simvastatin increases circulating endothelial progenitor cells and inhibits the formation of intracranial aneurysms in rats with diet-induced hyperhomocysteinemia. Neurosci Lett 2021; 760:136072. [PMID: 34147541 DOI: 10.1016/j.neulet.2021.136072] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/01/2021] [Accepted: 06/15/2021] [Indexed: 01/21/2023]
Abstract
BACKGROUND AND PURPOSE Endothelial dysfunction triggers early pathological changes in artery, leading to the formation of intracranial aneurysm (ICA). Increase in plasma homocysteine (Hcy) impairs endothelium and endothelial progenitor cells (EPCs) are critical in repairing damaged endothelium. The aim of this study was to assess the impact of simvastatin on ICA formation in rats with hyperhomocysteinemia (HHcy). METHODS ICAs were induced in Male Sprague-Dawley rats after surgical induction in the presence of HHcy induced by a high L-methionine diet with or without oral simvastatin treatment. The size and media thickness of ICAs were evaluated 2 months after aneurysm induction. EPCs and serum vascular endothelial grow factor (VEGF) were measured be flow cytometry and ELISA respectively. Plasma Hcy levels and expression of VEGF, endothelial nitric oxide synthase (eNOS), inducible nitric oxide synthase (iNOS), matrix metalloproteinase-2 (MMP-2), and MMP-9 in aneurysmal walls were examined and correlated with ICA formation. RESULTS HHcy accelerates ICA formation and rats treated with simvastatin exhibited a significant increase in media thickness and a reduction in aneurysmal size. Simvastatin increased levels of circulating EPCs and decreased iNOS, MMP-2, MMP-9 and VEGF mRNA levels, while increased eNOS mRNA in aneurysmal tissue. CONCLUSION In a rat model, HHcy reduces circulating EPCs and accelerates ICA formation. Simvastatin treatment increases circulating EPCs and inhabits the formation of ICA. We have shown a close association among circulating EPCs, biochemical markers related to vascular remodeling and the formation of ICA.
Collapse
Affiliation(s)
- Yong Xu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Bin Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yu Chen
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiu Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yong Li
- Department of Neurosurgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jiangping Wu
- Department of Neurosurgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Hao Dong
- Department of Neurosurgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Shuo Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
7
|
Effect of Natural Cytokine Complex on Metabolism of Smooth Muscle Cells in Myocardial Arteries under Normal Conditions and during Hemodynamic Overload. Bull Exp Biol Med 2020; 168:430-434. [PMID: 32146620 DOI: 10.1007/s10517-020-04725-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Indexed: 10/24/2022]
Abstract
Histoenzymological methods were employed to examine the effects of systemically administered natural cytokine complex including IL-1, IL-2, IL-6, TNFα, MIF, and TGFβ on metabolism of smooth muscle cells in intramural myocardial arteries under physiological conditions and during acute hemodynamic overload of the heart. Natural cytokine complex markedly inhibited metabolism of vascular smooth muscle cells under control conditions and during acute experimental aortal stenosis. In vascular smooth muscle cells, deceleration of tricarboxylic acid cycle, redistribution of the fluxes in glycolytic cascade and its inhibition, down-regulation of oxidation of free fatty acids and their metabolites, and inhibition of the shuttle systems and biosynthetic processes were observed. Inhibition of metabolism in the vascular wall of myocardial arteries correlated with a decrease in their tone and could be partially determined by a decrease in contractile activity of smooth muscle cells. These findings do not exclude the involvement of other factors and mechanisms in down-regulation of metabolism in vascular myocytes in response to increased cytokines levels of in the blood, including their direct effect on biochemical processes in cells.
Collapse
|
8
|
Liu Y, Yuan X, Muñoz N, Logan TM, Ma T. Commitment to Aerobic Glycolysis Sustains Immunosuppression of Human Mesenchymal Stem Cells. Stem Cells Transl Med 2018; 8:93-106. [PMID: 30272389 PMCID: PMC6312448 DOI: 10.1002/sctm.18-0070] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 08/16/2018] [Indexed: 12/13/2022] Open
Abstract
Human mesenchymal stem cells (hMSCs) promote endogenous tissue repair in part by coordinating multiple components of the host immune system in response to environmental stimuli. Recent studies have shown that hMSCs are metabolically heterogeneous and actively reconfigure metabolism to support the biochemical demands of tissue repair. However, how hMSCs regulate their energy metabolism to support their immunomodulatory properties is largely unknown. This study investigates hMSC metabolic reconfiguration during immune activation and provides evidence that the hMSC metabolic state significantly influences their immunomodulatory properties. Specifically, hMSC immune polarization by interferon‐gamma (IFN‐γ) treatment leads to remodeling of hMSC metabolic pathways toward glycolysis, which is required to sustain the secretion of immunosuppressive factors. IFN‐γ exposure also inhibited mitochondrial electron transport activity, and the accumulation of mitochondrial reactive oxygen species plays an important signaling role in this metabolic reconfiguration. The results also show that activation of the Akt/mTOR signaling pathway is required for metabolic reconfiguration during immune polarization and that interruption of these metabolic changes alters the immune response in IFN‐γ licensed hMSCs. The results demonstrate the potential of altering hMSC metabolism to enhance their immunomodulatory properties and therapeutic efficacy in various diseases. Stem Cells Translational Medicine2019;8:93–106
Collapse
Affiliation(s)
- Yijun Liu
- Department of Chemical and Biomedical Engineering, Florida State University, Tallahassee, Florida, USA
| | - Xuegang Yuan
- Department of Chemical and Biomedical Engineering, Florida State University, Tallahassee, Florida, USA
| | - Nathalie Muñoz
- Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida, USA
| | - Timothy M Logan
- Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida, USA.,Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida, USA
| | - Teng Ma
- Department of Chemical and Biomedical Engineering, Florida State University, Tallahassee, Florida, USA.,Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida, USA
| |
Collapse
|
9
|
Tse MCL, Herlea-Pana O, Brobst D, Yang X, Wood J, Hu X, Liu Z, Lee CW, Zaw AM, Chow BKC, Ye K, Chan CB. Tumor Necrosis Factor-α Promotes Phosphoinositide 3-Kinase Enhancer A and AMP-Activated Protein Kinase Interaction to Suppress Lipid Oxidation in Skeletal Muscle. Diabetes 2017; 66:1858-1870. [PMID: 28404596 PMCID: PMC5482076 DOI: 10.2337/db16-0270] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 03/29/2017] [Indexed: 12/12/2022]
Abstract
Tumor necrosis factor-α (TNF-α) is an inflammatory cytokine that plays a central role in obesity-induced insulin resistance. It also controls cellular lipid metabolism, but the underlining mechanism is poorly understood. We report in this study that phosphoinositide 3-kinase enhancer A (PIKE-A) is a novel effector of TNF-α to facilitate its metabolic modulation in the skeletal muscle. Depletion of PIKE-A in C2C12 myotubes diminished the inhibitory activities of TNF-α on mitochondrial respiration and lipid oxidation, whereas PIKE-A overexpression exacerbated these cellular responses. We also found that TNF-α promoted the interaction between PIKE-A and AMP-activated protein kinase (AMPK) to suppress its kinase activity in vitro and in vivo. As a result, animals with PIKE ablation in the skeletal muscle per se display an upregulation of AMPK phosphorylation and a higher preference to use lipid as the energy production substrate under high-fat diet feeding, which mitigates the development of diet-induced hyperlipidemia, ectopic lipid accumulation, and muscle insulin resistance. Hence, our data reveal PIKE-A as a new signaling factor that is important for TNF-α-initiated metabolic changes in skeletal muscle.
Collapse
Affiliation(s)
- Margaret Chui Ling Tse
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, People's Republic of China
| | - Oana Herlea-Pana
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Daniel Brobst
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Xiuying Yang
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
- Drug Screening Center, Institute of Materia Medica, Beijing, People's Republic of China
| | - John Wood
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Xiang Hu
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Zhixue Liu
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Chi Wai Lee
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, People's Republic of China
| | - Aung Moe Zaw
- School of Biological Sciences, The University of Hong Kong, Hong Kong SAR, People's Republic of China
| | - Billy K C Chow
- School of Biological Sciences, The University of Hong Kong, Hong Kong SAR, People's Republic of China
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA
| | - Chi Bun Chan
- School of Biological Sciences, The University of Hong Kong, Hong Kong SAR, People's Republic of China
| |
Collapse
|
10
|
Lone Y, Bhide M, Koiri RK. Amelioratory effect of coenzyme Q10 on potential human carcinogen Microcystin-LR induced toxicity in mice. Food Chem Toxicol 2017; 102:176-185. [PMID: 28219701 DOI: 10.1016/j.fct.2017.02.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 12/28/2016] [Accepted: 02/14/2017] [Indexed: 10/20/2022]
Abstract
Microcystins are a group of cyclic heptapeptide toxins produced by cyanobacteria. More than 100 microcystin analogues have been detected, among which microcystin-LR is the most abundant and toxic variant. Present study was designed to reveal whether potential human carcinogen microcystin-LR could imbalance the glycolytic-oxidative-nitrosative status of heart, kidney and spleen of mice and also to explore the amelioratory effect of coenzyme Q10 on microcystin-LR induced toxicity. Microcystin-LR was administered at a dose of 10 μg/kg bw/day, ip for 14 days in male mice. In microcystin-LR treated mice as compared to control, significant increase in the level of lipid peroxidation, hydrogen peroxide, lactate dehydrogenase, nitric oxide with a concomitant decrease in the level of glutathione was observed, suggesting microcystin-LR induced toxicity via induction of oxidative-nitrosative-glycolytic pathway. Although several studies have evaluated numerous antioxidants but still there is no effective chemoprotectant against microcystin-LR induced toxicity. When microcystin-LR treated mice were co-administered coenzyme Q10 (10 mg/kg bw/day, im) for 14 days, it was observed that coenzyme Q10 ameliorates microcystin-LR induced toxicity via modulation of glycolytic-oxidative-nitrosative stress pathway. Thus, the results suggest that coenzyme Q10 has a potential to be developed as preventive agent against microcystin-LR induced toxicity.
Collapse
Affiliation(s)
- Yaqoob Lone
- Department of Zoology, Dr. Harisingh Gour Central University, Sagar, Madhya Pradesh, 470003, India
| | - Mangla Bhide
- Department of Zoology, Dr. Harisingh Gour Central University, Sagar, Madhya Pradesh, 470003, India
| | - Raj Kumar Koiri
- Department of Zoology, Dr. Harisingh Gour Central University, Sagar, Madhya Pradesh, 470003, India.
| |
Collapse
|
11
|
Sigala F, Savvari P, Liontos M, Sigalas P, Pateras IS, Papalampros A, Basdra EK, Kolettas E, Papavassiliou AG, Gorgoulis VG. Increased expression of bFGF is associated with carotid atherosclerotic plaques instability engaging the NF-κB pathway. J Cell Mol Med 2016; 14:2273-80. [PMID: 20455997 PMCID: PMC3822568 DOI: 10.1111/j.1582-4934.2010.01082.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Unstable atherosclerotic plaques of the carotid arteries are at great risk for the development of ischemic cerebrovascular events. The degradation of the extracellular matrix by matrix metalloproteinases (MMPs) and NO-induced apoptosis of vascular smooth muscle cells (VSMCs) contribute to the vulnerability of the atherosclerotic plaques. Basic fibroblast growth factor (bFGF) through its mitogenic and angiogenic properties has already been implicated in the pathogenesis of atherosclerosis. However, its role in plaque stability remains elusive. To address this issue, a panel of human carotid atherosclerotic plaques was analyzed for bFGF, FGF-receptors-1 and -2 (FGFR-1/-2), inducible nitric oxide synthase (iNOS) and MMP-9 expression. Our data revealed increased expression of bFGF and FGFR-1 in VSMCs of unstable plaques, implying the existence of an autocrine loop, which significantly correlated with high iNOS and MMP-9 levels. These results were recapitulated in vitro by treatment of VSMCs with bFGF. bFGF administration led to up-regulation of both iNOS and MMP-9 that was specifically mediated by nuclear factor-kappaB (NF-kappaB) activation. Collectively, our data demonstrate a novel NF-kappaB-mediated pathway linking bFGF with iNOS and MMP-9 expression that is associated with carotid plaque vulnerability.
Collapse
Affiliation(s)
- Fragiska Sigala
- Molecular Carcinogenesis Group, Laboratory of Histology and Embryology, Medical School, University of Athens, Athens, Greece
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Cortier M, Boina-Ali R, Racoeur C, Paul C, Solary E, Jeannin JF, Bettaieb A. H89 enhances the sensitivity of cancer cells to glyceryl trinitrate through a purinergic receptor-dependent pathway. Oncotarget 2016; 6:6877-86. [PMID: 25762630 PMCID: PMC4466656 DOI: 10.18632/oncotarget.3124] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 01/09/2015] [Indexed: 01/08/2023] Open
Abstract
High doses of the organic nitrate glyceryl trinitrate (GTN), a nitric oxide (NO) donor, are known to trigger apoptosis in human cancer cells. Here, we show that such a cytotoxic effect can be obtained with subtoxic concentrations of GTN when combined with H89, N-[2-(p-bromocinnamylamino)ethyl]-5-isoquinolinesulphonamide.2HCl. This synergistic effect requires the generation of reactive oxygen species (ROS) from H89 and NO from GTN treatment that causes cGMP production and PKG activation. Furthermore, the GTN/H89 synergy was attenuated by inhibition of P2-purinergic receptors with suramin and competition with ATP/UDP. By down-regulating genes with antisense oligonucleotides, P2-purinergic receptors P2X3, P2Y1, and P2Y6 were found to have a role in creating this cytotoxic effect. Thus, H89 likely acts as an ATP mimetic synergizing with GTN to trigger apoptosis in aggressive cancer cells.
Collapse
Affiliation(s)
- Marion Cortier
- EPHE, Tumor Immunology and Immunotherapy Laboratory, Dijon, F-21000, France.,Inserm U866, Dijon, F-21000, France.,EA7269, University of Burgundy, Dijon, F-21000, France
| | - Rahamata Boina-Ali
- EPHE, Tumor Immunology and Immunotherapy Laboratory, Dijon, F-21000, France.,Inserm U866, Dijon, F-21000, France.,EA7269, University of Burgundy, Dijon, F-21000, France
| | - Cindy Racoeur
- EPHE, Tumor Immunology and Immunotherapy Laboratory, Dijon, F-21000, France.,Inserm U866, Dijon, F-21000, France.,EA7269, University of Burgundy, Dijon, F-21000, France
| | - Catherine Paul
- EPHE, Tumor Immunology and Immunotherapy Laboratory, Dijon, F-21000, France.,Inserm U866, Dijon, F-21000, France.,EA7269, University of Burgundy, Dijon, F-21000, France
| | - Eric Solary
- Inserm U866, Dijon, F-21000, France.,Inserm UMR1009, Gustave Roussy Institute, Villejuif F-94805, France.,University Paris-Sud, Faculty of Medicine, Le Kremlin-Bicêtre, F-94800, France
| | - Jean-François Jeannin
- EPHE, Tumor Immunology and Immunotherapy Laboratory, Dijon, F-21000, France.,Inserm U866, Dijon, F-21000, France.,EA7269, University of Burgundy, Dijon, F-21000, France
| | - Ali Bettaieb
- EPHE, Tumor Immunology and Immunotherapy Laboratory, Dijon, F-21000, France.,Inserm U866, Dijon, F-21000, France.,EA7269, University of Burgundy, Dijon, F-21000, France
| |
Collapse
|
13
|
IL-4 Protects the Mitochondria Against TNFα and IFNγ Induced Insult During Clearance of Infection with Citrobacter rodentium and Escherichia coli. Sci Rep 2015; 5:15434. [PMID: 26481427 PMCID: PMC4613366 DOI: 10.1038/srep15434] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 09/24/2015] [Indexed: 11/08/2022] Open
Abstract
Citrobacter rodentium is a murine pathogen that serves as a model for enteropathogenic Escherichia coli. C. rodentium infection reduced the quantity and activity of mitochondrial respiratory complexes I and IV, as well as phosphorylation capacity, mitochondrial transmembrane potential and ATP generation at day 10, 14 and 19 post infection. Cytokine mRNA quantification showed increased levels of IFNγ, TNFα, IL-4, IL-6, and IL-12 during infection. The effects of adding these cytokines, C. rodentium and E. coli were hence elucidated using an in vitro colonic mucosa. Both infection and TNFα, individually and combined with IFNγ, decreased complex I and IV enzyme levels and mitochondrial function. However, IL-4 reversed these effects, and IL-6 protected against loss of complex IV. Both in vivo and in vitro, the dysfunction appeared caused by nitric oxide-generation, and was alleviated by an antioxidant targeting mitochondria. IFNγ −/− mice, containing a similar pathogen burden but higher IL-4 and IL-6, displayed no loss of any of the four complexes. Thus, the cytokine environment appears to be a more important determinant of mitochondrial function than direct actions of the pathogen. As IFNγ and TNFα levels increase during clearance of infection, the concomitant increase in IL-4 and IL-6 protects mitochondrial function.
Collapse
|
14
|
Yokomizo CH, Pessoto FS, Prieto T, Cunha RLOR, Nantes IL. Effects of Trichlorotelluro-dypnones on Mitochondrial Bioenergetics and Their Relationship to the Reactivity with Protein Thiols. Chem Res Toxicol 2015; 28:1167-75. [DOI: 10.1021/tx5005166] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- César H. Yokomizo
- Departamento
de Biologia Molecular, Universidade Federal de São Paulo, R. Botucatu, 740, São Paulo, SP Brazil
| | - Felipe S. Pessoto
- Centro
de Ciências Naturais e Humanas, Universidade Federal do ABC, Av. dos
Estados, 5001, Santo
André, SP Brazil
| | - Tatiana Prieto
- Centro
de Ciências Naturais e Humanas, Universidade Federal do ABC, Av. dos
Estados, 5001, Santo
André, SP Brazil
| | - Rodrigo L. O. R. Cunha
- Centro
de Ciências Naturais e Humanas, Universidade Federal do ABC, Av. dos
Estados, 5001, Santo
André, SP Brazil
| | - Iseli L. Nantes
- Centro
de Ciências Naturais e Humanas, Universidade Federal do ABC, Av. dos
Estados, 5001, Santo
André, SP Brazil
| |
Collapse
|
15
|
Lengert N, Drossel B. In silico analysis of exercise intolerance in myalgic encephalomyelitis/chronic fatigue syndrome. Biophys Chem 2015; 202:21-31. [PMID: 25899994 DOI: 10.1016/j.bpc.2015.03.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 03/26/2015] [Accepted: 03/28/2015] [Indexed: 11/16/2022]
Abstract
Post-exertional malaise is commonly observed in patients with myalgic encephalomyelitis/chronic fatigue syndrome, but its mechanism is not yet well understood. A reduced capacity for mitochondrial ATP synthesis is associated with the pathogenesis of CFS and is suspected to be a major contribution to exercise intolerance in CFS patients. To demonstrate the connection between a reduced mitochondrial capacity and exercise intolerance, we present a model which simulates metabolite dynamics in skeletal muscles during exercise and recovery. CFS simulations exhibit critically low levels of ATP, where an increased rate of cell death would be expected. To stabilize the energy supply at low ATP concentrations the total adenine nucleotide pool is reduced substantially causing a prolonged recovery time even without consideration of other factors, such as immunological dysregulations and oxidative stress. Repeated exercises worsen this situation considerably. Furthermore, CFS simulations exhibited an increased acidosis and lactate accumulation consistent with experimental observations.
Collapse
Affiliation(s)
- Nicor Lengert
- Institute for Condensed Matter Physics, Technische Universität Darmstadt, Hochschulstr. 6, 64289 Darmstadt, Germany.
| | - Barbara Drossel
- Institute for Condensed Matter Physics, Technische Universität Darmstadt, Hochschulstr. 6, 64289 Darmstadt, Germany
| |
Collapse
|
16
|
Hansson GK. A journey in science: medical scientist in translation. Mol Med 2014; 20:381-9. [PMID: 25356751 DOI: 10.2119/molmed.2014.00092] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 05/01/2014] [Indexed: 11/06/2022] Open
Abstract
Real innovations in medicine and science are historic and singular; the stories behind each occurrence are precious. At Molecular Medicine we have established the Anthony Cerami Award in Translational Medicine to document and preserve these histories. The monographs recount the seminal events as told in the voice of the original investigators who provided the crucial early insight. These essays capture the essence of discovery, chronicling the birth of ideas that created new fields of research; and launched trajectories that persisted and ultimately influenced how disease is prevented, diagnosed and treated. In this volume, the Cerami Award Monograph is by Göran K Hansson, MD, PhD, Karolinska Institute. A visionary in the field of cardiovascular research, this is the story of Dr. Hansson's scientific journey.
Collapse
Affiliation(s)
- Göran K Hansson
- Karolinska Institute, Department of Medicine Solna and Center for Molecular Medicine at Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
17
|
Therapy with immunoglobulin in patients with acute myocarditis and cardiomyopathy: analysis of leukocyte balance. Heart Vessels 2013; 29:336-42. [DOI: 10.1007/s00380-013-0368-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 05/10/2013] [Indexed: 10/26/2022]
|
18
|
Predictors of mortality in patients with stress-induced cardiomyopathy developed during critical care. J Crit Care 2013; 28:618-24. [PMID: 23683567 DOI: 10.1016/j.jcrc.2013.03.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 03/06/2013] [Accepted: 03/19/2013] [Indexed: 12/12/2022]
Abstract
PURPOSE The aims of this study were to define predictors of in-hospital mortality and to explore the implication of Acute Physiology and Chronic Health Evaluation (APACHE) II score in patients with stress-induced cardiomyopathy (SCM) developed during critical care. MATERIALS AND METHODS All patients admitted to intensive care unit and underwent transthoracic echocardiography (TTE) were consecutively enrolled from January 2008 to May 2011. Clinical, demographic and laboratory data, APACHE II score, and transthoracic echocardiography finding were analyzed using a logistic regression model to investigate predictors of in-hospital mortality. RESULTS A total of 71 patients (60 ± 18 years, 37% male) were included in the final analysis. In univariate and multivariate logistic regression analyses, underlying malignancies, male sex, age less than 65 years, and APACHE II score higher than 15 remained independent risk factors for in-hospital mortality of SCM. The area under the receiver operating characteristic curve for APACHE II was 0.745 (95% confidence interval, 0.630-0.861; P = .001), and an APACHE II score of 15 (sensitivity 73%, specificity 68%) was the optimal cutoff value in predicting in-hospital mortality of SCM during critical care. CONCLUSION The in-hospital mortality in patients with SCM that developed during critical care was associated with underlying malignancy, male sex, old age, and APACHE II score.
Collapse
|
19
|
Tonelli AR, Haserodt S, Aytekin M, Dweik RA. Nitric oxide deficiency in pulmonary hypertension: Pathobiology and implications for therapy. Pulm Circ 2013; 3:20-30. [PMID: 23662172 PMCID: PMC3641730 DOI: 10.4103/2045-8932.109911] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Nitric oxide (NO) is a diffusible gas with diverse roles in human physiology and disease. Significant progress in the understanding of its biological effects has taken place in recent years. This has led to a better understanding of the pathobiology of pulmonary hypertension (PH) and the development of new therapies. This article provides an overview of the NO physiology and its role in the pathobiology of lung diseases, particularly PH. We also discuss current and emerging specific treatments that target NO signaling pathways in PH.
Collapse
Affiliation(s)
- Adriano R Tonelli
- Department of Pulmonary, Allergy and Critical Care Medicine, Respiratory Institute, Cleveland, Ohio, USA
| | | | | | | |
Collapse
|
20
|
Karagiannis GS, Weile J, Bader GD, Minta J. Integrative pathway dissection of molecular mechanisms of moxLDL-induced vascular smooth muscle phenotype transformation. BMC Cardiovasc Disord 2013; 13:4. [PMID: 23324130 PMCID: PMC3556327 DOI: 10.1186/1471-2261-13-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 12/29/2012] [Indexed: 01/08/2023] Open
Abstract
Background Atherosclerosis (AT) is a chronic inflammatory disease characterized by the accumulation of inflammatory cells, lipoproteins and fibrous tissue in the walls of arteries. AT is the primary cause of heart attacks and stroke and is the leading cause of death in Western countries. To date, the pathogenesis of AT is not well-defined. Studies have shown that the dedifferentiation of contractile and quiescent vascular smooth muscle cells (SMC) to the proliferative, migratory and synthetic phenotype in the intima is pivotal for the onset and progression of AT. To further delineate the mechanisms underlying the pathogenesis of AT, we analyzed the early molecular pathways and networks involved in the SMC phenotype transformation. Methods Quiescent human coronary artery SMCs were treated with minimally-oxidized LDL (moxLDL), for 3 hours and 21 hours, respectively. Transcriptomic data was generated for both time-points using microarrays and was subjected to pathway analysis using Gene Set Enrichment Analysis, GeneMANIA and Ingenuity software tools. Gene expression heat maps and pathways enriched in differentially expressed genes were compared to identify functional biological themes to elucidate early and late molecular mechanisms of moxLDL-induced SMC dedifferentiation. Results Differentially expressed genes were found to be enriched in cholesterol biosynthesis, inflammatory cytokines, chemokines, growth factors, cell cycle control and myogenic contraction themes. These pathways are consistent with inflammatory responses, cell proliferation, migration and ECM production, which are characteristic of SMC dedifferentiation. Furthermore, up-regulation of cholesterol synthesis and dysregulation of cholesterol metabolism was observed in moxLDL-induced SMC. These observations are consistent with the accumulation of cholesterol and oxidized cholesterol esters, which induce proinflammatory reactions during atherogenesis. Our data implicate for the first time IL12, IFN-α, HGF, CSF3, and VEGF signaling in SMC phenotype transformation. GPCR signaling, HBP1 (repressor of cyclin D1 and CDKN1B), and ID2 and ZEB1 transcriptional regulators were also found to have important roles in SMC dedifferentiation. Several microRNAs were observed to regulate the SMC phenotype transformation via an interaction with IFN-γ pathway. Also, several “nexus” genes in complex networks, including components of the multi-subunit enzyme complex involved in the terminal stages of cholesterol synthesis, microRNAs (miR-203, miR-511, miR-590-3p, miR-346*/miR- 1207-5p/miR-4763-3p), GPCR proteins (GPR1, GPR64, GPRC5A, GPR171, GPR176, GPR32, GPR25, GPR124) and signal transduction pathways, were found to be regulated. Conclusions The systems biology analysis of the in vitro model of moxLDL-induced VSMC phenotype transformation was associated with the regulation of several genes not previously implicated in SMC phenotype transformation. The identification of these potential candidate genes enable hypothesis generation and in vivo functional experimentation (such as gain and loss-of-function studies) to establish causality with the process of SMC phenotype transformation and atherogenesis.
Collapse
Affiliation(s)
- George S Karagiannis
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, and Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON, M5S 1A8, Canada
| | | | | | | |
Collapse
|
21
|
Geng YJ, Jonasson L. Linking immunity to atherosclerosis: implications for vascular pharmacology--a tribute to Göran K. Hansson. Vascul Pharmacol 2012; 56:29-33. [PMID: 22120836 PMCID: PMC3268894 DOI: 10.1016/j.vph.2011.11.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Revised: 11/10/2011] [Accepted: 11/11/2011] [Indexed: 12/26/2022]
Abstract
For the past decade, we have deepened our understanding of the pathogenesis of atherosclerosis, a chronic arterial disease that causes cardiac and cerebral infarction and peripheral vascular disorders. Because of this extended understanding, more effective strategies for prevention and treatment of this disease are emerging. One of the fundamental mechanisms that lead to progress or regression in atherosclerosis, thus influencing its life-threatening complications, occurs through functional changes in vascular immunity and inflammation. This review briefly summarizes the discoveries in basic and translational sciences in this area and recent advances in clinical medicine against atherosclerotic vascular diseases.
Collapse
Affiliation(s)
- Yong-Jian Geng
- Center of Cardiovascular Biology and Atherosclerosis Research, University of Texas Medical School at Houston, TX 77030, USA
| | - Lena Jonasson
- Division of Cardiovascular Medicine, Department of Medical and Health Sciences, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
22
|
Widlansky ME, Gutterman DD. Regulation of endothelial function by mitochondrial reactive oxygen species. Antioxid Redox Signal 2011; 15:1517-30. [PMID: 21194353 PMCID: PMC3151425 DOI: 10.1089/ars.2010.3642] [Citation(s) in RCA: 143] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2010] [Revised: 12/07/2010] [Accepted: 01/01/2011] [Indexed: 12/19/2022]
Abstract
Mitochondria are well known for their central roles in ATP production, calcium homeostasis, and heme and steroid biosynthesis. However, mitochondrial reactive oxygen species (ROS), including superoxide and hydrogen peroxide, once thought to be toxic byproducts of mitochondrial physiologic activities, have recently been recognized as important cell-signaling molecules in the vascular endothelium, where their production, conversion, and destruction are highly regulated. Mitochondrial reactive oxygen species appear to regulate important vascular homeostatic functions under basal conditions in a variety of vascular beds, where, in particular, they contribute to endothelium-dependent vasodilation. On exposure to cardiovascular risk factors, endothelial mitochondria produce excessive ROS in concert with other cellular ROS sources. Mitochondrial ROS, in this setting, act as important signaling molecules activating prothrombotic and proinflammatory pathways in the vascular endothelium, a process that initially manifests itself as endothelial dysfunction and, if persistent, may lead to the development of atherosclerotic plaques. This review concentrates on emerging appreciation of the importance of mitochondrial ROS as cell-signaling molecules in the vascular endothelium under both physiologic and pathophysiologic conditions. Future potential avenues of research in this field also are discussed.
Collapse
Affiliation(s)
- Michael E Widlansky
- Department of Medicine, Cardiovascular Medicine Division and Department of Pharmacology, Medical College of Wisconsin , Milwaukee, Wisconsin
| | | |
Collapse
|
23
|
Chen J, Chen CL, Alevriadou BR, Zweier JL, Chen YR. Excess no predisposes mitochondrial succinate-cytochrome c reductase to produce hydroxyl radical. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2011; 1807:491-502. [PMID: 21406178 DOI: 10.1016/j.bbabio.2011.03.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2010] [Revised: 03/07/2011] [Accepted: 03/08/2011] [Indexed: 11/30/2022]
Abstract
Mitochondria-derived oxygen-free radical(s) are important mediators of oxidative cellular injury. It is widely hypothesized that excess NO enhances O(2)(•-) generated by mitochondria under certain pathological conditions. In the mitochondrial electron transport chain, succinate-cytochrome c reductase (SCR) catalyzes the electron transfer reaction from succinate to cytochrome c. To gain the insights into the molecular mechanism of how NO overproduction may mediate the oxygen-free radical generation by SCR, we employed isolated SCR, cardiac myoblast H9c2, and endothelial cells to study the interaction of NO with SCR in vitro and ex vivo. Under the conditions of enzyme turnover in the presence of NO donor (DEANO), SCR gained pro-oxidant function for generating hydroxyl radical as detected by EPR spin trapping using DEPMPO. The EPR signal associated with DEPMPO/(•)OH adduct was nearly completely abolished in the presence of catalase or an iron chelator and partially inhibited by SOD, suggesting the involvement of the iron-H(2)O(2)-dependent Fenton reaction or O(2)(•-)-dependent Haber-Weiss mechanism. Direct EPR measurement of SCR at 77K indicated the formation of a nonheme iron-NO complex, implying that electron leakage to molecular oxygen was enhanced at the FAD cofactor, and that excess NO predisposed SCR to produce (•)OH. In H9c2 cells, SCR-dependent oxygen-free radical generation was stimulated by NO released from DEANO or produced by the cells following exposure to hypoxia/reoxygenation. With shear exposure that led to overproduction of NO by the endothelium, SCR-mediated oxygen-free radical production was also detected in cultured vascular endothelial cells.
Collapse
Affiliation(s)
- Jingfeng Chen
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | |
Collapse
|
24
|
Xu Y, Tian Y, Wei HJ, Dong JF, Zhang JN. Methionine diet-induced hyperhomocysteinemia accelerates cerebral aneurysm formation in rats. Neurosci Lett 2011; 494:139-44. [PMID: 21382440 DOI: 10.1016/j.neulet.2011.02.076] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2010] [Revised: 02/05/2011] [Accepted: 02/27/2011] [Indexed: 11/25/2022]
Abstract
BACKGROUND AND PURPOSE The pathophysiology of cerebral aneurysms (CA) is linked to chronic inflammation. Endothelial damage is one of the first changes in CA walls resulted from inflammation. It has been shown that increase in plasma homocysteine (Hcy) impairs vascular endothelium and correlates with the development of atherosclerosis. However, the effect of hyperhomocysteinemia (HHcy) on the formation of cerebral aneurysm remains unknown. METHODS Male Sprague-Dawley rats examined for developing cerebral aneurysms after surgical induction in the presence and absence of hypercysteinemia induced by a high L-methionine diet (1 g/kg/d). Aneurysms developed at the anterior cerebral-olfactory artery bifurcation were classified as 4 stages from no abnormality to saccular aneurysm. Plasma homocysteine levels and expression of vascular endothelial growth factor (VEGF), endothelial nitric oxide synthase (eNOS), inducible nitric oxide synthase (iNOS), matrix metalloproteinase-2 (MMP-2), and MMP-9 in aneurysmal walls was examined and correlated with CA formation 3 months after surgery. RESULTS Methionine diet significantly increased plasma homocysteine levels, accelerates CA formation after ligation of the left common carotid artery. Expression of VEGF, iNOS, MMP-2, and MMP-9 in aneurysmal walls was also increased by methionine treatment. CONCLUSION Hyperhomocysteinemia accelerates cerebral aneurysm formation, potentially through differential effects on expression of molecules critical for vascular wall modeling in a rat model.
Collapse
Affiliation(s)
- Yong Xu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300052 PR China
| | | | | | | | | |
Collapse
|
25
|
Barbiro-Michaely E, Mendelman A, Mayevsky A. The evaluation of nitric oxide involvement in Metrazol induced status epilepticus using multiparametric monitoring. Brain Res 2011; 1377:50-9. [DOI: 10.1016/j.brainres.2011.01.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Revised: 12/30/2010] [Accepted: 01/02/2011] [Indexed: 11/26/2022]
|
26
|
Sprague AH, Khalil RA. Inflammatory cytokines in vascular dysfunction and vascular disease. Biochem Pharmacol 2009; 78:539-52. [PMID: 19413999 PMCID: PMC2730638 DOI: 10.1016/j.bcp.2009.04.029] [Citation(s) in RCA: 958] [Impact Index Per Article: 59.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2009] [Revised: 04/23/2009] [Accepted: 04/27/2009] [Indexed: 12/12/2022]
Abstract
The vascular inflammatory response involves complex interaction between inflammatory cells (neutrophils, lymphocytes, monocytes, macrophages), endothelial cells (ECs), vascular smooth muscle cells (VSMCs), and extracellular matrix (ECM). Vascular injury is associated with increased expression of adhesion molecules by ECs and recruitment of inflammatory cells, growth factors, and cytokines, with consequent effects on ECs, VSMCs and ECM. Cytokines include tumor necrosis factors, interleukins, lymphokines, monokines, interferons, colony stimulating factors, and transforming growth factors. Cytokines are produced by macrophages, T-cells and monocytes, as well as platelets, ECs and VSMCs. Circulating cytokines interact with specific receptors on various cell types and activate JAK-STAT, NF-kappaB, and Smad signaling pathways leading to an inflammatory response involving cell adhesion, permeability and apoptosis. Cytokines also interact with mitochondria to increase the production of reactive oxygen species. Cytokine-induced activation of these pathways in ECs modifies the production/activity of vasodilatory mediators such as nitric oxide, prostacyclin, endothelium-derived hyperpolarizing factor, and bradykinin, as well as vasoconstrictive mediators such as endothelin and angiotensin II. Cytokines interact with VSMCs to activate Ca(2+), protein kinase C, Rho-kinase, and MAPK pathways, which promote cell growth and migration, and VSM reactivity. Cytokines also interact with integrins and matrix metalloproteinases (MMPs) and modify ECM composition. Persistent increases in cytokines are associated with vascular dysfunction and vascular disease such as atherosclerosis, abdominal aortic aneurysm, varicose veins and hypertension. Genetic and pharmacological tools to decrease the production of cytokines or to diminish their effects using cytokine antagonists could provide new approaches in the management of inflammatory vascular disease.
Collapse
Affiliation(s)
- Alexander H Sprague
- Division of Vascular Surgery and Endovascular Therapy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
27
|
Hall CN, Garthwaite J. What is the real physiological NO concentration in vivo? Nitric Oxide 2009; 21:92-103. [PMID: 19602444 PMCID: PMC2779337 DOI: 10.1016/j.niox.2009.07.002] [Citation(s) in RCA: 268] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2009] [Accepted: 07/06/2009] [Indexed: 01/10/2023]
Abstract
Clarity about the nitric oxide (NO) concentrations existing physiologically is essential for developing a quantitative understanding of NO signalling, for performing experiments with NO that emulate reality, and for knowing whether or not NO concentrations become abnormal in disease states. A decade ago, a value of about 1 μM seemed reasonable based on early electrode measurements and a provisional estimate of the potency of NO for its guanylyl cyclase-coupled receptors, which mediate physiological NO signal transduction. Since then, numerous efforts to measure NO concentrations directly using electrodes in cells and tissues have yielded an irreconcilably large spread of values. In compensation, data from several alternative approaches have now converged to provide a more coherent picture. These approaches include the quantitative analysis of NO-activated guanylyl cyclase, computer modelling based on the type, activity and amount of NO synthase enzyme contained in cells, the use of novel biosensors to monitor NO release from single endothelial cells and neurones, and the use of guanylyl cyclase as an endogenous NO biosensor in tissue subjected to a variety of challenges. All these independent lines of evidence suggest the physiological NO concentration range to be 100 pM (or below) up to ∼5 nM, orders of magnitude lower than was once thought.
Collapse
Affiliation(s)
- Catherine N Hall
- Department of Neuroscience, Physiology and Pharmacology, University College London, UK
| | | |
Collapse
|
28
|
Mitochondrial Genetics and Sepsis. Intensive Care Med 2009. [DOI: 10.1007/978-0-387-92278-2_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
29
|
Wollin M, Abele S, Bruns H, Weyand M, Kalden JR, Ensminger SM, Spriewald BM. Inhibition of TNF-alpha reduces transplant arteriosclerosis in a murine aortic transplant model. Transpl Int 2008; 22:342-9. [PMID: 19055619 DOI: 10.1111/j.1432-2277.2008.00802.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Experimental and clinical data provide evidence that TNF-alpha contributes to acute and chronic allograft rejection. In this study, we explored the effect of TNF-alpha blockade using the chimeric monoclonal antibody infliximab on the development of transplant arterisoclerosis in a fully mismatched aortic allograft model. Post-transplant treatment of CBA (H2(k)) recipients with 250 mug infliximab (cumulative dose 1.25 mg) reduced luminal occlusion of C57Bl/6 (H2(b)) aortic grafts on day 30 from 77 +/- 5% in untreated controls to 52 +/- 6%. Increasing the dose of anti-TNF-alpha antibody had no further beneficial effect. Treatment with human control immunoglobulin had no effect on intima proliferation. Under TNF-alpha blockade, ICAM-1 and PDGF mRNA expression within the grafts was strongly reduced, whereas iNOS expression was enhanced. The data show that TNF-alpha blockade using infliximab can reduce the development of transplant arteriosclerosis in fully mismatched murine aortic grafts.
Collapse
Affiliation(s)
- Martina Wollin
- Department for Internal Medicine 3 and Institute for Clinical Immunology, University Erlangen-Nürnberg, Erlangen, Germany
| | | | | | | | | | | | | |
Collapse
|
30
|
Howes LG, Brillante DG. Expert opinion on tilarginine in the treatment of shock. Expert Opin Investig Drugs 2008; 17:1573-80. [PMID: 18808317 DOI: 10.1517/13543784.17.10.1573] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Tilarginine is L-N-monomethyl arginine (L-NMMA) or N(G)-monomethyl-L-arginine HCL, a non-selective inhibitor of nitric oxide synthase (NOS), which has been studied in the treatment of septic shock and cardiogenic shock complicating myocardial infarction. Despite strong evidence that excessive nitric oxide (NO) production plays a pivotal role in the pathogenesis of septic shock and may contribute to the pathogenesis of cardiogenic shock complicating myocardial infarction, outcome studies in these two disorders have proved disappointing. L-NMMA therapy was associated with an excess mortality, particularly at doses > 5 mg/(kg h), in septic shock whereas the effects of a lower dose (1 mg/(kg h)) in cardiogenic shock complicating myocardial infarction were neutral. The excess mortality in patients with septic shock was almost certainly the result of unfavourable haemodynamic changes induced by L-NMMA (decreased cardiac output, increased pulmonary vascular resistance and reduced tissue oxygen delivery) whereas the lack of benefit in patients with cardiogenic shock complicating myocardial infarction may have been because the dose of L-NMMA was too low. Further studies of L-NMMA at doses < 5 mg/(kg h) in conjunction with inotrope support may produce more beneficial results. Conversely, the use of a selective inducible NOS inhibitor to reduce the pathological effects of excessive NO production although leaving the beneficial effects of vascular NO production by endothelial NOS unaltered may prove to be of value.
Collapse
Affiliation(s)
- Laurence Guy Howes
- Griffith University and Bond University Medical Schools, Gold Coast Hospital, Department of Pharmacology and Therapeutics, Nerang St, Southport, Queensland 4215, Australia.
| | | |
Collapse
|
31
|
Loppnow H, Werdan K, Buerke M. Vascular cells contribute to atherosclerosis by cytokine- and innate-immunity-related inflammatory mechanisms. Innate Immun 2008; 14:63-87. [PMID: 18713724 DOI: 10.1177/1753425908091246] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular diseases are the human diseases with the highest death rate and atherosclerosis is one of the major underlying causes of cardiovascular diseases. Inflammatory and innate immune mechanisms, employing monocytes, innate receptors, innate cytokines, or chemokines are suggested to be involved in atherogenesis. Among the inflammatory pathways the cytokines are central players. Plasma levels of cytokines and related proteins, such as CRP, have been investigated in cardiovascular patients, tissue mRNA expression was analyzed and correlations to vascular diseases established. Consistent with these findings the generation of cytokine-deficient animals has provided direct evidence for a role of cytokines in atherosclerosis. In vitro cell culture experiments further support the suggestion that cytokines and other innate mechanisms contribute to atherogenesis. Among the initiation pathways of atherogenesis are innate mechanisms, such as toll-like-receptors (TLRs), including the endotoxin receptor TLR4. On the other hand, innate cytokines, such as IL-1 or TNF, or even autoimmune triggers may activate the cells. Cytokines potently activate multiple functions relevant to maintain or spoil homeostasis within the vessel wall. Vascular cells, not least smooth muscle cells, can actively contribute to the inflammatory cytokine-dependent network in the blood vessel wall by: (i) production of cytokines; (ii) response to these potent cell activators; and (iii) cytokine-mediated interaction with invading cells, such as monocytes, T-cells, or mast cells. Activation of these pathways results in accumulation of cells and increased LDL- and ECM-deposition which may serve as an 'immunovascular memory' resulting in an ever-growing response to subsequent invasions. Thus, vascular cells may potently contribute to the inflammatory pathways involved in development and acceleration of atherosclerosis.
Collapse
Affiliation(s)
- Harald Loppnow
- Martin-Luther-Universität Halle-Wittenberg, Universitätsklinik und Poliklinik für Innere Medizin , Halle (Saale), Germany.
| | | | | |
Collapse
|
32
|
Hansson GK. Atherosclerosis--an immune disease: The Anitschkov Lecture 2007. Atherosclerosis 2008; 202:2-10. [PMID: 18951547 DOI: 10.1016/j.atherosclerosis.2008.08.039] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2008] [Revised: 08/13/2008] [Accepted: 08/21/2008] [Indexed: 01/09/2023]
Abstract
Atherosclerosis is an inflammatory disease. This article reviews the emergence of this concept from studies of patients and their lesions, experimental animal models, and epidemiological cohorts. Immunohistochemical studies identified immune cells and mediators and provided evidence for inflammatory activation in the atherosclerotic lesion. In parallel, cell culture studies demonstrated the capacity of vascular cells to interact with immune cells. Subsequent studies of clinical and epidemiological materials have identified inflammatory markers and immunoregulatory genes as contributors of risk for myocardial infarction and stroke. Finally, experiments using gene-targeted mice have provided mechanistic understanding of the disease process. It is now thought that the atherosclerotic process is initiated when low-density lipoproteins accumulate in the intima, activate the endothelium, and promote recruitment of monocytes and T cells. Monocytes differentiate into macrophages, internalize modified lipoproteins, and end up as foam cells. T cells in lesions recognize local antigens and mount T helper-1 responses that contribute to local inflammation and plaque growth. This atherogenic pathway is counterbalanced by anti-inflammatory signals provided by regulatory immunity. Intensified inflammatory activation may lead to local proteolysis, plaque rupture, thrombus formation, ischemia and infarction. Novel therapeutic opportunities may emerge from understanding the role of inflammation in atherosclerosis.
Collapse
Affiliation(s)
- Göran K Hansson
- Center for Molecular Medicine and Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
33
|
Hall CN, Attwell D. Assessing the physiological concentration and targets of nitric oxide in brain tissue. J Physiol 2008; 586:3597-615. [PMID: 18535091 DOI: 10.1113/jphysiol.2008.154724] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Low nanomolar concentrations of nitric oxide activate guanylyl cyclase to produce cGMP, which has diverse physiological effects. Higher concentrations inhibit mitochondrial respiration at cytochrome c oxidase and this has been proposed to be important physiologically, increasing oxygen permeation into tissue (by reducing the oxygen use of cells near blood vessels), activating AMP kinase, and regulating the relationship between cerebral blood flow and oxygen use. It is unclear, however, whether nitric oxide can accumulate physiologically to concentrations at which inhibition of respiration occurs. In rat cerebellar slices, we activated nitric oxide production from each isoform of nitric oxide synthase. Only activation of inducible nitric oxide synthase, which is expressed pathologically, caused any significant inhibition of respiration. Modelling oxygen and nitric oxide concentrations predicted that, in vivo, physiological nitric oxide levels are too low to affect respiration. Even pathologically, the nitric oxide concentration may only rise to 2.5 nm, producing a 1.5% inhibition of respiration. Thus, under physiological conditions, nitric oxide signals do not inhibit respiration but are well-tuned to the dynamic range of guanylyl cyclase activation.
Collapse
Affiliation(s)
- Catherine N Hall
- Department of Physiology, University College London, Gower St, London WC1E 6BT, UK.
| | | |
Collapse
|
34
|
Morton J, Coles B, Wright K, Gallimore A, Morrow JD, Terry ES, Anning PB, Morgan BP, Dioszeghy V, Kühn H, Chaitidis P, Hobbs AJ, Jones SA, O'Donnell VB. Circulating neutrophils maintain physiological blood pressure by suppressing bacteria and IFNgamma-dependent iNOS expression in the vasculature of healthy mice. Blood 2008; 111:5187-94. [PMID: 18281503 PMCID: PMC2602588 DOI: 10.1182/blood-2007-10-117283] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2007] [Accepted: 02/12/2008] [Indexed: 12/29/2022] Open
Abstract
Whether leukocytes exert an influence on vascular function in vivo is not known. Here, genetic and pharmacologic approaches show that the absence of neutrophils leads to acute blood pressure dysregulation. Following neutrophil depletion, systolic blood pressure falls significantly over 3 days (88.0 +/- 3.5 vs 104.0 +/- 2.8 mm Hg, day 3 vs day 0, mean +/- SEM, P < .001), and aortic rings from neutropenic mice do not constrict properly. The constriction defect is corrected using l-nitroarginine-methyl ester (L-NAME) or the specific inducible nitric oxide synthase (iNOS) inhibitor 1400W, while acetylcholine relaxation is normal. iNOS- or IFNgamma-deficient mice are protected from neutropenia-induced hypotension, indicating that iNOS-derived nitric oxide (NO) is responsible and that its induction involves IFNgamma. Oral enrofloxacin partially inhibited hypotension, implicating bacterial products. Roles for cyclooxygenase, complement C5, or endotoxin were excluded, although urinary prostacyclin metabolites were elevated. Neutrophil depletion required complement opsinization, with no evidence for intravascular degranulation. In summary, circulating neutrophils contribute to maintaining physiological tone in the vasculature, at least in part through suppressing early proinflammatory effects of infection. The speed with which hypotension developed provides insight into early changes that occur in the absence of neutrophils and illustrates the importance of constant surveillance of mucosal sites by granulocytes in healthy mice.
Collapse
Affiliation(s)
- Jonathan Morton
- Department of Medical Biochemistry & Immunology, Cardiff University, Cardiff, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Chan JYH, Cheng HL, Chou JLJ, Li FCH, Dai KY, Chan SHH, Chang AYW. Heat Shock Protein 60 or 70 Activates Nitric-oxide Synthase (NOS) I- and Inhibits NOS II-associated Signaling and Depresses the Mitochondrial Apoptotic Cascade during Brain Stem Death. J Biol Chem 2007; 282:4585-4600. [PMID: 17150954 DOI: 10.1074/jbc.m603394200] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The cellular and molecular basis of brain stem death remains an enigma. As the origin of a "life-and-death" signal that reflects the progression toward brain stem death, the rostral ventrolateral medulla (RVLM) is a suitable neural substrate for mechanistic delineation of this phenomenon. Here, we evaluated the hypothesis that heat shock proteins (HSPs) play a neuroprotective role in the RVLM during brain stem death and delineated the underlying mechanisms, using a clinically relevant animal model that employed the organophosphate pesticide mevinphos (Mev) as the experimental insult. In Sprague-Dawley rats, proteomic, Western blot, and real-time PCR analyses demonstrated that Mev induced de novo synthesis of HSP60 or HSP70 in the RVLM without affecting HSP90 level. Loss-of-function manipulations of HSP60 or HSP70 in the RVLM using anti-serum or antisense oligonucleotide potentiated Mev-elicited cardiovascular depression alongside reduced nitric-oxide synthase (NOS) I/protein kinase G signaling, enhanced NOS II/peroxynitrite cascade, intensified nucleosomal DNA fragmentation, elevated cytoplasmic histone-associated DNA fragments or activated caspase-3, and augmented the cytochrome c/caspase-3 cascade of apoptotic signaling in the RVLM. Co-immunoprecipitation experiments further revealed a progressive increase in the complex formed between HSP60 and mitochondrial or cytosolic Bax or mitochondrial Bcl-2 during Mev intoxication, alongside a dissociation of the cytosolic HSP60-Bcl-2 complex. We conclude that HSP60 and HSP70 confer neuroprotection against Mev intoxication by ameliorating cardiovascular depression via an anti-apoptotic action in the RVLM. The possible underlying intracellular processes include enhancing NOS I/protein kinase G signaling and inhibiting the NOS II/peroxynitrite cascade. In addition, HSP60 exerts its effects against apoptosis by blunting Mev-induced activation of the Bax/cytochrome c/caspase-3 cascade.
Collapse
Affiliation(s)
- Julie Y H Chan
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 81346
| | - Hsiao-Lei Cheng
- Center for Neuroscience, National Sun Yat-sen University, Kaohsiung 80424, and the
| | - Jimmy L J Chou
- Center for Neuroscience, National Sun Yat-sen University, Kaohsiung 80424, and the
| | - Faith C H Li
- Center for Neuroscience, National Sun Yat-sen University, Kaohsiung 80424, and the
| | - Kuang-Yu Dai
- Center for Neuroscience, National Sun Yat-sen University, Kaohsiung 80424, and the
| | - Samuel H H Chan
- Center for Neuroscience, National Sun Yat-sen University, Kaohsiung 80424, and the; Center for Gene Regulation and Signal Transduction Research, National Cheng Kung University, Tainan 70101, Taiwan
| | - Alice Y W Chang
- Center for Neuroscience, National Sun Yat-sen University, Kaohsiung 80424, and the; Center for Gene Regulation and Signal Transduction Research, National Cheng Kung University, Tainan 70101, Taiwan.
| |
Collapse
|
36
|
Pinti M, Salomoni P, Cossarizza A. Anti-HIV drugs and the mitochondria. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2006; 1757:700-7. [PMID: 16782042 DOI: 10.1016/j.bbabio.2006.05.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2005] [Revised: 05/02/2006] [Accepted: 05/05/2006] [Indexed: 12/11/2022]
Abstract
Several drugs are currently used that can significantly prolong the course of the infection with the human immunodeficiency virus (HIV), the cause of the acquired immunodeficiency syndrome (AIDS). Among these drugs, the nucleosidic inhibitors of viral reverse transcriptase can alter mitochondrial (mt) function by inhibiting the mitochondrial DNA polymerase gamma (the enzyme responsible for the replication of mtDNA). Decreased mtDNA content provokes a diminished synthesis of respiratory chain enzymes, leading to alterations in mt function. These are in turn responsible for a variety of side effects frequently observed in HIV+ patients, that range from hyperlactatemia and lactic acidosis to lipodystrophy, a pathology characterized by accumulation of visceral fat, breast adiposity, cervical fat-pads, hyperlipidemia, insulin resistance and fat wasting in face and limbs. In this paper, data concerning the effects of different compounds on mitochondria, their role in the pathogenesis of lipodystrophy, and problems related to studies on the mt toxicity of antiviral drugs are reviewed and thoroughly discussed.
Collapse
Affiliation(s)
- Marcello Pinti
- Department of Biomedical Sciences, Section of General Pathology, University of Modena and Reggio Emilia School of Medicine, via Campi 287, 41100 Modena, Italy
| | | | | |
Collapse
|
37
|
Chen YR, Chen CL, Yeh A, Liu X, Zweier JL. Direct and Indirect Roles of Cytochrome b in the Mediation of Superoxide Generation and NO Catabolism by Mitochondrial Succinate-Cytochrome c Reductase. J Biol Chem 2006; 281:13159-13168. [PMID: 16531408 DOI: 10.1074/jbc.m513627200] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Mitochondrial superoxide (O2*-) production is an important mediator of oxidative cellular injury. Succinate-cytochrome c reductase (SCR) of the electron transport chain has been implicated as an essential part of the mediation of O2*- generation and an alternative target of nitric oxide (NO) in the regulation of mitochondrial respiration. The Q cycle mechanism plays a central role in controlling both events. In the present work, O2*- generation by SCR was measured with the EPR spin-trapping technique using DEPMPO (5-diethoxylphosphoryl-5-methyl-1-pyrroline N-oxide) as the spin trap. In the presence of succinate, O2*- generation from SCR was detected as the spin adduct DEPMPO/*OOH. Inhibitors of the Q(o*-) site only marginally reduced (20-30%) this O2*- production, suggesting a secondary role of Q(o*-) in the mediation of O2*- generation. Addition of cyanide significantly decreased (approximately 70%) O2*- production, indicating the involvement of the heme component. UV-visible spectral analysis revealed that oxidation of ferrocytochrome b was accompanied by cytochrome c(1) reduction, and the reaction was mediated by the formation of an O2*- intermediate, indicating a direct role for cytochrome b in O2*- generation. In the presence of NO, DEPMPO/*OOH production was progressively diminished, implying that NO interacted with SCR or trapped the O2*-. The consumption of NO by SCR was investigated by electrochemical detection using an NO electrode. In the presence of succinate, SCR-mediated NO consumption was observed and inhibited by the addition of superoxide dismutase, suggesting the involvement of O2*-. Under the conditions of argon saturation, the NO consumption rate was not enhanced by succinate, suggesting a direct role for O2*- in the mediation of NO consumption. In the presence of succinate, oxidation of the ferrocytochrome b moiety of SCR was accelerated by the addition of NO, and was inhibited by argon saturation, indicating an indirect role for cytochrome b in the mediation of NO consumption.
Collapse
Affiliation(s)
- Yeong-Renn Chen
- Davis Heart & Lung Research Institute, Division of Cardiovascular Medicine, Department of Internal Medicine, Ohio State University, Columbus, Ohio 43210; Department of Molecular and Cellular Biochemistry, College of Medicine, Ohio State University, Columbus, Ohio 43210.
| | - Chwen-Lih Chen
- Davis Heart & Lung Research Institute, Division of Cardiovascular Medicine, Department of Internal Medicine, Ohio State University, Columbus, Ohio 43210
| | - Alexander Yeh
- Davis Heart & Lung Research Institute, Division of Cardiovascular Medicine, Department of Internal Medicine, Ohio State University, Columbus, Ohio 43210
| | - Xiaoping Liu
- Davis Heart & Lung Research Institute, Division of Cardiovascular Medicine, Department of Internal Medicine, Ohio State University, Columbus, Ohio 43210
| | - Jay L Zweier
- Davis Heart & Lung Research Institute, Division of Cardiovascular Medicine, Department of Internal Medicine, Ohio State University, Columbus, Ohio 43210; Department of Molecular and Cellular Biochemistry, College of Medicine, Ohio State University, Columbus, Ohio 43210
| |
Collapse
|
38
|
Chan JYH, Chang AYW, Chan SHH. New insights on brain stem death: From bedside to bench. Prog Neurobiol 2005; 77:396-425. [PMID: 16376477 DOI: 10.1016/j.pneurobio.2005.11.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2005] [Revised: 10/31/2005] [Accepted: 11/03/2005] [Indexed: 01/07/2023]
Abstract
As much as brain stem death is currently the clinical definition of death in many countries and is a phenomenon of paramount medical importance, there is a dearth of information on its mechanistic underpinnings. A majority of the clinical studies are concerned only with methods to determine brain stem death. Whereas a vast amount of information is available on the cellular and molecular mechanisms of cell death, rarely are these studies directed specifically towards the understanding of brain stem death. This review presents a framework for translational research on brain stem death that is based on systematically coordinated clinical and laboratory efforts that center on this phenomenon. It begins with the identification of a novel clinical marker from patients that is related specifically to brain stem death. After realizing that this "life-and-death" signal is related to the functional integrity of the brain stem, its origin is traced to the rostral ventrolateral medulla (RVLM). Subsequent laboratory studies on this neural substrate in animal models of brain stem death provide credence to the notion that both "pro-life" and "pro-death" programs are at work during the progression towards death. Those programs (mitochondrial functions, nitric oxide, peroxynitrite, superoxide anion, coenzyme Q10, heat shock proteins and ubiquitin-proteasome system) hitherto identified from the RVLM are presented, along with their cellular and molecular mechanisms. It is proposed that outcome of the interplay between the "pro-life" and "pro-death" programs (dying) in this neural substrate determines the final fate of the individual (being dead). Thus, identification of additional programs in the RVLM and delineation of their regulatory mechanisms should shed new lights on future directions for clinical management of life-and-death.
Collapse
Affiliation(s)
- Julie Y H Chan
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 81346, Taiwan, ROC
| | | | | |
Collapse
|
39
|
Hassan T, Timofeev EV, Saito T, Shimizu H, Ezura M, Matsumoto Y, Takayama K, Tominaga T, Takahashi A. A proposed parent vessel geometry-based categorization of saccular intracranial aneurysms: computational flow dynamics analysis of the risk factors for lesion rupture. J Neurosurg 2005; 103:662-80. [PMID: 16266049 DOI: 10.3171/jns.2005.103.4.0662] [Citation(s) in RCA: 139] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
OBJECT The authors created a simple, broadly applicable classification of saccular intracranial aneurysms into three categories: sidewall (SW), sidewall with branching vessel (SWBV), and endwall (EW) according to the angiographically documented patterns of their parent arteries. Using computational flow dynamics analysis (CFDA) of simple models representing the three aneurysm categories, the authors analyzed geometry-related risk factors such as neck width, parent artery curvature, and angulation of the branching vessels. METHODS The authors performed CFDAs of 68 aneurysmal geometric formations documented on angiograms that had been obtained in patients with 45 ruptured and 23 unruptured lesions. In successfully studied CFDA cases, the wall shear stress, blood velocity, and pressure maps were examined and correlated with aneurysm rupture points. Statistical analysis of the cases involving aneurysm rupture revealed a statistically significant correlation between aneurysm depth and both neck size (p < 0.0001) and caliber of draining arteries (p < 0.0001). Wider-necked aneurysms or those with wider-caliber draining vessels were found to be high-flow lesions that tended to rupture at larger sizes. Smaller-necked aneurysms or those with smaller-caliber draining vessels were found to be low-flow lesions that tended to rupture at smaller sizes. The incidence of ruptured aneurysms with an aspect ratio (depth/neck) exceeding 1.6 was 100% in the SW and SWBV categories, whereas the incidence was only 28.75% for the EW aneurysms. CONCLUSIONS The application of standardized categories enables the comparison of results for various aneurysms' geometric formations, thus assisting in their management. The proposed classification system may provide a promising means of understanding the natural history of saccular intracranial aneurysms.
Collapse
Affiliation(s)
- Tamer Hassan
- Department of Neuroendovascular Therapy, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Rakesh K, Agrawal DK. Cytokines and growth factors involved in apoptosis and proliferation of vascular smooth muscle cells. Int Immunopharmacol 2005; 5:1487-506. [PMID: 16023601 DOI: 10.1016/j.intimp.2005.05.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2004] [Revised: 05/09/2005] [Accepted: 05/09/2005] [Indexed: 11/21/2022]
Abstract
This review focuses on the role of cytokines and growth factors involved in the regulation of smooth muscle cells in an atherosclerotic plaque. As a plaque begins to develop, upon endothelial injury inflammatory cells within the lesion interact with the accumulating LDL, other inflammatory cells and smooth muscle cells and release cytokines and growth factors. The mediators released from the activated cells regulate the proliferation and/or survival of smooth muscle cells. This determines the stability and integrity of a plaque. New data emerging from various studies have provided novel insights into many of the cellular interactions and signaling mechanisms involving apoptosis of smooth muscle cells in the atherosclerotic plaques. A number of these studies, focusing on activation of inflammatory cells and the roles of chemokines, cytokines and growth factors, are addressed in this review.
Collapse
Affiliation(s)
- Kriti Rakesh
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE 68178, United States
| | | |
Collapse
|
41
|
Odeh M, Sabo E, Srugo I, Oliven A. Correlation between levels of tumour necrosis factor-alpha and levels of pH, glucose, and lactate dehydrogenase in parapneumonic effusions. J Infect 2005; 50:114-9. [PMID: 15667911 DOI: 10.1016/j.jinf.2004.05.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/19/2004] [Indexed: 11/25/2022]
Abstract
OBJECTIVES This study was undertaken to investigate the correlation, which has not been previously investigated, between levels of tumour necrosis factor-alpha (TNF) and levels of pH, glucose, and lactate dehydrogenase (LDH) in pleural fluid of patients with uncomplicated parapneumonic effusion (UCPPE), and patients with complicated parapneumonic effusion (CPPE). METHODS Using a commercially-available high sensitivity ELISA kit, levels of TNF were measured in pleural fluid of patients with UCPPE (n = 23), and CPPE (n = 15), and were compared with levels of pH, glucose, and LDH in these two groups. RESULTS The mean +/- SD values of pleural fluid TNF, pH, glucose, and LDH in the UCPPE group were 11.05 +/- 7.65 pg/ml, 7.41 +/- 0.08, 125 +/- 48 mg/dl, and 306 +/- 182 IU/l, respectively. In the CPPE group the values were 56.07 +/- 28.5 pg/ml, 6.82 +/- 0.25, 42 +/- 36 mg/dl, and 2096 +/- 1916 IU/l, respectively. The only significant correlation, which was negative, was found between levels of TNF and pH in the CPPE group (r = -0.62, P = 0.01). Levels of pleural fluid TNF and LDH were significantly higher, and levels of glucose were significantly lower in the CPPE group than in the UCPPE group (P < 0.0001). CONCLUSIONS This study demonstrates, for the first time that TNF levels correlate inversely with levels of pH in pleural fluid of patients with CPPE but not of patients with UCPPE. This correlation may, in part, explain the pathophysiology of the pleural complications which occur in the presence of CPPE.
Collapse
Affiliation(s)
- M Odeh
- Department of Internal Medicine B, Bnai Zion Medical Center, Technion Faculty of Medicine, Israel Institute of Technology, P.O. Box 6477, Haifa 31063, Israel.
| | | | | | | |
Collapse
|
42
|
Madonna R, Di Napoli P, Massaro M, Grilli A, Felaco M, De Caterina A, Tang D, De Caterina R, Geng YJ. Simvastatin Attenuates Expression of Cytokine-inducible Nitric-oxide Synthase in Embryonic Cardiac Myoblasts. J Biol Chem 2005; 280:13503-11. [PMID: 15705589 DOI: 10.1074/jbc.m411859200] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cardiac stem cells or myoblasts are vulnerable to inflammatory stimulation in hearts with infarction or ischemic injury. Widely used for the prevention and treatment of atherosclerotic heart disease, the cholesterol-lowering drugs statins may exert anti-inflammatory effects. In this study, we examined the impact of inhibition of hydroxymethylglutaryl coenzyme A (HMG-CoA) reductase with simvastatin on the expression of inducible nitric-oxide synthase (iNOS) in embryonic cardiac myoblasts stimulated with the proinflammatory cytokines, interleukin-1 or tumor necrosis factor. Treatment with simvastatin significantly reduced the levels of iNOS mRNA and protein in cytokine-treated rat H9c2 cardiac embryonic myoblasts. Addition of the HMG-CoA reductase product, L-mevalonate, and the by-product of cholesterol synthesis, geranylgeranyl pyrophosphate, could reverse the statin inhibitory effect on iNOS expression. Simvastatin treatment lowered the Rho GTPase activities, whereas the Rho-associated kinase inhibitor Y27632 partially blocked the statin inhibitory effect on nitrite production in the cytokine-treated H9c2 cells. Treatment with simvastatin led to inactivation of NF-kappaB by elevation of the NF-kappaB inhibitor IkappaB and reduction of the NF-kappaB nuclear contents in the cytokine-stimulated H9c2 cells. Hence, treatment with simvastatin can attenuate iNOS expression and NO synthesis in cytokine-stimulated embryonic cardiac myoblasts. The statin inhibitory effect may occur through isoprenoid-mediated intracellular signal transduction, which involves several key signal proteins, such as Rho kinase and IkappaB/NF-kappaB. These data suggest that statin therapy may protect the cardiac myocyte progenitors against the cytotoxicity of cytokine-induced high output of NO production in infarcted or ischemic hearts with inflammation.
Collapse
Affiliation(s)
- Rosalinda Madonna
- Institute of Cardiology and Center of Excellence on Aging, G. d'Annuzio Univeristy, Chieti, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Tatsumi T, Akashi K, Keira N, Matoba S, Mano A, Shiraishi J, Yamanaka S, Kobara M, Hibino N, Hosokawa S, Asayama J, Fushiki S, Fliss H, Nakagawa M, Matsubara H. Cytokine-induced nitric oxide inhibits mitochondrial energy production and induces myocardial dysfunction in endotoxin-treated rat hearts. J Mol Cell Cardiol 2005; 37:775-84. [PMID: 15350850 DOI: 10.1016/j.yjmcc.2004.06.014] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2004] [Revised: 05/24/2004] [Accepted: 06/18/2004] [Indexed: 11/27/2022]
Abstract
The mechanism responsible for cardiac depression in septic shock remains unknown. The present study examined whether nitric oxide (NO) overproduced by inducible NO synthase (iNOS) can inhibit aerobic energy metabolism and impair the myocardial function in endotoxin-treated rat hearts. Lipopolysaccharide (LPS) significantly decreased systolic blood pressure (BP) to 44% of control during the 48 h treatment. Hearts from control and LPS-treated rats were perfused in a Langendorff apparatus. After LPS injection, left ventricular (LV) developed pressure (LVDP) was significantly depressed, plasma NO2-/NO3- (NO(x)) concentration was markedly increased, and myocardial adenosine 5'-triphosphate (ATP), creatine phosphate (CrP), and the ratio of ATP/adenosine 5'-diphosphate were progressively decreased with time. Immunological examination showed a significant expression of iNOS protein in the LPS-treated myocytes. Aminoguanidine, an inhibitor of iNOS, significantly attenuated these LPS-induced functional and metabolic changes. Myocardial cyclic guanosine 3',5'-monophosphate (cGMP) content was significantly increased after LPS injection. Methylene blue, an inhibitor of soluble guanylate cyclase, blunted this increase in cGMP and significantly restored the LPS-induced contractile dysfunction 6 h after LPS injection. In addition, there was a significant negative correlation between LVDP and myocardial cGMP levels as well as a significant negative correlation between LVDP and plasma NO(x) levels. In contrast, 48 h after LPS injection, methylene blue no longer affected cardiac performance, and there was a significant positive correlation between LVDP and myocardial ATP content. Furthermore, the normalized activities (as a ratio of the citrate synthase activity) of mitochondrial NADH-CoQ reductase, succinate-CoQ reductase, and ATPase, were significantly inhibited, and the swelling or disruption of mitochondria cristae was seen in the 48 h LPS treatment. These LPS-induced functional and morphological disorders in the mitochondria were significantly improved by aminoguanidine. The findings suggest that sustained production of NO by iNOS leads to contractile dysfunction via cGMP in the early stage, but that it can directly impair the mitochondrial function, lower myocardial energy production, and contribute significantly to the myocardial dysfunction in the later stage of septic shock.
Collapse
Affiliation(s)
- Tetsuya Tatsumi
- Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602 8566, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Almeida A, Cidad P, Delgado-Esteban M, Fernández E, García-Nogales P, Bolaños JP. Inhibition of mitochondrial respiration by nitric oxide: its role in glucose metabolism and neuroprotection. J Neurosci Res 2005; 79:166-71. [PMID: 15573411 DOI: 10.1002/jnr.20281] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
There is an increasing body of evidence demonstrating that inhibition of cytochrome c oxidase by nitric oxide (NO) may be one more step in a signaling cascade involved in the physiologic regulation of cell functions. For example, in both astrocytes and neurons the inhibition of mitochondrial respiration by endogenously produced NO induces transient and modest decreases in cellular ATP concentrations. This mitochondrial impairment may serve as a cellular sensor of energy charges, hence modulating metabolic pathways, such as glycolysis, through AMP-activated protein kinase (AMPK) in astrocytes. In neurons, the NO derivative peroxynitrite anion triggers signaling pathways leading to glucose oxidation through the pentose-phosphate pathway to form reducing equivalents in the form of NADPH. The modulation of these metabolic pathways by nitric oxide or its derivatives may be important for understanding the mechanisms by which this free radical affects neuronal death or survival.
Collapse
Affiliation(s)
- Angeles Almeida
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Campus Miguel de Unamuno. 37007 Salamanca, Spain
| | | | | | | | | | | |
Collapse
|
45
|
Moyle G. Mechanisms of HIV and Nucleoside Reverse Transcriptase Inhibitor Injury to Mitochondria. Antivir Ther 2005. [DOI: 10.1177/135965350501002s05] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Graeme Moyle
- St Stephens HIV Research, Chelsea and Westminster Hospital, London, UK
| |
Collapse
|
46
|
Wang Z, Rao PJ, Castresana MR, Newman WH. TNF-α induces proliferation or apoptosis in human saphenous vein smooth muscle cells depending on phenotype. Am J Physiol Heart Circ Physiol 2005; 288:H293-301. [PMID: 15358608 DOI: 10.1152/ajpheart.00165.2004] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Tumor necrosis factor (TNF)-α is implicated in development of restenotic and atherosclerotic vascular lesions, which are pathological processes involving both proliferation and apoptosis of vascular smooth muscle cells (VSMCs). Human VSMCs were recently found to contain heterogeneous subpopulations. We therefore examined whether TNF has different effects on distinct subpopulations of VSMCs. With the use of cloning techniques, two stable subpopulations of VSMCs were isolated from human saphenous vein: spindle- and epithelioid-shaped smooth muscle cells (Sp- and Ep-SMCs, respectively). We found that TNF stimulated growth in Sp-SMCs but had a toxic effect on Ep-SMCs. TNF did not induce apoptosis in Sp-SMCs as determined by nuclear staining and cellular DNA electrophoresis. In contrast, the reduction of viability in Ep-SMCs was associated with induction of apoptosis as characterized by cellular DNA fragmentation and nuclear condensation. Higher levels of the TNF-R1 receptor subtype were detected in membrane preparations from Ep-SMCs than in membranes from Sp-SMCs. Activation of caspase-3 was also selectively induced in Ep-SMCs but not in Sp-SMCs. Cycloheximide, an inhibitor of protein synthesis, enhanced the toxicity of TNF in Ep-SMCs. This effect of cycloheximide was not seen in Sp-SMCs. The data presented here demonstrate for the first time that TNF either promotes growth or induces apoptosis in human VSMCs depending on phenotype.
Collapse
MESH Headings
- Apoptosis/drug effects
- Caspase 3
- Caspases/metabolism
- Cell Proliferation/drug effects
- Cell Survival/drug effects
- Cells, Cultured
- Cycloheximide/pharmacology
- Enzyme Activation/drug effects
- Humans
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/physiology
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/physiology
- Phenotype
- Protein Synthesis Inhibitors/pharmacology
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Recombinant Proteins/pharmacology
- Saphenous Vein/cytology
- Saphenous Vein/drug effects
- Saphenous Vein/physiology
- Tumor Necrosis Factor-alpha/pharmacology
Collapse
Affiliation(s)
- Zhongbiao Wang
- Division of Basic Medical Science, Mercer University School of Medicine, and Medical Center of Central Georgia, 1550 College St., Macon, GA 31207, USA.
| | | | | | | |
Collapse
|
47
|
Tsai AG, Cabrales P, Hangai-Hoger N, Intaglietta M. Oxygen distribution and respiration by the microcirculation. Antioxid Redox Signal 2004; 6:1011-8. [PMID: 15548898 DOI: 10.1089/ars.2004.6.1011] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Longitudinal and radial oxygen gradients in the microcirculation due to oxygen release from arterioles show that in some tissues oxygen is primarily supplied by arterioles and secondarily by capillaries. In several tissues, the arteriolar rate of oxygen exit is too large to be explained by diffusion alone, indicating that in these tissues oxygen consumption of the arteriolar wall in vivo is much greater than that shown in in vitro studies of endothelium and vascular smooth muscle, a phenomenon that may be related to the synthesis autocoids by the endothelium in vivo. The functional significance of the high metabolic rate of the arteriolar vessels may be related to the need of providing a metabolic barrier for protecting the parenchymal tissue from high oxygen levels in arterial blood, thus reducing formation of oxygen free radicals in the perivascular tissue, a supposition supported by the finding that the radial oxygen gradient at the microvascular wall and therefore its rate of oxygen consumption are proportional to local blood oxygen partial pressure (pO(2)). Oxygen consumption by the endothelium and/or smooth muscle is also a factor in causing terminal lymphatic pO(2) to have the lowest oxygen level in the tissue, rendering this compartment most vulnerable in hypoxic conditions.
Collapse
Affiliation(s)
- Amy G Tsai
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093-0412, USA
| | | | | | | |
Collapse
|
48
|
Mitra AK, Dhume AS, Agrawal DK. "Vulnerable plaques" — ticking of the time bomb. Can J Physiol Pharmacol 2004; 82:860-71. [PMID: 15573146 DOI: 10.1139/y04-095] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Atherosclerosis and its sequelae are one of the leading causes of morbidity and mortality, especially in the developed nations. Over the years, treatment protocols have changed with the changing understanding of the disease process. Inflammatory mechanisms have emerged as key players in the formation of the atherosclerotic plaque. For the majority of its life span, the plaque develops silently and only some exhibit overt clinical manifestations. The purpose of this review is to examine the inherent properties of some of these "vulnerable" or symptomatic plaques. Rupture of the plaque is related to the thickness of the fibrous cap overlying the necrotic lipid core. A thin cap is more likely to lead to rupture. Multiple factors broadly grouped as the "determinants of vulnerability" are responsible for directly or indirectly influencing the plaque dynamics. Apoptosis is considered an important underlying mechanism that contributes to plaque instability. Inflammatory reactions within the plaque trigger apoptosis by cell–cell contact and intra cellular death signaling. Once started, the apoptotic process affects all of the components that make up the plaque, including vascular smooth muscle cells, endothelial cells, and macrophages. Extensive research has identified many of the key cellular and molecular regulators that play a part in apoptosis within the atherosclerotic lesion. This information will help us to gain a better understanding of the underlying mechanisms at the cellular and molecular level and enable us to formulate better therapeutic strategies to combat this disease.Key words: apoptosis, atherosclerosis, inflammation, plaque stability, vulnerable plaques.
Collapse
Affiliation(s)
- Amit K Mitra
- Department of Biomedical Sciences, CRISS, Creighton, University School of Medicine, Omaha, NE 68178, USA
| | | | | |
Collapse
|
49
|
Li YJ, Du GH. Effects of alpinetin on rat vascular smooth muscle cells. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2004; 6:87-92. [PMID: 15008454 DOI: 10.1080/1028602031000135558] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
The object of this work was to study the effects of alpinetin on cultured rat aortic smooth muscle cells. It was observed that H2O2 (100 micromol L(-1)) induced increase of LDH (lactate dehydrogenase) leakage in the medium of VSMC by 7.4% (p < 0.01) and 10(-7) mol L(-1) alpinetin significantly decreased LDH leakage induced by H2O2 (p < 0.01). Alpinetin had the effects of inhibiting VSMC proliferation in a dose-dependent manner under the condition of serum stimulation for 24 and 48 h, but with serum stimulation for 72 h adverse effects on VSMC proliferation appeared. 10(-7) and 10(-8) mol L(-1) alpinetin had significantly inhibitory effects on VSMC migration by 67.9% (p < 0.001) and 34.1% (p < 0.01) respectively. It was also found that alpinetin (10(-7)-10(-9) mol L(-1)) could significantly inhibit the production of NO in cultured VSMC induced by TNFalpha (200 U ml(-1)). At 10(-7), 10(-8) and 10(-9) mol L(-1) the modulation of NO was by 22.6% (p < 0.001), 20.6% (p < 0.01) and 13.9% (p < 0.05), respectively. In summary, the data show that alpinetin has, to some extent, protective effects on VSMC.
Collapse
MESH Headings
- Animals
- Cell Division/drug effects
- Cell Movement/drug effects
- Cells, Cultured
- Flavanones
- Flavonoids/pharmacology
- Hydrogen Peroxide/pharmacology
- L-Lactate Dehydrogenase/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/metabolism
- Nitric Oxide/pharmacology
- Rats
- Rats, Wistar
- Tumor Necrosis Factor-alpha/pharmacology
Collapse
Affiliation(s)
- Yu-Juan Li
- Institute of Materia Medica, Peking Union Medical College, Chinese Academy of Medical Sciences, 1 Xian Nong Tan Street, Beijing 100050, China
| | | |
Collapse
|
50
|
Yen DHT, Chan JYH, Tseng HP, Huang CI, Lee CH, Chan SHH, Chang AYW. DEPRESSION OF MITOCHONDRIAL RESPIRATORY ENZYME ACTIVITY IN ROSTRAL VENTROLATERAL MEDULLA DURING ACUTE MEVINPHOS INTOXICATION IN THE RAT. Shock 2004; 21:358-63. [PMID: 15179137 DOI: 10.1097/00024382-200404000-00011] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
We investigated possible changes in bioenergetics at the rostral ventrolateral medulla (RVLM), a medullary site where sympathetic vasomotor tone originates and where the organophosphate poison mevinphos (Mev) acts to elicit cardiovascular intoxication. In Sprague-Dawley rats maintained under propofol anesthesia, microinjection bilaterally of Mev (10 nmol) into the RVLM induced progressive hypotension that was accompanied by an early augmentation (80-100 min post-Mev; Phase I), followed by a decrease (>100 min post-Mev; Phase II) in the power density of the vasomotor components (0-0.8 Hz) in systemic arterial pressure (SAP) signals. Enzyme assay revealed that local application of Mev into the RVLM also significantly and progressively depressed the activity of NADH cytochrome c reductase (marker for Complexes I and III) and cytochrome c oxidase (marker for Complex IV) in the mitochondrial respiratory chain of the RVLM, but not the heart. On the other hand, the activity of succinate cytochrome c reductase (marker for Complexes II and III) remained unaltered. Both the cardiovascular consequences and depression of mitochondrial respiratory chain enzymes elicited by Mev were significantly antagonized on comicroinjection of atropine (3.5 or 7 nmol) bilaterally into the RVLM. We conclude that Mev adversely effects cardiovascular control by acting as a cholinesterase inhibitor in the RVLM, whose neuronal activity is intimately related to the death process. The resulting accumulation of acetylcholine and prolonged activation of muscarinic receptors in the RVLM is manifested by a selective dysfunction of respiratory enzyme Complexes I and IV in the mitochondrial respiratory chain that underlies cardiovascular toxicity associated with organophosphate poisons such as Mev.
Collapse
Affiliation(s)
- David H T Yen
- Department of Emergency Medicine, Taipei-Veterans General Hospital, Taipei 112, Republic of China
| | | | | | | | | | | | | |
Collapse
|