1
|
Kirby NV, Meade RD, McCormick JJ, King KE, Notley SR, Kenny GP. Brain-derived neurotrophic factor in older adults exposed to simulated indoor overheating. Eur J Appl Physiol 2024:10.1007/s00421-024-05623-y. [PMID: 39417862 DOI: 10.1007/s00421-024-05623-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024]
Abstract
PURPOSE Brain-derived neurotrophic factor (BDNF) is a neuroprotective growth factor that increases in young adults during short, intense bouts of passive heat stress. However, this may not reflect the response in heat-vulnerable populations exposed to air temperatures more consistent with indoor overheating during hot weather and heatwaves, especially as the BDNF response to acute stressors may diminish with increasing age. We therefore evaluated the ambient and body temperature-dependent responses of BDNF in older adults during daylong passive heating. METHODS Sixteen older adults (6 females; aged 66-78 years) completed 8-h exposure to four randomized ambient conditions simulating those experienced indoors during hot weather and heatwaves in continental climates: 22 °C (air-conditioning; control), 26 °C (health-agency-recommended indoor temperature limit), 31 °C, and 36 °C (non-airconditioned home); all 45% relative humidity. To further investigate upstream mechanisms of BDNF regulation during thermal strain, we also explored associations between BDNF and circulating heat shock protein 70 (HSP70; taken as an indicator of the heat shock response). RESULTS Circulating BDNF was elevated by ~ 28% (1139 [95%CI: 166, 2112] pg/mL) at end-exposure in the 36 °C compared to the 22 °C control condition (P = 0.026; 26 °C-and 31 °C-22 °C differences: P ≥ 0.090), increasing 90 [22, 158] pg/mL per 1 °C rise in ambient temperature (linear trend: P = 0.011). BDNF was also positively correlated with mean body temperatures (P = 0.013), which increased 0.12 [0.10, 0.13]°C per 1 °C rise in ambient temperature (P < 0.001). By contrast, serum HSP70 did not change across conditions (P ≥ 0.156), nor was it associated with BDNF (P = 0.376). CONCLUSION Our findings demonstrate a progressive increase in circulating BDNF during indoor overheating in older adults.
Collapse
Affiliation(s)
- Nathalie V Kirby
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, 125 University Private, Montpetit Hall, Room 367, Ottawa, ON, K1N 6N5, Canada
| | - Robert D Meade
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, 125 University Private, Montpetit Hall, Room 367, Ottawa, ON, K1N 6N5, Canada
| | - James J McCormick
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, 125 University Private, Montpetit Hall, Room 367, Ottawa, ON, K1N 6N5, Canada
| | - Kelli E King
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, 125 University Private, Montpetit Hall, Room 367, Ottawa, ON, K1N 6N5, Canada
| | - Sean R Notley
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, 125 University Private, Montpetit Hall, Room 367, Ottawa, ON, K1N 6N5, Canada
| | - Glen P Kenny
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, 125 University Private, Montpetit Hall, Room 367, Ottawa, ON, K1N 6N5, Canada.
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.
| |
Collapse
|
2
|
Gatta C, Avallone L, Costagliola A, Scocco P, D’Angelo L, de Girolamo P, De Felice E. Immunolocalization of Two Neurotrophins, NGF and BDNF, in the Pancreas of the South American Sea Lion Otaria flavescens and Bottlenose Dolphin Tursiops truncatus. Animals (Basel) 2024; 14:2336. [PMID: 39199870 PMCID: PMC11350702 DOI: 10.3390/ani14162336] [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: 07/22/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 09/01/2024] Open
Abstract
In this study, we have investigated the immunolocalization of NGF (Nerve Growth Factor) and BDNF (Brain-Derived Neurotrophic Factor) in the pancreas of two species of marine mammals: Tursiops truncatus (common bottlenose dolphin), belonging to the order of the Artiodactyla, and Otaria flavescens (South American sea lion), belonging to the order of the Carnivora. Our results demonstrated a significant presence of NGF and BDNF in the pancreas of both species with a wide distribution pattern observed in the exocrine and endocrine components. We identified some differences that can be attributed to the different feeding habits of the two species, which possess a different morphological organization of the digestive system. Altogether, these preliminary observations open new perspectives on the function of neurotrophins and the adaptive mechanisms of marine mammals in the aquatic environment, suggesting potential parallels between the physiology of marine and terrestrial mammals.
Collapse
Affiliation(s)
- Claudia Gatta
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Via F. Delpino 1, 80137 Naples, Italy; (C.G.); (L.A.); (A.C.); (L.D.); (P.d.G.)
| | - Luigi Avallone
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Via F. Delpino 1, 80137 Naples, Italy; (C.G.); (L.A.); (A.C.); (L.D.); (P.d.G.)
| | - Anna Costagliola
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Via F. Delpino 1, 80137 Naples, Italy; (C.G.); (L.A.); (A.C.); (L.D.); (P.d.G.)
| | - Paola Scocco
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032 Camerino, Italy;
| | - Livia D’Angelo
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Via F. Delpino 1, 80137 Naples, Italy; (C.G.); (L.A.); (A.C.); (L.D.); (P.d.G.)
| | - Paolo de Girolamo
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Via F. Delpino 1, 80137 Naples, Italy; (C.G.); (L.A.); (A.C.); (L.D.); (P.d.G.)
| | - Elena De Felice
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032 Camerino, Italy;
| |
Collapse
|
3
|
Dubin RF, Deo R, Ren Y, Wang J, Pico AR, Mychaleckyj JC, Kozlitina J, Arthur V, Lee H, Shah A, Feldman H, Bansal N, Zelnick L, Rao P, Sukul N, Raj DS, Mehta R, Rosas SE, Bhat Z, Weir MR, He J, Chen J, Kansal M, Kimmel PL, Ramachandran VS, Waikar SS, Segal MR, Ganz P. Incident heart failure in chronic kidney disease: proteomics informs biology and risk stratification. Eur Heart J 2024; 45:2752-2767. [PMID: 38757788 PMCID: PMC11313584 DOI: 10.1093/eurheartj/ehae288] [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: 07/30/2023] [Revised: 04/09/2024] [Accepted: 04/25/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND AND AIMS Incident heart failure (HF) among individuals with chronic kidney disease (CKD) incurs hospitalizations that burden patients and health care systems. There are few preventative therapies, and the Pooled Cohort equations to Prevent Heart Failure (PCP-HF) perform poorly in the setting of CKD. New drug targets and better risk stratification are urgently needed. METHODS In this analysis of incident HF, SomaScan V4.0 (4638 proteins) was analysed in 2906 participants of the Chronic Renal Insufficiency Cohort (CRIC) with validation in the Atherosclerosis Risk in Communities (ARIC) study. The primary outcome was 14-year incident HF (390 events); secondary outcomes included 4-year HF (183 events), HF with reduced ejection fraction (137 events), and HF with preserved ejection fraction (165 events). Mendelian randomization and Gene Ontology were applied to examine causality and pathways. The performance of novel multi-protein risk models was compared to the PCP-HF risk score. RESULTS Over 200 proteins were associated with incident HF after adjustment for estimated glomerular filtration rate at P < 1 × 10-5. After adjustment for covariates including N-terminal pro-B-type natriuretic peptide, 17 proteins remained associated at P < 1 × 10-5. Mendelian randomization associations were found for six proteins, of which four are druggable targets: FCG2B, IGFBP3, CAH6, and ASGR1. For the primary outcome, the C-statistic (95% confidence interval [CI]) for the 48-protein model in CRIC was 0.790 (0.735, 0.844) vs. 0.703 (0.644, 0.762) for the PCP-HF model (P = .001). C-statistic (95% CI) for the protein model in ARIC was 0.747 (0.707, 0.787). CONCLUSIONS Large-scale proteomics reveal novel circulating protein biomarkers and potential mediators of HF in CKD. Proteomic risk models improve upon the PCP-HF risk score in this population.
Collapse
Affiliation(s)
- Ruth F Dubin
- Division of Nephrology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, H5.122E, Dallas, TX 75390, USA
| | - Rajat Deo
- Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Yue Ren
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jianqiao Wang
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Alexander R Pico
- Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA, USA
| | - Josyf C Mychaleckyj
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Julia Kozlitina
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Victoria Arthur
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hongzhe Lee
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amil Shah
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Harold Feldman
- Patient-Centered Outcomes Research Institute, Washington, DC, USA
| | - Nisha Bansal
- Division of Nephrology, University of Washington Medical Center, Seattle, WA, USA
| | - Leila Zelnick
- Division of Nephrology, University of Washington Medical Center, Seattle, WA, USA
| | - Panduranga Rao
- Division of Nephrology, University of Michigan, Ann Arbor, MI, USA
| | - Nidhi Sukul
- Division of Nephrology, University of Michigan, Ann Arbor, MI, USA
| | - Dominic S Raj
- Division of Kidney Diseases and Hypertension, George Washington University School of Medicine, Washington, DC, USA
| | - Rupal Mehta
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, USA
| | - Sylvia E Rosas
- Joslin Diabetes Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Zeenat Bhat
- Division of Nephrology, University of Michigan, Ann Arbor, MI, USA
| | - Matthew R Weir
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jiang He
- Department of Epidemiology, Tulane University, New Orleans, LA, USA
| | - Jing Chen
- Department of Epidemiology, Tulane University, New Orleans, LA, USA
| | - Mayank Kansal
- Division of Cardiology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Paul L Kimmel
- Division of Kidney, Urologic, and Hematologic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Vasan S Ramachandran
- University of Texas School of Public Health San Antonio and the University of Texas Health Sciences Center in San Antonio, Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Sushrut S Waikar
- Section of Nephrology, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Mark R Segal
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Peter Ganz
- Division of Cardiology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
4
|
Medved S, Salinas J, Kojis D, Weinstein G, Vasan RS, Beiser A, Seshadri S. The association between levels of brain-derived neurotrophic factor and comorbid depression in patients with cardiovascular disease: The Framingham Heart Study. Psychiatry Clin Neurosci 2024; 78:438-445. [PMID: 38842141 PMCID: PMC11410362 DOI: 10.1111/pcn.13664] [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: 10/17/2023] [Revised: 02/14/2024] [Accepted: 03/01/2024] [Indexed: 06/07/2024]
Abstract
AIM The current study aims to investigate the association of serum brain-derived neurotrophic factor (BDNF) levels with symptoms of depression in adults with and without prevalent cardiovascular disease (CVD), an often burdensome comorbidity. METHODS This cross-sectional study included participants from FHS (Framingham Heart Study) who had available serum BDNF levels. Depressive symptoms were assessed using the Center for Epidemiological Studies-Depression Scale (CES-D) with a score ≥16 indicating mild to moderate and ≥21 severe depression. Participants taking antidepressant medications were excluded from the study. RESULTS Altogether 3716 FHS participants were included in the final analysis (mean age, 64.3 ± 11.5 years; 55% women). After adjusting for potential confounders, greater BDNF levels were associated with reduced severe depression risk (odds ratio [OR], 0.78 [95% CI, 0.64-0.96]; P = 0.016). Among participants with CVD, greater BDNF levels were related to lower risk of depressive symptoms (CES-D ≥ 16 OR, 0.63 [95% CI, 0.45-0.89], P = 0.008; CES-D ≥ 21 OR, 0.49 [95% CI, 0.31-0.76], P = 0.002). The inverse relationship between BDNF and depressive symptom risk was present in women with CVD (CES-D ≥ 16 OR, 0.63 [95% CI, 0.40-0.99], P = 0.047; CES-D ≥ 21 OR, 0.38 [95% CI, 0.21-0.70], P = 0.002) but not in men. CONCLUSION Lower serum BDNF levels are associated with a higher risk of depressive symptoms in CVD, particularly among women. These findings implicate BDNF in the complex biological mechanisms that underlie prior associations observed between CVD and depression. To reduce the burden of depression in the large proportion of midlife and older adults with CVD, a better understanding of how BDNF may modify these pathways is merited.
Collapse
Affiliation(s)
- Sara Medved
- Department of Psychiatry and Psychological Medicine, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Joel Salinas
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, USA
| | - Daniel Kojis
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
- Boston University’s and National Heart, Lung, and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts, USA
| | | | - Ramachandran S. Vasan
- Boston University’s and National Heart, Lung, and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts, USA
- Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Alexa Beiser
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
- Boston University’s and National Heart, Lung, and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts, USA
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Sudha Seshadri
- Boston University’s and National Heart, Lung, and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts, USA
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, USA
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, Texas, USA
| |
Collapse
|
5
|
Paribello P, Manchia M, Isayeva U, Upali M, Orrù D, Pinna F, Collu R, Primavera D, Deriu L, Caboni E, Iaselli MN, Sundas D, Tusconi M, Scherma M, Pisanu C, Meloni A, Zai CC, Congiu D, Squassina A, Fratta W, Fadda P, Carpiniello B. A Secondary Analysis of the Complex Interplay between Psychopathology, Cognitive Functions, Brain Derived Neurotrophic Factor Levels, and Suicide in Psychotic Disorders: Data from a 2-Year Longitudinal Study. Int J Mol Sci 2024; 25:7922. [PMID: 39063164 PMCID: PMC11276839 DOI: 10.3390/ijms25147922] [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: 06/20/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Identifying phenotypes at high risk of suicidal behaviour is a relevant objective of clinical and translational research and can facilitate the identification of possible candidate biomarkers. We probed the potential association and eventual stability of neuropsychological profiles and serum BDNF concentrations with lifetime suicide ideation and attempts (LSI and LSA, respectively) in individuals with schizophrenia (SCZ) and schizoaffective disorder (SCA) in a 2-year follow-up study. A secondary analysis was conducted on a convenience sample of previously recruited subjects from a single outpatient clinic. Retrospectively assessed LSI and LSA were recorded by analysing the available longitudinal clinical health records. LSI + LSA subjects consistently exhibited lower PANSS-defined negative symptoms and better performance in the BACS-letter fluency subtask. There was no significant association between BDNF levels and either LSI or LSA. We found a relatively stable pattern of lower negative symptoms over two years among patients with LSI and LSA. No significant difference in serum BDNF concentrations was detected. The translational viability of using neuropsychological profiles as a possible avenue for the identification of populations at risk for suicide behaviours rather than the categorical diagnosis represents a promising option but requires further confirmation.
Collapse
Affiliation(s)
- Pasquale Paribello
- Unit of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy; (P.P.); (D.P.); (L.D.); (M.T.)
- Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, 09124 Cagliari, Italy
| | - Mirko Manchia
- Unit of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy; (P.P.); (D.P.); (L.D.); (M.T.)
- Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, 09124 Cagliari, Italy
- Department of Pharmacology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Ulker Isayeva
- Unit of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy; (P.P.); (D.P.); (L.D.); (M.T.)
- Division of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (R.C.); (C.P.); (A.S.); (W.F.); (P.F.)
| | - Marco Upali
- Unit of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy; (P.P.); (D.P.); (L.D.); (M.T.)
- Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, 09124 Cagliari, Italy
| | - Davide Orrù
- Unit of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy; (P.P.); (D.P.); (L.D.); (M.T.)
- Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, 09124 Cagliari, Italy
| | - Federica Pinna
- Unit of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy; (P.P.); (D.P.); (L.D.); (M.T.)
- Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, 09124 Cagliari, Italy
| | - Roberto Collu
- Division of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (R.C.); (C.P.); (A.S.); (W.F.); (P.F.)
| | - Diego Primavera
- Unit of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy; (P.P.); (D.P.); (L.D.); (M.T.)
| | - Luca Deriu
- Unit of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy; (P.P.); (D.P.); (L.D.); (M.T.)
- Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, 09124 Cagliari, Italy
| | - Edoardo Caboni
- Unit of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy; (P.P.); (D.P.); (L.D.); (M.T.)
- Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, 09124 Cagliari, Italy
| | - Maria Novella Iaselli
- Unit of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy; (P.P.); (D.P.); (L.D.); (M.T.)
- Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, 09124 Cagliari, Italy
| | - Davide Sundas
- Unit of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy; (P.P.); (D.P.); (L.D.); (M.T.)
- Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, 09124 Cagliari, Italy
| | - Massimo Tusconi
- Unit of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy; (P.P.); (D.P.); (L.D.); (M.T.)
| | - Maria Scherma
- Division of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (R.C.); (C.P.); (A.S.); (W.F.); (P.F.)
| | - Claudia Pisanu
- Division of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (R.C.); (C.P.); (A.S.); (W.F.); (P.F.)
| | - Anna Meloni
- Division of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (R.C.); (C.P.); (A.S.); (W.F.); (P.F.)
| | - Clement C. Zai
- Tanenbaum Centre for Pharmacogenetics, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada;
- Laboratory Medicine and Pathobiology, Department of Psychiatry, Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Donatella Congiu
- Division of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (R.C.); (C.P.); (A.S.); (W.F.); (P.F.)
| | - Alessio Squassina
- Division of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (R.C.); (C.P.); (A.S.); (W.F.); (P.F.)
| | - Walter Fratta
- Division of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (R.C.); (C.P.); (A.S.); (W.F.); (P.F.)
- Centre of Excellence “Neurobiology of Dependence”, University of Cagliari, 09124 Cagliari, Italy
| | - Paola Fadda
- Division of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (R.C.); (C.P.); (A.S.); (W.F.); (P.F.)
- Centre of Excellence “Neurobiology of Dependence”, University of Cagliari, 09124 Cagliari, Italy
| | - Bernardo Carpiniello
- Unit of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy; (P.P.); (D.P.); (L.D.); (M.T.)
- Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, 09124 Cagliari, Italy
| |
Collapse
|
6
|
Pressler SJ, Jung M, Dorsey SG, Giordani B, Titler MG, Wierenga KL, Clark DG, Smith DG, Smith AB, Gradus-Pizlo I, Pressler ML. Atrial Fibrillation and Older Age Predict Serum Brain-Derived Neurotrophic Factor Levels Among Patients With Heart Failure. J Cardiovasc Nurs 2024:00005082-990000000-00206. [PMID: 39007747 DOI: 10.1097/jcn.0000000000001110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
BACKGROUND Predictors have not been determined of serum brain-derived neurotrophic factor (BDNF) levels among patients with heart failure (HF). OBJECTIVE The primary purpose was to evaluate history of atrial fibrillation, age, gender, and left ventricular ejection fraction as predictors of serum BDNF levels at baseline, 10 weeks, and 4 and 8 months after baseline among patients with HF. METHODS This study was a retrospective cohort analyses of 241 patients with HF. Data were retrieved from the patients' health records (coded history of atrial fibrillation, left ventricular ejection fraction), self-report (age, gender), and serum BDNF. Linear multiple regression analyses were conducted. RESULTS One hundred three patients (42.7%) had a history of atrial fibrillation. History of atrial fibrillation was a significant predictor of serum BDNF levels at baseline (β = -0.16, P = .016), 4 months (β = -0.21, P = .005), and 8 months (β = -0.19, P = .015). Older age was a significant predictor at 10 weeks (β = -0.17, P = .017) and 4 months (β = -0.15, P = .046). CONCLUSIONS Prospective studies are needed to validate these results. Clinicians need to assess patients with HF for atrial fibrillation and include treatment of it in management plans.
Collapse
|
7
|
Ning B, Ge T, Wu Y, Wang Y, Zhao M. Role of Brain-Derived Neurotrophic Factor in Anxiety or Depression After Percutaneous Coronary Intervention. Mol Neurobiol 2024; 61:2921-2937. [PMID: 37946008 DOI: 10.1007/s12035-023-03758-1] [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: 08/17/2023] [Accepted: 10/31/2023] [Indexed: 11/12/2023]
Abstract
Anxiety or depression after percutaneous coronary intervention (PCI) is one of the key clinical problems in cardiology that need to be solved urgently. Brain-derived neurotrophic factor (BDNF) may be a potential biomarker for the pathogenesis and treatment of anxiety or depression after PCI. This article reviews the correlation between BDNF and cardiovascular system and nervous system from the aspects of synthesis, release and action site of BDNF, and focuses on the latest research progress of the mechanism of BDNF in anxiety or depression after PCI. It includes the specific mechanisms by which BDNF regulates the levels of inflammatory factors, reduces oxidative stress damage, and mediates multiple signaling pathways. In addition, this review summarizes the therapeutic potential of BDNF as a potential biomarker for anxiety or depression after PCI.
Collapse
Affiliation(s)
- Bo Ning
- First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Teng Ge
- First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Yongqing Wu
- First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Yuting Wang
- First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
- Affiliated Hospital, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Mingjun Zhao
- First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, 712046, China.
- Affiliated Hospital, Shaanxi University of Chinese Medicine, Xianyang, 712046, China.
- Shaanxi Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Cardiovascular Diseases, Xianyang, 712046, China.
| |
Collapse
|
8
|
Zou J, Hao S. A potential research target for cardiac rehabilitation: brain-derived neurotrophic factor. Front Cardiovasc Med 2024; 11:1348645. [PMID: 38707889 PMCID: PMC11069312 DOI: 10.3389/fcvm.2024.1348645] [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: 12/03/2023] [Accepted: 03/19/2024] [Indexed: 05/07/2024] Open
Abstract
Cardiovascular diseases pose a major threat to human life, functional activity, and quality of life. Once the disease is present, patients can experience varying degrees of problems or limitations on three levels: physical, psychological, and social. Patients with cardiovascular disease are always at risk for adverse cardiac events, decreased physical activity, psychoemotional disturbances, and limited social participation due to their varying pathologies. Therefore, personalized cardiac rehabilitation is of great significance in improving patients' physical and mental functions, controlling disease progression, and preventing deterioration. There is a consensus on the benefits of cardiac rehabilitation in improving patients' quality of life, enhancing functional activity, and reducing mortality. As an important part of cardiac rehabilitation, Exercise plays an irreplaceable role. Aerobic exercise, resistance training, flexibility training, and other forms of exercise are recommended by many experts. Improvements in exercise tolerance, lipid metabolism, cardiac function, and psychological aspects of the patients were evident with appropriate exercise interventions based on a comprehensive assessment. Further studies have found that brain-derived neurotrophic factor may be an important mediator of exercise's ability to improve cardiovascular health. Brain-derived neurotrophic factor exerts multiple biological effects on the cardiovascular system. This article provides another perspective on the cardiac effects of exercise and further looks at the prospects for the use of brain-derived neurotrophic factor in cardiac rehabilitation. Meanwhile, the new idea that brain-derived neurotrophic factor is a key mediator connecting the brain-cardiac axis is proposed in light of the current research progress, to provide new ideas for clinical rehabilitation and scientific research.
Collapse
Affiliation(s)
- Jianpeng Zou
- Department of Rehabilitation and Physiotherapy, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shijie Hao
- College of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
9
|
Boukhatem I, Fleury S, Jourdi G, Lordkipanidzé M. The intriguing role of platelets as custodians of brain-derived neurotrophic factor. Res Pract Thromb Haemost 2024; 8:102398. [PMID: 38706782 PMCID: PMC11066552 DOI: 10.1016/j.rpth.2024.102398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/26/2024] [Accepted: 03/18/2024] [Indexed: 05/07/2024] Open
Abstract
A State of the Art lecture titled "Platelets and neurotrophins" was presented at the International Society on Thrombosis and Haemostasis Congress in 2023. Neurotrophins, a family of neuronal growth factors known to support cognitive function, are increasingly recognized as important players in vascular health. Indeed, along with their canonical receptors, neurotrophins are expressed in peripheral tissues, particularly in the vasculature. The better-characterized neurotrophin in vascular biology is the brain-derived neurotrophic factor (BDNF). Its largest extracerebral pool resides within platelets, partly inherited from megakaryocytes and also likely internalized from circulation. Activation of platelets releases vast amounts of BDNF into their milieu and interestingly leads to platelet aggregation through binding of its receptor, the tropomyosin-related kinase B, on the platelet surface. As BDNF is readily available in plasma, a mechanism to preclude excessive platelet activation and aggregation appears critical. As such, binding of BDNF to α2-macroglobulin hinders its ability to bind its receptor and limits its platelet-activating effects to the site of vascular injury. Altogether, addition of BDNF to a forming clot facilitates not only paracrine platelet activation but also binding to fibrinogen, rendering the resulting clot more porous and plasma-permeable. Importantly, release of BDNF into circulation also appears to be protective against adverse cardiovascular and cerebrovascular outcomes, which has been reported in both animal models and epidemiologic studies. This opens an avenue for platelet-based strategies to deliver BDNF to vascular lesions and facilitate wound healing through its regenerative properties. Finally, we summarize relevant new data on this topic presented during the 2023 International Society on Thrombosis and Haemostasis Congress.
Collapse
Affiliation(s)
- Imane Boukhatem
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
| | - Samuel Fleury
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
| | - Georges Jourdi
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
- Université Paris Cité, Institut National de la Santé Et de la Recherche Médicale, Innovative Therapies in Haemostasis, Paris, France
- Service d’Hématologie Biologique, Assistance Publique : Hôpitaux de Paris, Hôpital Lariboisière, Paris, France
| | - Marie Lordkipanidzé
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
10
|
Shi L, Luo J, Wei X, Xu X, Tu L. The protective role of ginsenoside Rg3 in heart diseases and mental disorders. Front Pharmacol 2024; 15:1327033. [PMID: 38469409 PMCID: PMC10926849 DOI: 10.3389/fphar.2024.1327033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 02/07/2024] [Indexed: 03/13/2024] Open
Abstract
Ginsenoside Rg3, a compound derived from Panax ginseng C. A. Mey., is increasingly recognized for its wide range of pharmacological effects. Under the worldwide healthcare challenges posed by heart diseases, Rg3 stands out as a key subject in modern research on Chinese herbal medicine, offering a novel approach to therapy. Mental illnesses are significant contributors to global disease mortality, and there is a well-established correlation between cardiac and psychiatric conditions. This connection is primarily due to dysfunctions in the sympathetic-adrenomedullary system (SAM), the hypothalamic-pituitary-adrenal axis, inflammation, oxidative stress, and brain-derived neurotrophic factor impairment. This review provides an in-depth analysis of Rg3's therapeutic benefits and its pharmacological actions in treating cardiac and mental health disorders respectively. Highlighting its potential for the management of these conditions, Rg3 emerges as a promising, multifunctional therapeutic agent.
Collapse
Affiliation(s)
- Lili Shi
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Jinlan Luo
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Xiupan Wei
- Department of Rehabilitation Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| | - Xizhen Xu
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Tu
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| |
Collapse
|
11
|
Wsół A. Cardiovascular safety of psychedelic medicine: current status and future directions. Pharmacol Rep 2023; 75:1362-1380. [PMID: 37874530 PMCID: PMC10661823 DOI: 10.1007/s43440-023-00539-4] [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: 08/29/2023] [Revised: 10/02/2023] [Accepted: 10/02/2023] [Indexed: 10/25/2023]
Abstract
Psychedelics are powerful psychoactive substances that alter perception and mood processes. Their effectiveness in the treatment of psychiatric diseases was known before their prohibition. An increasing number of recent studies, due to the indisputable resurgence of serotonergic hallucinogens, have shown their efficacy in alleviating depression, anxiety, substance abuse therapies, and existential distress treatment in patients facing life-threatening illness. Psychedelics are generally considered to be physiologically safe with low toxicity and low addictive potential. However, their agonism at serotonergic receptors should be considered in the context of possible serotonin-related cardiotoxicity (5-HT2A/2B and 5-HT4 receptors), influence on platelet aggregation (5-HT2A receptor), and their proarrhythmic potential. The use of psychedelics has also been associated with significant sympathomimetic effects in both experimental and clinical studies. Therefore, the present review aims to provide a critical discussion of the cardiovascular safety of psilocybin, d-lysergic acid diethylamide (LSD), N,N-dimethyltryptamine, ayahuasca, and mescaline, based on the results of experimental research and clinical trials in humans. Experimental studies provide inconsistent information on the potential cardiovascular effects and toxicity of psychedelics. Data from clinical trials point to the relative cardiovascular safety of psychedelic-assisted therapies in the population of "healthy" volunteers. However, there is insufficient evidence from therapies carried out with microdoses of psychedelics, and there is still a lack of data on the safety of psychedelics in the population of patients with cardiovascular disease. Therefore, the exact determination of the cardiovascular safety of psychedelic therapies (especially long-term therapies) requires further research.
Collapse
Affiliation(s)
- Agnieszka Wsół
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland.
| |
Collapse
|
12
|
Fioranelli M, Garo ML, Roccia MG, Prizbelek B, Sconci FR. Brain-Heart Axis: Brain-Derived Neurotrophic Factor and Cardiovascular Disease-A Review of Systematic Reviews. Life (Basel) 2023; 13:2252. [PMID: 38137853 PMCID: PMC10744648 DOI: 10.3390/life13122252] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/22/2023] [Accepted: 11/24/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND The brain-heart axis is an intra- and bidirectional complex that links central nervous system dysfunction and cardiac dysfunction. In recent decades, brain-derived neurotrophic factor (BDNF) has emerged as a strategic molecule involved in both brain and cardiovascular disease (CVD). This systematic review of systematic reviews aimed to (1) identify and summarize the evidence for the BDNF genotype and BDNF concentration in CVD risk assessment, (2) evaluate the evidence for the use of BDNF as a biomarker of CVD recovery, and (3) evaluate rehabilitation approaches that can restore BDNF concentration. METHODS A comprehensive search strategy was developed using PRISMA. The risk of bias was assessed via ROBIS. RESULTS Seven studies were identified, most of which aimed to evaluate the role of BDNF in stroke patients. Only two systematic reviews examined the association of BDNF concentration and polymorphism in CVDs other than stroke. CONCLUSIONS The overall evidence showed that BDNF plays a fundamental role in assessing the risk of CVD occurrence, because lower BDNF concentrations and rs6265 polymorphism are often associated with CVD. Nevertheless, much work remains to be carried out in current research to investigate how BDNF is modulated in different cardiovascular diseases and in different populations.
Collapse
Affiliation(s)
- Massimo Fioranelli
- Department of Human Sciences, Guglielmo Marconi University, 00193 Rome, Italy; (M.F.); (M.G.R.)
| | - Maria Luisa Garo
- Istituto Terapie Sistemiche Integrate, Casa di Cura Sanatrix, 00199 Rome, Italy; (B.P.); (F.R.S.)
| | - Maria Grazia Roccia
- Department of Human Sciences, Guglielmo Marconi University, 00193 Rome, Italy; (M.F.); (M.G.R.)
| | - Bianca Prizbelek
- Istituto Terapie Sistemiche Integrate, Casa di Cura Sanatrix, 00199 Rome, Italy; (B.P.); (F.R.S.)
| | - Francesca Romana Sconci
- Istituto Terapie Sistemiche Integrate, Casa di Cura Sanatrix, 00199 Rome, Italy; (B.P.); (F.R.S.)
| |
Collapse
|
13
|
Shobeiri P, Behnoush AH, Khalaji A, Teixeira AL, Rezaei N. Peripheral Levels of the Brain-Derived Neurotrophic Factor in Coronary Artery Disease: A Systematic Review and Meta-Analysis. J Tehran Heart Cent 2023; 18:244-255. [PMID: 38680638 PMCID: PMC11053235 DOI: 10.18502/jthc.v18i4.14823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 08/05/2023] [Indexed: 05/01/2024] Open
Abstract
Background Among its functions, brain-derived neurotrophic factor (BDNF) regulates endothelial and macrophage activation, possibly playing a role in atherosclerotic plaque pathophysiology. Given contradicting reports, this study sought to investigate whether blood levels of BDNF differed between patients with coronary heart disease (CHD) and controls. Methods We explored PubMed, Embase, Web of Science, and Cochrane Library for studies comparing BDNF blood levels in patients with CHD and controls. Random-effect meta-analysis was conducted to calculate the standardized mean differences (SMD) and 95% confidence intervals (CI). The Newcastle-Ottawa scale was used to evaluate the quality of included articles, and statistical analyses were conducted using R version 4.0.4. Results The final analysis comprised 12 investigations covering 1422 CHD cases and 929 controls with mean ages of 59.66±13.56 and 53.78±13.61 years, respectively. The initial analyses revealed a tendency toward low levels of BDNF in the CHD group compared with the control group (SMD= -0.41; 95% CI, -1.12 to 0.30; P=0.26). After the removal of outliers, the difference achieved statistical difference (SMD= -0.56; 95% CI, -0.93 to -0.19; P<0.01). Subgroup analysis demonstrated no significant difference between serum and plasma BDNF levels (P=0.54); however, subgroup analyses of studies investigating plasma BDNF showed that patients with CHD had significantly lower BDNF levels. Conclusion Serum and plasma BDNF concentrations were considerably lower in patients with CHD than in healthy controls. Further studies of higher quality are required on the potential role of BDNF as a biomarker of CHD pathophysiology and severity.
Collapse
Affiliation(s)
- Parnian Shobeiri
- Children’s Medical Center Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity, Universal Scientific Education and Research Network, Tehran, Iran
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Hossein Behnoush
- Children’s Medical Center Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity, Universal Scientific Education and Research Network, Tehran, Iran
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirmohammad Khalaji
- Children’s Medical Center Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity, Universal Scientific Education and Research Network, Tehran, Iran
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Antonio Lucio Teixeira
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, TX, United States
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity, Universal Scientific Education and Research Network, Tehran, Iran
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Fioranelli M, Roccia MG, Przybylek B, Garo ML. The Role of Brain-Derived Neurotrophic Factor (BDNF) in Depression and Cardiovascular Disease: A Systematic Review. Life (Basel) 2023; 13:1967. [PMID: 37895349 PMCID: PMC10608007 DOI: 10.3390/life13101967] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/09/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Several studies have been conducted to prove the bidirectional relationship between cardiovascular disease (CVD) and depression. These two major illnesses share several common risk factors such that the development of either condition may increase the risk of the occurrence of the other. Brain-derived neurotrophic factor (BDNF) has been suggested as a reliable biomarker for depression and a strong predictor of CVD because it plays an important role in neuron survival and growth, serves as a neurotransmitter modulator, and promotes neuronal plasticity. The aim of this systematic review was to examine the bidirectional relationship between CVD and depression, focusing on the potential role of low serum BDNF levels in the development of either disease in the presence of the other. METHODS A systematic search strategy was developed using PRISMA guidelines. RESULTS Six studies (comprising 1251 patients) were identified, all of which examined the association between CVD and depression. CONCLUSIONS It was found that there may be a strong association between low serum BDNF levels and the risk of post-stroke depression. However, the studies on the role of altered serum BDNF levels and other types of CVD are few. Therefore, the inverse association between depression and CVD cannot be proven.
Collapse
Affiliation(s)
- Massimo Fioranelli
- Department of Human Sciences, Guglielmo Marconi University, Via Plinio 44, 00193 Rome, Italy; (M.F.); (M.G.R.)
| | - Maria Grazia Roccia
- Department of Human Sciences, Guglielmo Marconi University, Via Plinio 44, 00193 Rome, Italy; (M.F.); (M.G.R.)
| | - Bianca Przybylek
- Istituto Terapie Sistemiche Integrate, Casa di Cura Villa del Rosario, Via Flaminia 449, 00181 Rome, Italy;
| | - Maria Luisa Garo
- Istituto Terapie Sistemiche Integrate, Casa di Cura Sanatrix, Via di Trasone, 6, 00199 Rome, Italy
| |
Collapse
|
15
|
Çerçi B, Gök A, Akyol A. Brain-derived neurotrophic factor: Its role in energy balance and cancer cachexia. Cytokine Growth Factor Rev 2023; 71-72:105-116. [PMID: 37500391 DOI: 10.1016/j.cytogfr.2023.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 07/14/2023] [Accepted: 07/16/2023] [Indexed: 07/29/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) plays an important role in the development of the central and peripheral nervous system during embryogenesis. In the mature central nervous system, BDNF is required for the maintenance and enhancement of synaptic transmissions and the survival of neurons. Particularly, it is involved in the modulation of neurocircuits that control energy balance through food intake, energy expenditure, and locomotion. Regulation of BDNF in the central nervous system is complex and environmental factors affect its expression in murine models which may reflect to phenotype dramatically. Furthermore, BDNF and its high-affinity receptor tropomyosin receptor kinase B (TrkB), as well as pan-neurotrophin receptor (p75NTR) is expressed in peripheral tissues in adulthood and their signaling is associated with regulation of energy balance. BDNF/TrkB signaling is exploited by cancer cells as well and BDNF expression is increased in tumors. Intriguingly, previously demonstrated roles of BDNF in regulation of food intake, adipose tissue and muscle overlap with derangements observed in cancer cachexia. However, data about the involvement of BDNF in cachectic cancer patients and murine models are scarce and inconclusive. In the future, knock-in and/or knock-out experiments with murine cancer models could be helpful to explore potential new roles for BDNF in the development of cancer cachexia.
Collapse
Affiliation(s)
- Barış Çerçi
- Medical School, Hacettepe University, Ankara, Turkey.
| | - Ayşenur Gök
- Department of Stem Cell Sciences, Graduate School of Health Sciences, Hacettepe University, Ankara, Turkey; Hacettepe University Transgenic Animal Technologies Research and Application Center, Sıhhiye, Ankara 06100, Turkey
| | - Aytekin Akyol
- Departmant of Pathology, Medical School, Hacettepe University, Ankara, Turkey; Hacettepe University Transgenic Animal Technologies Research and Application Center, Sıhhiye, Ankara 06100, Turkey
| |
Collapse
|
16
|
Burovenko IY, Sonin DL, Borshcheva OV, Istomina MS, Zaitseva EA, Protsak ES, Borshchev VY, Borshchev YY, Galagudza MM. Influence of Interleukin-2 and Brain-Derived Neurotrophic Factor (BDNF) on the Size of Myocardial Infarction in Rats with Systemic Inflammatory Response. Bull Exp Biol Med 2023; 175:219-224. [PMID: 37464198 DOI: 10.1007/s10517-023-05838-7] [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: 10/24/2022] [Indexed: 07/20/2023]
Abstract
We studied the influence of recombinant IL-2 and brain-derived neurotrophic factor (BDNF) on the size of the myocardial necrosis zone of rats with systemic inflammatory response syndrome (SIRS). A significant increase in the necrosis zone and the levels of proinflammatory cytokines was revealed in animals with SIRS in comparison with the control. The administration of IL-2 to animals with SIRS significantly reduced the size of the necrosis zone, which was paralleled by a pronounced increase in IL-2 and BDNF in comparison with the corresponding parameters in rats with SIRS that did not receive IL-2. Administration of BDNF to animals with SIRS was followed by normalization of TNFα and IL-1α levels, but did not lead to a decrease in the size of the necrosis zone.
Collapse
Affiliation(s)
- I Yu Burovenko
- Institute of Experimental Medicine, V. A. Almazov National Medical Research Center, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - D L Sonin
- Institute of Experimental Medicine, V. A. Almazov National Medical Research Center, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - O V Borshcheva
- Institute of Experimental Medicine, V. A. Almazov National Medical Research Center, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - M S Istomina
- Institute of Experimental Medicine, V. A. Almazov National Medical Research Center, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - E A Zaitseva
- Institute of Experimental Medicine, V. A. Almazov National Medical Research Center, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - E S Protsak
- Institute of Experimental Medicine, V. A. Almazov National Medical Research Center, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - V Yu Borshchev
- Institute of Experimental Medicine, V. A. Almazov National Medical Research Center, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - Yu Yu Borshchev
- Institute of Experimental Medicine, V. A. Almazov National Medical Research Center, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - M M Galagudza
- Institute of Experimental Medicine, V. A. Almazov National Medical Research Center, Ministry of Health of the Russian Federation, St. Petersburg, Russia.
| |
Collapse
|
17
|
Cannavo A, Jun S, Rengo G, Marzano F, Agrimi J, Liccardo D, Elia A, Keceli G, Altobelli GG, Marcucci L, Megighian A, Gao E, Feng N, Kammers K, Ferrara N, Finos L, Koch WJ, Paolocci N. β3AR-Dependent Brain-Derived Neurotrophic Factor (BDNF) Generation Limits Chronic Postischemic Heart Failure. Circ Res 2023; 132:867-881. [PMID: 36884028 PMCID: PMC10281793 DOI: 10.1161/circresaha.122.321583] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 02/28/2023] [Indexed: 03/09/2023]
Abstract
BACKGROUND Loss of brain-derived neurotrophic factor (BDNF)/TrkB (tropomyosin kinase receptor B) signaling accounts for brain and cardiac disorders. In neurons, β-adrenergic receptor stimulation enhances local BDNF expression. It is unclear if this occurs in a pathophysiological relevant manner in the heart, especially in the β-adrenergic receptor-desensitized postischemic myocardium. Nor is it fully understood whether and how TrkB agonists counter chronic postischemic left ventricle (LV) decompensation, a significant unmet clinical milestone. METHODS We conducted in vitro studies using neonatal rat and adult murine cardiomyocytes, SH-SY5Y neuronal cells, and umbilical vein endothelial cells. We assessed myocardial ischemia (MI) impact in wild type, β3AR knockout, or myocyte-selective BDNF knockout (myoBDNF KO) mice in vivo (via coronary ligation [MI]) or in isolated hearts with global ischemia-reperfusion (I/R). RESULTS In wild type hearts, BDNF levels rose early after MI (<24 hours), plummeting at 4 weeks when LV dysfunction, adrenergic denervation, and impaired angiogenesis ensued. The TrkB agonist, LM22A-4, countered all these adverse effects. Compared with wild type, isolated myoBDNF KO hearts displayed worse infarct size/LV dysfunction after I/R injury and modest benefits from LM22A-4. In vitro, LM22A-4 promoted neurite outgrowth and neovascularization, boosting myocyte function, effects reproduced by 7,8-dihydroxyflavone, a chemically unrelated TrkB agonist. Superfusing myocytes with the β3AR-agonist, BRL-37344, increased myocyte BDNF content, while β3AR signaling underscored BDNF generation/protection in post-MI hearts. Accordingly, the β1AR blocker, metoprolol, via upregulated β3ARs, improved chronic post-MI LV dysfunction, enriching the myocardium with BDNF. Last, BRL-37344-imparted benefits were nearly abolished in isolated I/R injured myoBDNF KO hearts. CONCLUSIONS BDNF loss underscores chronic postischemic heart failure. TrkB agonists can improve ischemic LV dysfunction via replenished myocardial BDNF content. Direct cardiac β3AR stimulation, or β-blockers (via upregulated β3AR), is another BDNF-based means to fend off chronic postischemic heart failure.
Collapse
Affiliation(s)
- Alessandro Cannavo
- Department of Translational Medical Science, University of Naples Federico II, Italy
- Center For Translational Medicine LKSOM Temple University, Philadelphia, PA, U.S.A
| | - Seungho Jun
- Division of Cardiology, Johns Hopkins University Medical Institutions, Baltimore, MD, U.S.A
| | - Giuseppe Rengo
- Department of Translational Medical Science, University of Naples Federico II, Italy
- Istituti Clinici Scientifici Maugeri - Scientific Institute of Telese Terme (BN), Italy
| | - Federica Marzano
- Department of Translational Medical Science, University of Naples Federico II, Italy
- Center For Translational Medicine LKSOM Temple University, Philadelphia, PA, U.S.A
| | - Jacopo Agrimi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Daniela Liccardo
- Department of Translational Medical Science, University of Naples Federico II, Italy
- Center For Translational Medicine LKSOM Temple University, Philadelphia, PA, U.S.A
| | - Andrea Elia
- Department of Translational Medical Science, University of Naples Federico II, Italy
| | - Gizem Keceli
- Division of Cardiology, Johns Hopkins University Medical Institutions, Baltimore, MD, U.S.A
| | - Giovanna G. Altobelli
- Istituti Clinici Scientifici Maugeri - Scientific Institute of Telese Terme (BN), Italy
| | - Lorenzo Marcucci
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Aram Megighian
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Erhe Gao
- Center For Translational Medicine LKSOM Temple University, Philadelphia, PA, U.S.A
| | - Ning Feng
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, U.S.A
| | - Kai Kammers
- Quantitative Sciences Division – Department of Oncology, Johns Hopkins University School of Medicine, Padova, Italy
| | - Nicola Ferrara
- Department of Translational Medical Science, University of Naples Federico II, Italy
- Istituti Clinici Scientifici Maugeri - Scientific Institute of Telese Terme (BN), Italy
| | - Livio Finos
- Department of Statistical Science, University of Padova, Padova, Italy
| | - Walter J. Koch
- Center For Translational Medicine LKSOM Temple University, Philadelphia, PA, U.S.A
| | - Nazareno Paolocci
- Division of Cardiology, Johns Hopkins University Medical Institutions, Baltimore, MD, U.S.A
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| |
Collapse
|
18
|
Baumer Y, Pita M, Baez A, Ortiz-Whittingham L, Cintron M, Rose R, Gray V, Osei Baah F, Powell-Wiley T. By what molecular mechanisms do social determinants impact cardiometabolic risk? Clin Sci (Lond) 2023; 137:469-494. [PMID: 36960908 PMCID: PMC10039705 DOI: 10.1042/cs20220304] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 03/07/2023] [Accepted: 03/08/2023] [Indexed: 03/25/2023]
Abstract
While it is well known from numerous epidemiologic investigations that social determinants (socioeconomic, environmental, and psychosocial factors exposed to over the life-course) can dramatically impact cardiovascular health, the molecular mechanisms by which social determinants lead to poor cardiometabolic outcomes are not well understood. This review comprehensively summarizes a variety of current topics surrounding the biological effects of adverse social determinants (i.e., the biology of adversity), linking translational and laboratory studies with epidemiologic findings. With a strong focus on the biological effects of chronic stress, we highlight an array of studies on molecular and immunological signaling in the context of social determinants of health (SDoH). The main topics covered include biomarkers of sympathetic nervous system and hypothalamic-pituitary-adrenal axis activation, and the role of inflammation in the biology of adversity focusing on glucocorticoid resistance and key inflammatory cytokines linked to psychosocial and environmental stressors (PSES). We then further discuss the effect of SDoH on immune cell distribution and characterization by subset, receptor expression, and function. Lastly, we describe epigenetic regulation of the chronic stress response and effects of SDoH on telomere length and aging. Ultimately, we highlight critical knowledge gaps for future research as we strive to develop more targeted interventions that account for SDoH to improve cardiometabolic health for at-risk, vulnerable populations.
Collapse
Affiliation(s)
- Yvonne Baumer
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, Cardiovascular Branch, Division of Intramural Research, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, U.S.A
| | - Mario A. Pita
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, Cardiovascular Branch, Division of Intramural Research, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, U.S.A
| | - Andrew S. Baez
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, Cardiovascular Branch, Division of Intramural Research, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, U.S.A
| | - Lola R. Ortiz-Whittingham
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, Cardiovascular Branch, Division of Intramural Research, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, U.S.A
| | - Manuel A. Cintron
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, Cardiovascular Branch, Division of Intramural Research, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, U.S.A
| | - Rebecca R. Rose
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, Cardiovascular Branch, Division of Intramural Research, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, U.S.A
| | - Veronica C. Gray
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, Cardiovascular Branch, Division of Intramural Research, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, U.S.A
| | - Foster Osei Baah
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, Cardiovascular Branch, Division of Intramural Research, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, U.S.A
| | - Tiffany M. Powell-Wiley
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, Cardiovascular Branch, Division of Intramural Research, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, U.S.A
- Intramural Research Program, National Institute on Minority Health and Health Disparities, National Institutes of Health, Bethesda, MD, U.S.A
| |
Collapse
|
19
|
Kennedy KG, Mio M, Goldstein BI, Brambilla P, Delvecchio G. Systematic review and meta-analysis of retinal microvascular caliber in bipolar disorder, major depressive disorder, and schizophrenia. J Affect Disord 2023; 331:342-351. [PMID: 36958491 DOI: 10.1016/j.jad.2023.03.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/06/2023] [Accepted: 03/09/2023] [Indexed: 03/25/2023]
Abstract
BACKGROUND Individuals with a severe mental illness (SMI), such as bipolar disorder (BD), major depressive disorder (MDD), and schizophrenia (SZ), have increased rates of cardiovascular and cerebrovascular disease. Interestingly, it has been reported that retinal microvessels, a proxy cerebrovascular measure, non-invasively assessed via retinal imaging, predict future cardiovascular disease, with some studies also showing anomalous retinal microvascular caliber in SMI. Therefore, this review and meta-analysis evaluated whether retinal microvascular caliber differs between individuals with SMI vs controls and summarized current findings. METHODS A systematic literature search for retinal microvascular caliber and SMI was conducted in Embase and MEDLINE. Studies needed to be published in English before 2022 December 1st and examine retinal microvascular caliber in individuals diagnosed with a SMI. Finally, a meta-analysis of arteriolar and venular caliber in SMI case-controlled studies was also conducted. RESULTS The search yielded 65 unique articles, 11 were included in the review and 6 in the meta-analysis. The meta-analysis found that the SMI group had significantly wider venules than controls (SMD = -0.53; 95 % CI = 0.24, 0.81; p = 0.0004) but not arterioles (SMD = 0.07; 95 % CI = -0.29, 0.44; p = 0.70). Additionally, the systematic review found that poorer retinal microvascular health is associated with greater illness severity. LIMITATIONS Large heterogeneity of findings and small sample size. CONCLUSION This systematic review and meta-analysis found that SMI, specifically SZ, is associated with wider retinal venules. Retinal imaging, a fast, cost-effective, and non-invasive assay of cerebrovascular health, may provide insight into the pathophysiological processes of SMI. However, future longitudinal studies investigating these findings are warranted.
Collapse
Affiliation(s)
- Kody G Kennedy
- Centre for Youth Bipolar Disorder, Centre for Addictions and Mental Health, Toronto, Canada; Department of Pharmacology, University of Toronto, Toronto, Canada
| | - Megan Mio
- Centre for Youth Bipolar Disorder, Centre for Addictions and Mental Health, Toronto, Canada; Department of Pharmacology, University of Toronto, Toronto, Canada
| | - Benjamin I Goldstein
- Centre for Youth Bipolar Disorder, Centre for Addictions and Mental Health, Toronto, Canada; Department of Pharmacology, University of Toronto, Toronto, Canada
| | - Paolo Brambilla
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Giuseppe Delvecchio
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| |
Collapse
|
20
|
Schäfer K, Tello K, Pak O, Richter M, Gierhardt M, Kwapiszewska G, Veith C, Fink L, Gall H, Hecker M, Kojonazarov B, Kraut S, Lo K, Wilhelm J, Grimminger F, Seeger W, Schermuly RT, Ghofrani HA, Zahner D, Gerstberger R, Weissmann N, Sydykov A, Sommer N. Decreased plasma levels of the brain-derived neurotrophic factor correlate with right heart congestion in pulmonary arterial hypertension. ERJ Open Res 2023; 9:00230-2022. [PMID: 36891080 PMCID: PMC9986749 DOI: 10.1183/23120541.00230-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 09/28/2022] [Indexed: 01/27/2023] Open
Abstract
Background The brain-derived neurotrophic factor (BDNF) may promote development of pulmonary hypertension and right ventricular (RV) failure. However, BDNF plasma levels were decreased in patients with left ventricular (LV) failure. Therefore, we investigated BDNF plasma levels in pulmonary hypertension patients and the role of BDNF in mouse models of pulmonary hypertension and isolated RV failure. Methods BDNF plasma levels were correlated to pulmonary hypertension in two patient cohorts, including either post- and pre-capillary pulmonary hypertension patients (first cohort) or only pre-capillary pulmonary hypertension patients (second cohort). In the second cohort, RV dimensions and load-independent function were determined by imaging and pressure-volume catheter measurements, respectively. For induction of isolated RV pressure overload, heterozygous Bdnf knockout (Bdnf+/- ) mice were subjected to pulmonary arterial banding (PAB). For induction of pulmonary hypertension, mice with inducible knockout of BDNF in smooth muscle cells (Bdnf/Smmhc knockout) were exposed to chronic hypoxia. Results Plasma BDNF levels were decreased in patients with pulmonary hypertension. Following adjustment for covariables, BDNF levels negatively correlated in both cohorts with central venous pressure. In the second cohort, BDNF levels additionally negatively correlated with RV dilatation. In animal models, BDNF downregulation attenuated RV dilatation in Bdnf+ /- mice after PAB or hypoxic Bdnf/Smmhc knockout mice, although they developed pulmonary hypertension to a similar extent. Conclusions Similar to LV failure, circulating levels of BDNF were decreased in pulmonary hypertension patients, and low BDNF levels were associated with right heart congestion. Decreased BDNF levels did not worsen RV dilatation in animal models, and thus, may be the consequence, but not the cause of RV dilatation.
Collapse
Affiliation(s)
- Katharina Schäfer
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Excellence Cluster Cardio-Pulmonary Institute (CPI), Justus-Liebig University, Giessen, Germany.,These authors contributed equally
| | - Khodr Tello
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Excellence Cluster Cardio-Pulmonary Institute (CPI), Justus-Liebig University, Giessen, Germany.,These authors contributed equally
| | - Oleg Pak
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Excellence Cluster Cardio-Pulmonary Institute (CPI), Justus-Liebig University, Giessen, Germany.,These authors contributed equally
| | - Manuel Richter
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Excellence Cluster Cardio-Pulmonary Institute (CPI), Justus-Liebig University, Giessen, Germany
| | - Mareike Gierhardt
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Excellence Cluster Cardio-Pulmonary Institute (CPI), Justus-Liebig University, Giessen, Germany.,Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular Research, Otto Loewi research Center, Medical University Graz, Graz, Austria.,Institute for Lung Health (ILH), Giessen, Germany
| | - Christine Veith
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Excellence Cluster Cardio-Pulmonary Institute (CPI), Justus-Liebig University, Giessen, Germany
| | - Ludger Fink
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Excellence Cluster Cardio-Pulmonary Institute (CPI), Justus-Liebig University, Giessen, Germany
| | - Henning Gall
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Excellence Cluster Cardio-Pulmonary Institute (CPI), Justus-Liebig University, Giessen, Germany
| | - Matthias Hecker
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Excellence Cluster Cardio-Pulmonary Institute (CPI), Justus-Liebig University, Giessen, Germany
| | | | - Simone Kraut
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Excellence Cluster Cardio-Pulmonary Institute (CPI), Justus-Liebig University, Giessen, Germany
| | - Kevin Lo
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Excellence Cluster Cardio-Pulmonary Institute (CPI), Justus-Liebig University, Giessen, Germany
| | - Jochen Wilhelm
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Excellence Cluster Cardio-Pulmonary Institute (CPI), Justus-Liebig University, Giessen, Germany.,Institute for Lung Health (ILH), Giessen, Germany
| | - Friedrich Grimminger
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Excellence Cluster Cardio-Pulmonary Institute (CPI), Justus-Liebig University, Giessen, Germany
| | - Werner Seeger
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Excellence Cluster Cardio-Pulmonary Institute (CPI), Justus-Liebig University, Giessen, Germany.,Institute for Lung Health (ILH), Giessen, Germany
| | - Ralph T Schermuly
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Excellence Cluster Cardio-Pulmonary Institute (CPI), Justus-Liebig University, Giessen, Germany
| | - Hossein A Ghofrani
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Excellence Cluster Cardio-Pulmonary Institute (CPI), Justus-Liebig University, Giessen, Germany.,Department of Medicine, Imperial College London, London, UK
| | - Daniel Zahner
- Central Laboratory Animal Facility, Justus-Liebig University, Giessen, Germany
| | - Rüdiger Gerstberger
- Institute of Veterinary Physiology and Biochemistry, Justus-Liebig University, Giessen, Germany
| | - Norbert Weissmann
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Excellence Cluster Cardio-Pulmonary Institute (CPI), Justus-Liebig University, Giessen, Germany
| | - Akylbek Sydykov
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Excellence Cluster Cardio-Pulmonary Institute (CPI), Justus-Liebig University, Giessen, Germany.,These authors contributed equally
| | - Natascha Sommer
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Excellence Cluster Cardio-Pulmonary Institute (CPI), Justus-Liebig University, Giessen, Germany.,These authors contributed equally
| |
Collapse
|
21
|
Fatma R, Chauhan W, Shahi MH, Afzal M. Association of BDNF gene missense polymorphism rs6265 (Val66Met) with three quantitative traits, namely, intelligence quotient, body mass index, and blood pressure: A genetic association analysis from North India. Front Neurol 2023; 13:1035885. [PMID: 36742047 PMCID: PMC9894895 DOI: 10.3389/fneur.2022.1035885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 12/28/2022] [Indexed: 01/21/2023] Open
Abstract
Background Brain-derived neurotrophic factor (BDNF), a neurotransmitter modulator, plays a significant role in neuronal survival and growth and participates in neuronal plasticity, thus being essential for learning, memory, and the development of cognition. Additionally, it is crucial for appetite, weight, and metabolic control and plays a pivotal role in the cardiovascular system. The Val66Met polymorphism (rs6265) of the BDNF gene causes a decrease in BDNF secretion and plays a role in impairments in cognition, energy homeostasis, and cardiovascular events. The present study aimed to evaluate the association of polymorphism (rs6265) of the BDNF gene with three quantitative traits simultaneously, namely, intelligence quotient (IQ), body mass index (BMI), and blood pressure (BP). Methods Psychometric, morphometric, and physiometric data of the total participants (N = 246) were collected. WASI-IIINDIA was used to measure cognitive ability. Genotyping was carried out using allele-specific PCR for the rs6265 polymorphism (C196T), and genotypes were determined. Statistical analyses were performed at p < 0.05 significance level using MS-Excel and SigmaPlot. The odds ratio models with a 95% confidence interval were used to test the associations. The used models are co-dominant, recessive, dominant, over-dominant, and additive. Results The allelic frequencies of alleles C and T were 72 and 28%, respectively. Under the dominant genetic model, a significant susceptible association of minor allele T was observed with a lower average verbal comprehensive index (OR = 2.216, p = 0.003, CI (95%) =1.33-3.69), a lower average performance reasoning index (OR = 2.634, p < 0.001, CI (95%) = 1.573-4.41), and a lower average full-scale IQ-4 (OR = 3.159, p < 0.001, CI (95%) = 1.873-5.328). Carriers of Met-alleles were found to have an increased body mass index (OR = 2.538, p < 0.001, CI (95%) = 1.507-4.275), decreased systolic blood pressure (OR = 2.051, p = 0.012, CI (95%) = 1.202-3.502), and decreased diastolic blood pressure (OR = 2.162, p = 0.006, CI (95%) = 1.278-3.657). Under the recessive genetic model, several folds decrease in IQ and BP and an increase in BMI with the presence of the T allele was also detected. Conclusion This novel study may improve our understanding of genetic alterations to the traits and hence be helpful for clinicians and researchers to investigate the diagnostic and prognostic value of this neurotrophic factor.
Collapse
Affiliation(s)
- Rafat Fatma
- Human Genetics and Toxicology Laboratory, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh, India
| | - Waseem Chauhan
- Human Genetics and Toxicology Laboratory, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh, India
| | - Mehdi Hayat Shahi
- Interdisciplinary Brain Research Centre, Aligarh Muslim University, Aligarh, India
| | - Mohammad Afzal
- Human Genetics and Toxicology Laboratory, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh, India,*Correspondence: Mohammad Afzal ✉ ; ✉
| |
Collapse
|
22
|
Choi W, Kang HJ, Kim JW, Kim HK, Kang HC, Kim SW, Kim JC, Ahn Y, Jeong MH, Kim JM. Modifying effect of the serum level of brain-derived neurotrophic factor (BDNF) on the association between BDNF methylation and long-term cardiovascular outcomes in patients with acute coronary syndrome. Front Cardiovasc Med 2023; 9:1084834. [PMID: 36741831 PMCID: PMC9889833 DOI: 10.3389/fcvm.2022.1084834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/30/2022] [Indexed: 01/19/2023] Open
Abstract
Introduction This study investigated the potential modifying effects of the serum brain-derived neurotrophic factor (sBDNF) level on the association between BDNF methylation status and long-term cardiovascular outcomes in acute coronary syndrome (ACS) patients. Methods From 2006 to 2012, hospitalized ACS patients were consecutively recruited. The sBDNF level and BDNF methylation status were assessed at baseline in 969 patients who were followed up for major adverse cardiac events (MACEs) over 5-12 years, until 2017 or death. Cox proportional hazards models were utilized to compare the time to first composite or individual MACEs between individuals with lower and those with higher average BDNF methylation levels in the low and high sBDNF groups, respectively. The modifying effects of the sBDNF and average BDNF methylation levels on first composite and individual MACEs were analyzed using Cox proportional hazards models after adjusting for potential covariates. Results In the low sBDNF group, a higher average BDNF methylation level was linked to an increase in composite MACEs independent of confounding variables, but not in the high sBDNF group [HR (95 percent CI) = 1.04 (0.76-1.44)]. The interaction effect between the sBDNF and average BDNF methylation levels on composite MACEs was significant after adjusting for covariates (P = 0.008). Conclusion Combining the BDNF methylation status and sBDNF levels may help identify ACS patients who are likely to have unfavorable clinical outcomes.
Collapse
Affiliation(s)
- Wonsuk Choi
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Republic of Korea
| | - Hee-Ju Kang
- Department of Psychiatry, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Ju-Wan Kim
- Department of Psychiatry, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Hee Kyung Kim
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Republic of Korea
| | - Ho-Cheol Kang
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Republic of Korea
| | - Sung-Wan Kim
- Department of Psychiatry, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Jung-Chul Kim
- Department of Surgery, Chonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Youngkeun Ahn
- Department of Cardiology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Myung Ho Jeong
- Department of Cardiology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Jae-Min Kim
- Department of Psychiatry, Chonnam National University Medical School, Gwangju, Republic of Korea,*Correspondence: Jae-Min Kim ✉
| |
Collapse
|
23
|
Gabryelska A, Turkiewicz S, Ditmer M, Sochal M. Neurotrophins in the Neuropathophysiology, Course, and Complications of Obstructive Sleep Apnea-A Narrative Review. Int J Mol Sci 2023; 24:1808. [PMID: 36768132 PMCID: PMC9916304 DOI: 10.3390/ijms24031808] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/04/2022] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Obstructive sleep apnea (OSA) is a disorder characterized by chronic intermittent hypoxia and sleep fragmentation due to recurring airway collapse during sleep. It is highly prevalent in modern societies, and due to its pleiotropic influence on the organism and numerous sequelae, it burdens patients and physicians. Neurotrophins (NTs), proteins that modulate the functioning and development of the central nervous system, such as brain-derived neurotrophic factor (BDNF), have been associated with OSA, primarily due to their probable involvement in offsetting the decline in cognitive functions which accompanies OSA. However, NTs influence multiple aspects of biological functioning, such as immunity. Thus, extensive evaluation of their role in OSA might enlighten the mechanism behind some of its elusive features, such as the increased risk of developing an immune-mediated disease or the association of OSA with cardiovascular diseases. In this review, we examine the interactions between NTs and OSA and discuss their contribution to OSA pathophysiology, complications, as well as comorbidities.
Collapse
Affiliation(s)
- Agata Gabryelska
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 90-419 Lodz, Poland
| | | | | | - Marcin Sochal
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 90-419 Lodz, Poland
| |
Collapse
|
24
|
Galindo C, Nguyen VT, Hill B, Sims N, Heck A, Negron M, Lusk C. Brain-derived neurotrophic factor rs6265 (Val66Met) single nucleotide polymorphism as a master modifier of human pathophysiology. Neural Regen Res 2023. [PMID: 35799516 PMCID: PMC9241394 DOI: 10.4103/1673-5374.343894] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Brain-derived neurotrophic factor is the most prevalent member of the nerve growth factor family. Since its discovery in 1978, this enigmatic molecule has spawned more than 27,000 publications, most of which are focused on neurological disorders. Brain-derived neurotrophic factor is indispensable during embryogenesis and postnatally for the normal development and function of both the central and peripheral nervous systems. It is becoming increasingly clear, however, that brain-derived neurotrophic factor likewise plays crucial roles in a variety of other biological functions independently of sympathetic or parasympathetic involvement. Brain-derived neurotrophic factor is also increasingly recognized as a sophisticated environmental sensor and master coordinator of whole organismal physiology. To that point, we recently found that a common nonsynonymous (Val66→Met) single nucleotide polymorphism in the brain-derived neurotrophic factor gene (rs6265) not only substantially alters basal cardiac transcriptomics in mice but subtly influences heart gene expression and function differentially in males and females. In addition to a short description of recent results from associative neuropsychiatric studies, this review provides an eclectic assortment of research reports that support a modulatory role for rs6265 including and beyond the central nervous system.
Collapse
|
25
|
Haba MȘC, Tudorancea I, Mihai CT, Onofrei V, Costache II, Petriș AO, Șorodoc L. Brain-Derived Neurotrophic Factor Expression in Patients with Acute Pulmonary Embolism Compared to the General Population: Diagnostic and Prognostic Implications. J Clin Med 2022; 11:jcm11174948. [PMID: 36078878 PMCID: PMC9456489 DOI: 10.3390/jcm11174948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/15/2022] [Accepted: 08/19/2022] [Indexed: 11/28/2022] Open
Abstract
(1) Background: Pulmonary embolism (PE) is a severe condition, representing the third most important cardiovascular cause of death after myocardial infarction and stroke. Despite the use of clinical pre-test probability scores, D-dimer measuring, and computer tomography pulmonary angiography (CTPA), PE diagnosis remains a challenge. Brain-derived neurotrophic factor (BDNF) is the most important member of the neurotrophin family, which has also been shown to be involved in the physiopathology of cardiovascular conditions such as heart failure and myocardial infarction. In this study, we aimed to assess the BDNF expression in patients with acute PE compared to the general population, and to also investigate its diagnostic and prognostic role. (2) Methods: We conducted a single center prospective study, which included 90 patients with PE and 55 healthy volunteers. Clinical and paraclinical parameters, together with plasma levels of BDNF, were evaluated in all patients after admission. (3) Results: The plasma levels of BDNF were significantly lower in the PE patients compared with the control group (403 vs. 644 pg/mL, p < 0.001). ROC analysis revealed an AUC of 0.806 (95% CI 0.738−0.876, p < 0.001) and a cut-off value of 564 pg/mL, which associated a sensitivity of 74.4% and a specificity of 78.2% for PE. Low BDNF levels also correlated with prognostic markers of PE, such as PESI score (p = 0.023), NT-proBNP (p < 0.01), right ventricular diameter (p = 0.029), and tricuspid annular plane systolic elevation (p = 0.016). Moreover, we identified a decreased BDNF expression in patients with high-risk PE (p < 0.01), thrombolytic treatment (p = 0.01), and patients who died within 30 days (p = 0.05). (4) Conclusions: Our study revealed that plasma BNDF is significantly lower in patients with PE when compared with the general population, and may be considered as a promising biomarker in complementing the current diagnostic tools for PE. Furthermore, low levels of BDNF might also be used to predict a poor outcome of this condition.
Collapse
Affiliation(s)
- Mihai Ștefan Cristian Haba
- Department of Internal Medicine I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
| | - Ionuț Tudorancea
- Department of Morpho-Functional Sciences II-Physiology, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
- Correspondence: ; Tel.: +40-232-301-603
| | - Cosmin Teodor Mihai
- Advanced Research and Development Center for Experimental Medicine (CEMEX), University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
| | - Viviana Onofrei
- Department of Internal Medicine I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
| | - Irina Iuliana Costache
- Department of Internal Medicine I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
| | - Antoniu Octavian Petriș
- Department of Internal Medicine I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
| | - Laurențiu Șorodoc
- Department of Internal Medicine III, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
| |
Collapse
|
26
|
Maroofi A, Moro T, Agrimi J, Safari F. Cognitive decline in heart failure: Biomolecular mechanisms and benefits of exercise. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166511. [PMID: 35932891 DOI: 10.1016/j.bbadis.2022.166511] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/18/2022] [Accepted: 07/29/2022] [Indexed: 11/24/2022]
Abstract
By definition, heart failure (HF) is a human pathological condition affecting the structure and function of all organs in the body, and the brain is not an exception to that. Failure of the heart to pump enough blood centrally and peripherally is at the foundation of HF patients' inability to attend even the most ordinary daily activities and progressive deterioration of their cognitive capacity. What is more, between heart and brain exists a bidirectional relationship that goes well beyond hemodynamics and concerns bioelectric and endocrine signaling. This increasingly consolidated evidence makes the scenario even more complex. Studies have mainly chased how HF impairs cognition without focusing much on preventive measures, notably cardio-cerebral health proxies. Here, we aim to provide a brief account of known and hypothetical factors that may explain how exercise can help obviate cognitive dysfunction associated with HF in its different forms. As we shall see, there is a stringent need for a deeper grasp of such mechanisms. Indeed, gaining this new knowledge will automatically shed new light on the inner workings of HF itself, thus resulting in more effective prevention and treatment of this escalating syndrome.
Collapse
Affiliation(s)
- Abdulbaset Maroofi
- Department of Exercise Physiology, Faculty of Physical Education & Sport Sciences, University of Guilan, Rasht, Iran
| | - Tatiana Moro
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
| | - Jacopo Agrimi
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy.
| | - Fatemeh Safari
- Department of Physiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
27
|
Li X, Yuan RR, Wang Q, Chai S, Zhang Z, Wang Y, Huang SH. Brain-derived neurotrophic factor regulates LYN kinase-mediated myosin light chain kinase activation to modulate nonmuscle myosin II activity in hippocampal neurons. J Biol Chem 2022; 298:102054. [PMID: 35598826 PMCID: PMC9194867 DOI: 10.1016/j.jbc.2022.102054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 12/14/2022] Open
Abstract
Myosins belong to a large superfamily of actin-dependent molecular motors. Nonmuscle myosin II (NM II) is involved in the morphology and function of neurons, but little is known about how NM II activity is regulated. Brain-derived neurotrophic factor (BDNF) is a prevalent neurotrophic factor in the brain that encourages growth and differentiation of neurons and synapses. In this study, we report that BDNF upregulates the phosphorylation of myosin regulatory light chain (MLC2), to increases the activity of NM II. The role of BDNF on modulating the phosphorylation of MLC2 was validated by using Western blotting in primary cultured hippocampal neurons. This result was confirmed by injecting BDNF into the dorsal hippocampus of mice and detecting the phosphorylation level of MLC2 by Western blotting. We further perform coimmunoprecipitation assay to confirm that this process depends on the activation of the LYN kinase through binding with tyrosine kinase receptor B, the receptor of BDNF, in a kinase activity-dependent manner. LYN kinase subsequently phosphorylates MLCK, further promoting the phosphorylation of MLC2. Taken together, our results suggest a new molecular mechanism by which BDNF regulates MLC2 activity, which provides a new perspective for further understanding the functional regulation of NM II in the nervous system.
Collapse
Affiliation(s)
- Xiaobing Li
- Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Rong-Rong Yuan
- Institute of Basic Medicine, Shandong University, Jinan, Shandong, China
| | - Qixia Wang
- Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Shouyu Chai
- Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Zhengying Zhang
- Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yue Wang
- Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Shu-Hong Huang
- Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
28
|
Maroofi A, Bagheri Rouch A, Naderi N, Damirchi A. Effects of two different exercise paradigms on cardiac function, BDNF-TrkB expression, and myocardial protection in the presence and absence of Western diet. IJC HEART & VASCULATURE 2022; 40:101022. [PMID: 35399608 PMCID: PMC8991101 DOI: 10.1016/j.ijcha.2022.101022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/17/2022] [Accepted: 03/30/2022] [Indexed: 12/11/2022]
Abstract
Background Brain-derived neurotrophic factor (BDNF) -tropomyosin-related kinase receptor B (TrkB) signaling is a vital regulator of myocardial performance. Here, we tested the impact of high-intensity interval training (HIIT) and moderate-intensity continuous training (MICT) on heart function, metabolic parameters, and serum/cardiac BDNF (with its TrkB receptor) in animals fed a Western (WD) or regular diet (ND). Further, myocardial expression of pro-inflammatory cytokine interleukin-18 (IL-18) and cardioprotective molecule heme oxygens-1 (HO-1) were monitored. Methods Wistar rats were divided into HIIT, MICT, and sedentary (SED), all fed a WD or ND, for 12 weeks. Heart function, protein expression, and serum factors were assessed via echocardiography, western blotting, and ELISA, respectively. Results WD plus SED caused insulin resistance, dyslipidemia, visceral fat deposition, serum BDNF depletion as well as cardiac upregulation of IL-18 and downregulation of HO-1, without affecting, heart function and BDNF-TrkB expression. The cardiometabolic risk factors, serum BDNF losses, and IL-18 overexpression were similarly obviated by HIIT and MICT, although HO-1 expression was boosted by HIIT exclusively (even in ND). HIIT enhanced heart function, regardless of the diet. HIIT augmented cardiac BDNF expression, with a significant difference between ND and WD. Likewise, HIIT instigated TrkB expression only in ND. Conclusions HIIT and MICT can cope with myocardial inflammation and cardiometabolic risk factors in WD consumers and, exclusively, HIIT may grant further protection by increasing heart function, BDNF-TrkB expression, and HO-1 expression. Thus, the HIIT paradigm should be considered as a preference for subjects who require heart function to be preserved or enhanced.
Collapse
Affiliation(s)
- Abdulbaset Maroofi
- Department of Exercise Physiology, Faculty of Physical Education & Sport Sciences, University of Guilan, Rasht, Iran
| | - Ahmadreza Bagheri Rouch
- Department of Exercise Physiology, Faculty of Physical Education & Sport Sciences, University of Guilan, Rasht, Iran
| | - Nasim Naderi
- Rajaie Cardiovascular, Medical & Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Arsalan Damirchi
- Department of Exercise Physiology, Faculty of Physical Education & Sport Sciences, University of Guilan, Rasht, Iran
| |
Collapse
|
29
|
Alqudah M, Khanfar M, Alfaqih M, Al‑Shboul O, Al‑U'datt D, Al‑Dwairi A, Allouh M. Correlation between vitamin D and serum brain derived neurotropic factor levels in type 2 diabetes mellitus patients. Biomed Rep 2022; 16:54. [PMID: 35620310 PMCID: PMC9112377 DOI: 10.3892/br.2022.1537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/19/2022] [Indexed: 12/15/2022] Open
Abstract
Diabetes Mellitus (DM) currently ranks as the most common endocrine disorder worldwide. Current opinion views DM as a group of heterogeneous metabolic diseases characterized by hyperglycemia triggered by defects in the ability of the body to produce or use insulin in type 1 and 2 DM, respectively. Brain-derived neurotrophic factor (BDNF), one of the neurotrophin family of growth factors, has been linked to the pathogenesis of DM and insulin resistance. Moreover, vitamin D has been associated with insulin resistance and DM. Recently, the interactions between vitamin D and BDNF have been investigated in diabetic rats. However, this correlation has never been investigated in humans. Thus, the aim of the present study was to assess the alterations in serum BDNF and vitamin D levels in T2DM patients in Jordan, prior to and following vitamin D supplementation. A combination of non-experimental case-control and experimental designed studies were utilized to assess the relationship between serum BDNF and vitamin D levels in T2DM patients. The levels of BDNF and vitamin D were measured using commercially available ELISA kits, and fasting blood glucose (FBG) and HbA1c levels were measured in medical labs. The results showed that diabetic patients had lower levels of serum vitamin D and higher levels of BDNF compared with the healthy controls. Moreover, linear regression analysis indicated that BDNF levels were inversely correlated with serum vitamin D levels. Furthermore, vitamin D supplementation significantly increased vitamin D serum levels and decreased BDNF serum levels in diabetic patients. Intriguingly, FBG and HbA1c levels were significantly improved post vitamin D supplementation. These data demonstrate a positive effect of vitamin D supplementation in diabetic patients suggesting the implementation of vitamin D as part of future T2DM treatment plans. However, additional studies are needed to investigate the direct link between vitamin D, BDNF, and T2DM.
Collapse
Affiliation(s)
- Mohammad Alqudah
- Department of Physiology and Biochemistry, School of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Mariam Khanfar
- Department of Physiology and Biochemistry, School of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Mahmoud Alfaqih
- Department of Physiology and Biochemistry, School of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Othman Al‑Shboul
- Department of Physiology and Biochemistry, School of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Doa'a Al‑U'datt
- Department of Physiology and Biochemistry, School of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Ahmed Al‑Dwairi
- Department of Physiology and Biochemistry, School of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Mohammed Allouh
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 15551, United Arab Emirates
| |
Collapse
|
30
|
Lian H, Zhong XS, Xiao Y, Sun Z, Shen Y, Zhao K, Ma X, Li Y, Niu Q, Liu M, Powell DW, Liu C, Li Q. Exosomal miR-29b of Gut Origin in Patients With Ulcerative Colitis Suppresses Heart Brain-Derived Neurotrophic Factor. Front Mol Biosci 2022; 9:759689. [PMID: 35274002 PMCID: PMC8902158 DOI: 10.3389/fmolb.2022.759689] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 01/21/2022] [Indexed: 01/13/2023] Open
Abstract
Background and Aims: While the interplay between heart and gut in inflammatory bowel disease (IBD) has previously been noted, how the inflamed gut impairs heart function remain elusive. We hypothesized that exosomal miRNAs of gut origin induce cardiac remodeling in IBD. Our aim was to identify plasma exosomal miRNAs that not only are of diagnostic value but also contribute to cardiac remodeling in patients with ulcerative colitis (UC).Methods: Plasma exosomes were isolated from UC patients and healthy control subjects and exosomal miRNAs were profiled by next-generation sequencing. Exosomal miR-29b levels in CCD841 CoN colon epithelial cells were detected by RT-qPCR. Exosomes packaged with miR-29b were incubated with H9c2 cells or administered to live mice.Results: The plasma exosomal miRNA profiles of the UC patients were significantly different from that of the controls and 20 miRNAs including miR-29b were differentially expressed. In CCD841 CoN cells, TNFα, IL-1β, and H2O2 significantly elevated miR-29b in both the cells and their secreted exosomes (p < 0.01), suggesting that intestinal epithelium secrets exosomes rich in miR-29b in IBD. In H9c2 myoblast cells, miR-29b modulated multiple genes including brain-derived neurotrophic factor (BDNF). Epithelial cell-derived exosomes packaged with miR-29b also attenuated BDNF and increased cleaved caspase 3, suggestive of apoptosis. Furthermore, tail vein injection of engineered exosomes with high levels of miR-29b suppressed BDNF and augmented cleaved caspase 3 in the heart of adult mouse (p < 0.01).Conclusion: Plasma exosomal miRNA profile could be a novel diagnostic approach for IBD. Excessive plasma exosomal miR-29b suppresses critical proteins like BDNF in IBD, leading to cardiac impairment.
Collapse
Affiliation(s)
- Haifeng Lian
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, China
- *Correspondence: Haifeng Lian, ; Qingjie Li,
| | - Xiaoying S. Zhong
- Division of Gastroenterology, Department of Internal Medicine, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Ying Xiao
- Division of Gastroenterology, Department of Internal Medicine, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhe Sun
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, China
| | - Yuanyuan Shen
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, China
| | - Kaile Zhao
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, China
| | - Xingbin Ma
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, China
| | - Yanmin Li
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, China
| | - Qiong Niu
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, China
| | - Max Liu
- Division of Gastroenterology, Department of Internal Medicine, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Don W. Powell
- Division of Gastroenterology, Department of Internal Medicine, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Chengxia Liu
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, China
| | - Qingjie Li
- Division of Gastroenterology, Department of Internal Medicine, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
- *Correspondence: Haifeng Lian, ; Qingjie Li,
| |
Collapse
|
31
|
Roysommuti S, Wyss JM. Brain-Derived Neurotrophic Factor Potentiates Entorhinal-Dentate but not Hippocampus CA1 Pathway in Adult Male Rats: A Mechanism of Taurine-Modulated BDNF on Learning and Memory. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1370:369-379. [PMID: 35882811 PMCID: PMC9467516 DOI: 10.1007/978-3-030-93337-1_35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Taurine plays an important role in neural growth and function from early to adult life, particularly in learning and memory via BDNF action. This study tested the hypothesis that BDNF differentially potentiates entorhinal-hippocampal synaptic transmission in vivo in adult rats. In anesthetized male Sprague-Dawley rats, a stainless steel recording electrode with an attached microinjector was placed into CA1 and the dentate gyrus to record fEPSP, and a paired stainless steel electrode was inserted into entorhinal cortex for continuous paired-pulse stimulation of that brain region. In the dentate gyrus, microinjection of BDNF resulted in a gradual increase in the peak slope of the fEPSP. Following the infusion, the peak fEPSP began to rise in about 8 min, reached a maximum of 120 ± 2% (from baseline) by about 20 min, and remained near peak elevation (~115%) for more than 30 min. In contrast, the same dose of BDNF when injected into CA1 had no consistent effect on fEPSP slopes in the CA1. Further, an equimolar cytochrome C (horse heart) infusion had no significant effect on fEPSP slopes in either the dentate gyrus or CA1. The potentiation effect of BDNF in the dentate gyrus is consistent with a significant increase in power spectral density of dentate gyrus field potentials at 70-200 Hz, but not at frequencies below 70 Hz. In addition, the CA1 power spectral density was not affected by BDNF (compared to cytochrome C). These data indicate that in vivo BDNF potentiates entorhinal-hippocampal synaptic transmission in dentate gyrus, but not in CA1.
Collapse
Affiliation(s)
- Sanya Roysommuti
- Department of Cell, Developmental and Integrative Biology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
- Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.
| | - James Michael Wyss
- Department of Cell, Developmental and Integrative Biology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
32
|
Fujitani M, Otani Y, Miyajima H. Do Neurotrophins Connect Neurological Disorders and Heart Diseases? Biomolecules 2021; 11:1730. [PMID: 34827728 PMCID: PMC8615910 DOI: 10.3390/biom11111730] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/08/2021] [Accepted: 11/16/2021] [Indexed: 12/21/2022] Open
Abstract
Neurotrophins (NTs) are one of the most characterized neurotrophic factor family members and consist of four members in mammals. Growing evidence suggests that there is a complex inter- and bi-directional relationship between central nervous system (CNS) disorders and cardiac dysfunction, so-called "brain-heart axis". Recent studies suggest that CNS disorders, including neurodegenerative diseases, stroke, and depression, affect cardiovascular function via various mechanisms, such as hypothalamic-pituitary-adrenal axis augmentation. Although this brain-heart axis has been well studied in humans and mice, the involvement of NT signaling in the axis has not been fully investigated. In the first half of this review, we emphasize the importance of NTs not only in the nervous system, but also in the cardiovascular system from the embryonic stage to the adult state. In the second half, we discuss the involvement of NTs in the pathogenesis of cardiovascular diseases, and then examine whether an alteration in NTs could serve as the mediator between neurological disorders and heart dysfunction. The further investigation we propose herein could contribute to finding direct evidence for the involvement of NTs in the axis and new treatment for cardiovascular diseases.
Collapse
Affiliation(s)
- Masashi Fujitani
- Department of Anatomy and Neuroscience, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo-shi 693-8501, Shimane, Japan; (Y.O.); (H.M.)
| | | | | |
Collapse
|
33
|
Elia A, Cannavo A, Gambino G, Cimini M, Ferrara N, Kishore R, Paolocci N, Rengo G. Aging is associated with cardiac autonomic nerve fiber depletion and reduced cardiac and circulating BDNF levels. J Geriatr Cardiol 2021; 18:549-559. [PMID: 34404991 PMCID: PMC8352776 DOI: 10.11909/j.issn.1671-5411.2021.07.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023] Open
Abstract
BACKGROUND Aging is a multifactorial process associated with an impairment of autonomic nervous system (ANS) function. Progressive ANS remodeling includes upregulation of expression of circulating catecholamines and depletion of cardiac autonomic nerve fibers, and it is responsible, in part, for the increased susceptibility to cardiac diseases observed in elderly subjects. Neurotrophic factors, such as brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF), are involved in synaptogenesis and neurite outgrowth processes, supporting neuronal cell differentiation and maturation. However, whether and how these factors and their downstream signaling are involved in cardiac aging remains unclear. Here, we tested whether, in the aged heart, the overall extent of autonomic fibers is reduced, owing to lower production of trophic factors such as BDNF and NGF. METHODS In vivo, we used young (age: 3 months; n = 10) and old (age: 24 months; n = 11) male Fisher rats, whereas, we used human neuroblastoma (SH-SY5Y) cells in vitro. RESULTS Compared to the young rats, old rats displayed a marked reduction in the overall ANS fiber density, affecting both sympathetic and cholinergic compartments, as indicated by dopamine β-hydroxylase (dβh) and vesicular acetylcholine transporter (VaChT) immunohistochemical staining. In addition, a marked downregulation of GAP-43 and BDNF protein was observed in the left ventricular lysates of old rats compared to those of young rats. Interestingly, we did not find any significant difference in cardiac NGF levels between the young and old groups. To further explore the impact of aging on ANS fibers, we treated SH-SY5Y cells in vitro with serum obtained from young and old rats. Sera from both groups induced a remarkable increase in neuronal sprouting, as evidenced by a crystal violet assay. However, this effect was blunted in cells cultured with old rat serum and was accompanied by a marked reduction in GAP-43 and BDNF protein levels. CONCLUSIONS Our data indicate that physiological aging is associated with an impairment of ANS structure and function and that reduced BDNF levels are responsible, at least in part, for these phenomena.
Collapse
Affiliation(s)
- Andrea Elia
- Department of Translational Medical Sciences, Federico II University of Naples Italy
- Istituti Clinici Scientifici ICS-Maugeri, Telese Terme (BN), Italy
| | - Alessandro Cannavo
- Department of Translational Medical Sciences, Federico II University of Naples Italy
| | - Giuseppina Gambino
- Department of Translational Medical Sciences, Federico II University of Naples Italy
| | - Maria Cimini
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Nicola Ferrara
- Department of Translational Medical Sciences, Federico II University of Naples Italy
- Istituti Clinici Scientifici ICS-Maugeri, Telese Terme (BN), Italy
| | - Raj Kishore
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Nazareno Paolocci
- Division of Cardiology, Johns Hopkins University Medical Institutions, Baltimore, MD, USA
- Department of Biomedical Sciences, University of Padova, Italy
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, Federico II University of Naples Italy
- Istituti Clinici Scientifici ICS-Maugeri, Telese Terme (BN), Italy
| |
Collapse
|
34
|
Enhanced BDNF Actions Following Acute Hypoxia Facilitate HIF-1α-Dependent Upregulation of Cav3-T-Type Ca 2+ Channels in Rat Cardiomyocytes. MEMBRANES 2021; 11:membranes11070470. [PMID: 34202148 PMCID: PMC8307968 DOI: 10.3390/membranes11070470] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/16/2021] [Accepted: 06/23/2021] [Indexed: 01/19/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) has recently been recognized as a cardiovascular regulator particularly in the diseased condition, including coronary artery disease, heart failure, cardiomyopathy, and hypertension. Here, we investigate the role of BDNF on the T-type Ca2+ channel, Cav3.1 and Cav3.2, in rat neonatal cardiomyocytes exposed to normoxia (21% O2) and acute hypoxia (1% O2) in vitro for up to 3 h. The exposure of cardiomyocytes to hypoxia (1 h, 3 h) caused a significant upregulation of the mRNAs for hypoxia-inducible factor 1α (Hif1α), Cav3.1, Cav3.2 and Bdnf, but not tropomyosin-related kinase receptor B (TrkB). The upregulation of Cav3.1 and Cav3.2 caused by hypoxia was completely halted by small interfering RNA (siRNA) targeting Hif1a (Hif1a-siRNA) or Bdnf (Bdnf-siRNA). Immunocytochemical staining data revealed a distinct upregulation of Cav3.1- and Cav3.2-proteins caused by hypoxia in cardiomyocytes, which was markedly suppressed by Bdnf-siRNA. These results unveiled a novel regulatory action of BDNF on the T-type Ca2+ channels expression through the HIF-1α-dependent pathway in cardiomyocytes.
Collapse
|
35
|
Yang X, Liu S, Zhang Z. Sex difference in blood pressure, a combinatorial consequence of the differential in RAAS components, sex hormones and time course. Curr Hypertens Rev 2021; 18:11-16. [PMID: 33992057 DOI: 10.2174/1573402117666210511011444] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/26/2021] [Accepted: 03/02/2021] [Indexed: 11/22/2022]
Abstract
The longitudinal increment of blood pressure (BP) with age is attributed to lifestyle, internal and external environments. It is not limited to systemic brain-derived neurotrophic factor (BDNF), signaling to allow the individuals to better adapt to the developmental and environmental change. This regulation is necessary for all lives, regardless of sex. Basic levels of renin-angiotensin- aldosterone system (RAAS) components in males and females define the fundamental sex difference in BP, which may be set by prenatal programming and profoundly influence BP after birth. The innate sex difference in BP is magnified during puberty growth and further modified by menopause. At the age of 70 or older, blood pressure was similar in men and women. The understanding of the prenatal setup and development of sexual dimorphism in BP may provide preventative therapeutic strategies, including timing and drugs, for individuals with abnormal BP.
Collapse
Affiliation(s)
- Xiaomei Yang
- College of Basic Medicine, Henan University of Chinese Medicine,156,Jinshui East Road ,Zhengdong New District, Zhengzhou, Henan 450046. China
| | - Shien Liu
- College of Basic Medicine, Henan University of Chinese Medicine,156,Jinshui East Road ,Zhengdong New District, Zhengzhou, Henan 450046. China
| | - Zhongming Zhang
- College of Chinese Medicine, and Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology; 80 Changjiang Rd. Nanyang, Henan, 473004. China
| |
Collapse
|
36
|
Montone RA, Camilli M, Del Buono MG, Russo M, Rinaldi R, Canonico F, Pedicino D, Severino A, D'Amario D, Trani C, Liuzzo G, Crea F, Niccoli G. Brain-derived neurotrophic factor in patients with acute coronary syndrome. Transl Res 2021; 231:39-54. [PMID: 33221484 DOI: 10.1016/j.trsl.2020.11.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/13/2020] [Accepted: 11/13/2020] [Indexed: 12/20/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is a neurotrophic factor highly expressed in coronary plaques, particularly in macrophages, and in activated platelets. Thus, a possible role in the pathogenesis of acute coronary syndrome (ACS) has been suggested. We evaluated systemic BDNF levels according to the different clinical presentations of ACS. Moreover, we assessed the relationship between BDNF levels and the presence of optical coherence tomography (OCT)-defined macrophage infiltrates (MØI) and healed plaques along the culprit vessel. We enrolled consecutive patients presenting with ST-elevation myocardial infarction (STEMI) or non-ST-elevation (NSTE)-ACS. Serum BDNF levels were assessed by enzyme-linked immunosorbent assay. Plaque characteristics of the culprit vessel were assessed by OCT. Among 126 ACS patients (median age 68.00, interquartile range [IQR] 59.75-75.25 years, male 74.6%, 71 (56.3%) were NSTE-ACS and 55 (43.7%) were STEMI. BDNF levels were higher in STEMI patients compared to NSTE-ACS. OCT assessment was performed in 53 (42.1%) patients. Patients with MØI (n = 27) had higher BDNF levels compared to patients without MØI. Furthermore, patients with healed plaques (n = 13) had lower BDNF levels than patients without healed plaques. At multivariate regression analysis BDNF levels independently predicted the presence of MØI (odds ratio [OR] = 2.856; 95% confidence interval [CI] [1.151-7.090], P = 0.024) and the absence of healed plaques (OR = 0.438, 95% CI [0.185-0.992], P= 0.050). Among ACS patients, BDNF levels were higher in patients with STEMI. Moreover, BDNF levels were independently associated with MØI and with the absence of healed plaques along the culprit vessel, suggesting a possible role of BDNF in promoting plaque inflammation, destabilization and occlusive thrombosis.
Collapse
Affiliation(s)
- Rocco A Montone
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| | - Massimiliano Camilli
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Marco Giuseppe Del Buono
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Michele Russo
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Riccardo Rinaldi
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Francesco Canonico
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Daniela Pedicino
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Anna Severino
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Domenico D'Amario
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Carlo Trani
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy; Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Giovanna Liuzzo
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy; Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Filippo Crea
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy; Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Giampaolo Niccoli
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy; Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy; Division of Cardiology, University of Parma, Parma University Hospital
| |
Collapse
|
37
|
Luong H, Singh S, Patil M, Krishnamurthy P. Cardiac glycosaminoglycans and structural alterations during chronic stress-induced depression-like behavior in mice. Am J Physiol Heart Circ Physiol 2021; 320:H2044-H2057. [PMID: 33834865 DOI: 10.1152/ajpheart.00635.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Major depressive disorder (MDD) is an independent risk factor for cardiovascular disease (CVD) and its complications; however, causal mechanisms remain unclear. In the present study, we investigate cardiac structural and functional alterations and associated changes in myocardial glycosaminoglycans (GAGs) disaccharide profile in mice that exhibit depression-like behavior. Mice were assigned to the chronic mild stress (CMS) group and nonstress control group (CT). The CMS group was exposed to a series of mild, unpredictable stressors for 7 wk. Mice in the CMS group show a significant decrease in protein expression of hippocampal brain-derived neurotrophic factor (BDNF) and exhibit depression-like behavioral changes, such as learned helplessness and decreased exploration behavior, as compared with the control group. Although cardiac function remained unchanged between the groups, echocardiography analysis showed slightly increased left ventricular wall thickness in the CMS group. Furthermore, the CMS group shows an increase in cardiomyocyte cross-sectional area and an associated decrease in BDNF protein expression and increase in IL-6 mRNA expression, when compared with control mice. GAG disaccharide analysis of the left ventricles of the CMS and CT mice revealed an elevation in heparan (HS) and chondroitin sulfate (CS) content in the CMS hearts (35.3% and 17.9%, respectively, vs. control group). Furthermore, we also observed that unsulfated or monosulfated disaccharides were the most abundant units; however, we did not find any significant difference in mole percent or sulfation pattern of HS/CS disaccharides between the groups. The current investigation highlights a need for further research to explore the relationship between cardiac GAGs biology and myocardial remodeling as a causal mechanism that underlie cardiovascular complications in patients with MDD.NEW & NOTEWORTHY Comorbidity between depression and CVD is well established, whereas its etiology, especially the role of nonfibrous components (proteoglycans/GAGs) of the extracellular matrix, is unexplored. To the best of our knowledge, this is the first study to characterize cardiac proteoglycan/glycosaminoglycan profile in response to depression-like behavioral changes in mice. We observed that chronic mild stress (CMS)-induced depression-like behavior and alterations in glycosaminoglycan profile were associated with structural changes in the heart.
Collapse
Affiliation(s)
- Hien Luong
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| | - Sarojini Singh
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| | - Mallikarjun Patil
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| | - Prasanna Krishnamurthy
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
38
|
Zierold S, Buschmann K, Gachkar S, Bochenek ML, Velmeden D, Hobohm L, Vahl CF, Schäfer K. Brain-Derived Neurotrophic Factor Expression and Signaling in Different Perivascular Adipose Tissue Depots of Patients With Coronary Artery Disease. J Am Heart Assoc 2021; 10:e018322. [PMID: 33666096 PMCID: PMC8174206 DOI: 10.1161/jaha.120.018322] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Brain‐derived neurotrophic factor (BDNF) is expressed in neuronal and nonneuronal cells and may affect vascular functions via its receptor, tropomyosin‐related kinase B (TrkB). In this study, we determined the expression of BDNF in different perivascular adipose tissue (PVAT) depots of patients with established coronary atherosclerosis. Methods and Results Serum, vascular tissue, and PVAT surrounding the proximal aorta (C‐PVAT) or internal mammary artery (IMA‐PVAT) was obtained from 24 patients (79% men; mean age, 71.7±9.7 years; median body mass index, 27.4±4.8 kg/m2) with coronary atherosclerosis undergoing elective coronary artery bypass surgery. BDNF protein levels were significantly higher in C‐PVAT compared with IMA‐PVAT, independent of obesity, metabolic syndrome, or systemic biomarkers of inflammation. mRNA transcripts of TrkB, the BDNF receptor, were significantly reduced in aorta compared with IMA. Vessel wall TrkB immunosignals colocalized with cells expressing smooth muscle cell markers, and confocal microscopy and flow cytometry confirmed BDNF receptor expression in human aortic smooth muscle cells. Significantly elevated levels of protein tyrosine phosphatase 1B, a negative regulator of TrkB signaling in the brain, were also observed in C‐PVAT. In vitro, inhibition of protein tyrosine phosphatase 1B blunted the effects of BDNF on smooth muscle cell proliferation, migration, differentiation, and collagen production, possibly by upregulation of low‐affinity p75 neurotrophin receptors. Expression of nerve growth factor or its receptor tropomyosin‐related kinase A did not differ between C‐PVAT and IMA‐PVAT. Conclusions Elevated expression of BDNF in parallel with local upregulation of negative regulators of neurotrophin signaling in perivascular fat and lower TrkB expression suggest that vascular BDNF signaling is reduced or lost in patients with coronary atherosclerosis.
Collapse
Affiliation(s)
- Sarah Zierold
- Department of Cardiology Cardiology I University Medical Center Mainz Mainz Germany
| | - Katja Buschmann
- Department of Cardiothoracic and Vascular Surgery University Medical Center Mainz Mainz Germany
| | - Sogol Gachkar
- Department of Cardiology Cardiology I University Medical Center Mainz Mainz Germany
| | - Magdalena L Bochenek
- Department of Cardiology Cardiology I University Medical Center Mainz Mainz Germany.,Center for Thrombosis and Hemostasis University Medical Center Mainz Mainz Germany
| | - David Velmeden
- Department of Cardiology Cardiology I University Medical Center Mainz Mainz Germany
| | - Lukas Hobohm
- Department of Cardiology Cardiology I University Medical Center Mainz Mainz Germany.,Center for Thrombosis and Hemostasis University Medical Center Mainz Mainz Germany
| | | | - Katrin Schäfer
- Department of Cardiology Cardiology I University Medical Center Mainz Mainz Germany
| |
Collapse
|
39
|
Latchoumane CFV, Betancur MI, Simchick GA, Sun MK, Forghani R, Lenear CE, Ahmed A, Mohankumar R, Balaji N, Mason HD, Archer-Hartmann SA, Azadi P, Holmes PV, Zhao Q, Bellamkonda RV, Karumbaiah L. Engineered glycomaterial implants orchestrate large-scale functional repair of brain tissue chronically after severe traumatic brain injury. SCIENCE ADVANCES 2021; 7:7/10/eabe0207. [PMID: 33674306 PMCID: PMC7935369 DOI: 10.1126/sciadv.abe0207] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 01/21/2021] [Indexed: 05/14/2023]
Abstract
Severe traumatic brain injury (sTBI) survivors experience permanent functional disabilities due to significant volume loss and the brain's poor capacity to regenerate. Chondroitin sulfate glycosaminoglycans (CS-GAGs) are key regulators of growth factor signaling and neural stem cell homeostasis in the brain. However, the efficacy of engineered CS (eCS) matrices in mediating structural and functional recovery chronically after sTBI has not been investigated. We report that neurotrophic factor functionalized acellular eCS matrices implanted into the rat M1 region acutely after sTBI significantly enhanced cellular repair and gross motor function recovery when compared to controls 20 weeks after sTBI. Animals subjected to M2 region injuries followed by eCS matrix implantations demonstrated the significant recovery of "reach-to-grasp" function. This was attributed to enhanced volumetric vascularization, activity-regulated cytoskeleton (Arc) protein expression, and perilesional sensorimotor connectivity. These findings indicate that eCS matrices implanted acutely after sTBI can support complex cellular, vascular, and neuronal circuit repair chronically after sTBI.
Collapse
Affiliation(s)
- Charles-Francois V Latchoumane
- Regenerative Bioscience Center, University of Georgia, Athens, GA 30602, USA
- Edgar L. Rhodes Center for ADS, College of Agriculture and Environmental Sciences, University of Georgia, Athens, GA 30602, USA
| | - Martha I Betancur
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, 101 Science Drive, Durham, NC 27705, USA
| | - Gregory A Simchick
- Department of Physics and Astronomy, University of Georgia, Athens, GA 30602, USA
- Bio-Imaging Research Center, University of Georgia, Athens, GA 30602, USA
| | - Min Kyoung Sun
- Regenerative Bioscience Center, University of Georgia, Athens, GA 30602, USA
- Division of Neuroscience, Biomedical & Health Sciences Institute, University of Georgia, Athens, GA 30602, USA
| | - Rameen Forghani
- Regenerative Bioscience Center, University of Georgia, Athens, GA 30602, USA
| | - Christopher E Lenear
- Regenerative Bioscience Center, University of Georgia, Athens, GA 30602, USA
- Edgar L. Rhodes Center for ADS, College of Agriculture and Environmental Sciences, University of Georgia, Athens, GA 30602, USA
| | - Aws Ahmed
- Regenerative Bioscience Center, University of Georgia, Athens, GA 30602, USA
- Edgar L. Rhodes Center for ADS, College of Agriculture and Environmental Sciences, University of Georgia, Athens, GA 30602, USA
| | - Ramya Mohankumar
- Regenerative Bioscience Center, University of Georgia, Athens, GA 30602, USA
| | - Nivedha Balaji
- Regenerative Bioscience Center, University of Georgia, Athens, GA 30602, USA
| | - Hannah D Mason
- Regenerative Bioscience Center, University of Georgia, Athens, GA 30602, USA
| | | | - Parastoo Azadi
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Philip V Holmes
- Division of Neuroscience, Biomedical & Health Sciences Institute, University of Georgia, Athens, GA 30602, USA
- Psychology Department, University of Georgia, Athens, GA 30602, USA
| | - Qun Zhao
- Regenerative Bioscience Center, University of Georgia, Athens, GA 30602, USA
- Department of Physics and Astronomy, University of Georgia, Athens, GA 30602, USA
- Bio-Imaging Research Center, University of Georgia, Athens, GA 30602, USA
| | - Ravi V Bellamkonda
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, 101 Science Drive, Durham, NC 27705, USA
| | - Lohitash Karumbaiah
- Regenerative Bioscience Center, University of Georgia, Athens, GA 30602, USA.
- Edgar L. Rhodes Center for ADS, College of Agriculture and Environmental Sciences, University of Georgia, Athens, GA 30602, USA
- Division of Neuroscience, Biomedical & Health Sciences Institute, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
40
|
Ehinger Y, Phamluong K, Darevesky D, Welman M, Moffat JJ, Sakhai SA, Whiteley EL, Berger AL, Laguesse S, Farokhnia M, Leggio L, Lordkipanidzé M, Ron D. Differential correlation of serum BDNF and microRNA content in rats with rapid or late onset of heavy alcohol use. Addict Biol 2021; 26:e12890. [PMID: 32135570 DOI: 10.1111/adb.12890] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 02/13/2020] [Accepted: 02/15/2020] [Indexed: 12/22/2022]
Abstract
Heavy alcohol use reduces the levels of the brain-derived neurotrophic factor (BDNF) in the prefrontal cortex of rodents through the upregulation of microRNAs (miRs) targeting BDNF mRNA. In humans, an inverse correlation exists between circulating blood levels of BDNF and the severity of psychiatric disorders including alcohol abuse. Here, we set out to determine whether a history of heavy alcohol use produces comparable alterations in the blood of rats. We used an intermittent access to 20% alcohol using the two-bottle choice paradigm (IA20%2BC) and measured circulating levels of BDNF protein and miRs targeting BDNF in the serum of Long-Evans rats before and after 8 weeks of excessive alcohol intake. We observed that the drinking profile of heavy alcohol users is not unified, whereas 70% of the rats gradually escalate their alcohol intake (late onset), and 30% of alcohol users exhibit a very rapid onset of drinking (rapid onset). We found that serum BDNF levels are negatively correlated with alcohol intake in both rapid onset and late onset rats. In contrast, increased expression of the miRs targeting BDNF, miR30a-5p, miR-195-5p, miR191-5p and miR206-3p, was detected only in the rapid onset rats. Finally, we report that the alcohol-dependent molecular changes are not due to alterations in platelet number. Together, these data suggest that rats exhibit both late and rapid onset of alcohol intake. We further show that heavy alcohol use produces comparable changes in BDNF protein levels in both groups. However, circulating microRNAs are responsive to alcohol only in the rapid onset rats.
Collapse
Affiliation(s)
- Yann Ehinger
- Department of Neurology University of California, San Francisco San Francisco California
| | - Khanhky Phamluong
- Department of Neurology University of California, San Francisco San Francisco California
| | - David Darevesky
- Department of Neurology University of California, San Francisco San Francisco California
| | - Melanie Welman
- Research Center Montreal Heart Institute Montreal Quebec Canada
| | - Jeffrey J. Moffat
- Department of Neurology University of California, San Francisco San Francisco California
| | - Samuel A. Sakhai
- Department of Neurology University of California, San Francisco San Francisco California
| | - Ellanor L. Whiteley
- Department of Neurology University of California, San Francisco San Francisco California
| | - Anthony L. Berger
- Department of Neurology University of California, San Francisco San Francisco California
| | - Sophie Laguesse
- Department of Neurology University of California, San Francisco San Francisco California
| | - Mehdi Farokhnia
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section National Institute on Drug Abuse Intramural Research Program Baltimore Maryland
- Medication Development Program, National Institute on Drug Abuse Intramural Research Program National Institutes of Health Baltimore Maryland
- National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research National Institutes of Health Bethesda Maryland
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section National Institute on Drug Abuse Intramural Research Program Baltimore Maryland
- Medication Development Program, National Institute on Drug Abuse Intramural Research Program National Institutes of Health Baltimore Maryland
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences Brown University Providence Rhode Island
- National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research National Institutes of Health Bethesda Maryland
| | - Marie Lordkipanidzé
- Research Center Montreal Heart Institute Montreal Quebec Canada
- Faculty of Pharmacy University of Montreal Montreal Quebec Canada
| | - Dorit Ron
- Department of Neurology University of California, San Francisco San Francisco California
| |
Collapse
|
41
|
Szabó MR, Pipicz M, Csont T, Csonka C. Modulatory Effect of Myokines on Reactive Oxygen Species in Ischemia/Reperfusion. Int J Mol Sci 2020; 21:ijms21249382. [PMID: 33317180 PMCID: PMC7763329 DOI: 10.3390/ijms21249382] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/06/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022] Open
Abstract
There is a growing body of evidence showing the importance of physical activity against acute ischemic events in various organs. Ischemia/reperfusion injury (I/R) is characterized by tissue damage as a result of restriction and subsequent restoration of blood supply to an organ. Oxidative stress due to increased reactive oxygen species formation and/or insufficient antioxidant defense is considered to play an important role in I/R. Physical activity not only decreases the general risk factors for ischemia but also confers direct anti-ischemic protection via myokine production. Myokines are skeletal muscle-derived cytokines, representing multifunctional communication channels between the contracting skeletal muscle and other organs through an endocrine manner. In this review, we discuss the most prominent members of the myokines (i.e., brain-derived neurotrophic factor (BDNF), cathepsin B, decorin, fibroblast growth factors-2 and -21, follistatin, follistatin-like, insulin-like growth factor-1; interleukin-6, interleukin-7, interleukin-15, irisin, leukemia inhibitory factor, meteorin-like, myonectin, musclin, myostatin, and osteoglycin) with a particular interest in their potential influence on reactive oxygen and nitrogen species formation or antioxidant capacity. A better understanding of the mechanism of action of myokines and particularly their participation in the regulation of oxidative stress may widen their possible therapeutic use and, thereby, may support the fight against I/R.
Collapse
Affiliation(s)
- Márton Richárd Szabó
- Metabolic Diseases and Cell Signaling (MEDICS) Research Group, Department of Biochemistry, University of Szeged, Dóm tér 9, 6720 Szeged, Hungary; (M.R.S.); (M.P.); (T.C.)
- Interdisciplinary Centre of Excellence, University of Szeged, Dugonics tér 13, 6720 Szeged, Hungary
| | - Márton Pipicz
- Metabolic Diseases and Cell Signaling (MEDICS) Research Group, Department of Biochemistry, University of Szeged, Dóm tér 9, 6720 Szeged, Hungary; (M.R.S.); (M.P.); (T.C.)
- Interdisciplinary Centre of Excellence, University of Szeged, Dugonics tér 13, 6720 Szeged, Hungary
| | - Tamás Csont
- Metabolic Diseases and Cell Signaling (MEDICS) Research Group, Department of Biochemistry, University of Szeged, Dóm tér 9, 6720 Szeged, Hungary; (M.R.S.); (M.P.); (T.C.)
- Interdisciplinary Centre of Excellence, University of Szeged, Dugonics tér 13, 6720 Szeged, Hungary
| | - Csaba Csonka
- Metabolic Diseases and Cell Signaling (MEDICS) Research Group, Department of Biochemistry, University of Szeged, Dóm tér 9, 6720 Szeged, Hungary; (M.R.S.); (M.P.); (T.C.)
- Interdisciplinary Centre of Excellence, University of Szeged, Dugonics tér 13, 6720 Szeged, Hungary
- Department of Sports Medicine, University of Szeged, Tisza Lajos krt 107, 6725 Szeged, Hungary
- Correspondence: ; Tel.: +36-30-5432-693
| |
Collapse
|
42
|
Murthy VL, Reis JP, Pico AR, Kitchen R, Lima JAC, Lloyd-Jones D, Allen NB, Carnethon M, Lewis GD, Nayor M, Vasan RS, Freedman JE, Clish CB, Shah RV. Comprehensive Metabolic Phenotyping Refines Cardiovascular Risk in Young Adults. Circulation 2020; 142:2110-2127. [PMID: 33073606 PMCID: PMC7880553 DOI: 10.1161/circulationaha.120.047689] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 09/17/2020] [Indexed: 01/19/2023]
Abstract
BACKGROUND Whereas cardiovascular disease (CVD) metrics define risk in individuals >40 years of age, the earliest lesions of CVD appear well before this age. Despite the role of metabolism in CVD antecedents, studies in younger, biracial populations to define precise metabolic risk phenotypes are lacking. METHODS We studied 2330 White and Black young adults (mean age, 32 years; 45% Black) in the CARDIA study (Coronary Artery Risk Development in Young Adults) to identify metabolite profiles associated with an adverse CVD phenome (myocardial structure/function, fitness, vascular calcification), mechanisms, and outcomes over 2 decades. Statistical learning methods (elastic nets/principal components analysis) and Cox regression generated parsimonious, metabolite-based risk scores validated in >1800 individuals in the Framingham Heart Study. RESULTS In the CARDIA study, metabolite profiles quantified in early adulthood were associated with subclinical CVD development over 20 years, specifying known and novel pathways of CVD (eg, transcriptional regulation, brain-derived neurotrophic factor, nitric oxide, renin-angiotensin). We found 2 multiparametric, metabolite-based scores linked independently to vascular and myocardial health, with metabolites included in each score specifying microbial metabolism, hepatic steatosis, oxidative stress, nitric oxide modulation, and collagen metabolism. The metabolite-based vascular scores were lower in men, and myocardial scores were lower in Black participants. Over a nearly 25-year median follow-up in CARDIA, the metabolite-based vascular score (hazard ratio, 0.68 per SD [95% CI, 0.50-0.92]; P=0.01) and myocardial score (hazard ratio, 0.60 per SD [95% CI, 0.45-0.80]; P=0.0005) in the third and fourth decades of life were associated with clinical CVD with a synergistic association with outcome (Pinteraction=0.009). We replicated these findings in 1898 individuals in the Framingham Heart Study over 2 decades, with a similar association with outcome (including interaction), reclassification, and discrimination. In the Framingham Heart Study, the metabolite scores exhibited an age interaction (P=0.0004 for a combined myocardial-vascular score with incident CVD), such that young adults with poorer metabolite-based health scores had highest hazard of future CVD. CONCLUSIONS Metabolic signatures of myocardial and vascular health in young adulthood specify known/novel pathways of metabolic dysfunction relevant to CVD, associated with outcome in 2 independent cohorts. Efforts to include precision measures of metabolic health in risk stratification to interrupt CVD at its earliest stage are warranted.
Collapse
Affiliation(s)
| | - Jared P. Reis
- National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Alexander R. Pico
- Institute of Data Science and Biotechnology, Gladstone Institutes, University of California at San Francisco, San Francisco, CA
| | - Robert Kitchen
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Joao A. C. Lima
- Cardiology Division, Department of Medicine, Johns Hopkins Hospital, Baltimore, MD
| | | | | | | | - Gregory D. Lewis
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Matthew Nayor
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Ramachandran S. Vasan
- Sections of Preventive Medicine and Epidemiology and Cardiovascular Medicine, Department of Medicine, and Department of Epidemiology, Boston University Schools of Medicine and Public Health, Boston, MA, and the Framingham Heart Study, Framingham, MA
| | - Jane E. Freedman
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | | | - Ravi V. Shah
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
43
|
Raucci FJ, Singh AP, Soslow J, Markham LW, Zhong L, Aljafar W, Lessiohadi N, Awgulewitsch CP, Umbarkar P, Zhang Q, Cannon PL, Buchowski M, Roland JT, Carrier EJ, Burnette WB, Hatzopoulos AK, Lal H, Galindo CL. The BDNF rs6265 Polymorphism is a Modifier of Cardiomyocyte Contractility and Dilated Cardiomyopathy. Int J Mol Sci 2020; 21:ijms21207466. [PMID: 33050457 PMCID: PMC7593910 DOI: 10.3390/ijms21207466] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 09/25/2020] [Accepted: 10/08/2020] [Indexed: 12/22/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a neuronal growth and survival factor that harbors cardioprotective qualities that may attenuate dilated cardiomyopathy. In ~30% of the population, BDNF has a common, nonsynonymous single nucleotide polymorphism rs6265 (Val66Met), which might be correlated with increased risk of cardiovascular events. We previously showed that BDNF correlates with better cardiac function in Duchenne muscular dystrophy (DMD) patients. However, the effect of the Val66Met polymorphism on cardiac function has not been determined. The goal of the current study was to determine the effects of rs6265 on BDNF biomarker suitability and DMD cardiac functions more generally. We assessed cardiovascular and skeletal muscle function in human DMD patients segregated by polymorphic allele. We also compared echocardiographic, electrophysiologic, and cardiomyocyte contractility in C57/BL-6 wild-type mice with rs6265 polymorphism and in mdx/mTR (mDMD) mouse model of DMD. In human DMD patients, plasma BDNF levels had a positive correlation with left ventricular function, opposite to that seen in rs6265 carriers. There was also a substantial decrease in skeletal muscle function in carriers compared to the Val homozygotes. Surprisingly, the opposite was true when cardiac function of DMD carriers and non-carriers were compared. On the other hand, Val66Met wild-type mice had only subtle functional differences at baseline but significantly decreased cardiomyocyte contractility. Our results indicate that the Val66Met polymorphism alters myocyte contractility, conferring worse skeletal muscle function but better cardiac function in DMD patients. Moreover, these results suggest a mechanism for the relative preservation of cardiac tissues compared to skeletal muscle in DMD patients and underscores the complexity of BDNF signaling in response to mechanical workload.
Collapse
Affiliation(s)
- Frank J. Raucci
- Thomas P. Graham Division of Pediatric Cardiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (F.J.R.J.); (J.S.)
- Division of Pediatric Cardiology, Department of Pediatrics, Children’s Hospital of Richmond at Virginia Commonwealth University, Richmond, VA 23219, USA
| | - Anand Prakash Singh
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama Birmingham School of Medicine, Birmingham, AL 35233, USA; (A.P.S.); (P.U.); (Q.Z.); (H.L.)
| | - Jonathan Soslow
- Thomas P. Graham Division of Pediatric Cardiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (F.J.R.J.); (J.S.)
| | - Larry W. Markham
- Division of Cardiology, Department of Pediatrics, Riley Hospital for Children at Indiana University Health, Indianapolis, IN 46202, USA;
| | - Lin Zhong
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (L.Z.); (W.A.); (N.L.); (C.P.A.); (P.L.C.); (J.T.R.); (E.J.C.); (A.K.H.)
| | - Wejdan Aljafar
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (L.Z.); (W.A.); (N.L.); (C.P.A.); (P.L.C.); (J.T.R.); (E.J.C.); (A.K.H.)
| | - Natasja Lessiohadi
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (L.Z.); (W.A.); (N.L.); (C.P.A.); (P.L.C.); (J.T.R.); (E.J.C.); (A.K.H.)
| | - Cassandra P. Awgulewitsch
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (L.Z.); (W.A.); (N.L.); (C.P.A.); (P.L.C.); (J.T.R.); (E.J.C.); (A.K.H.)
| | - Prachi Umbarkar
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama Birmingham School of Medicine, Birmingham, AL 35233, USA; (A.P.S.); (P.U.); (Q.Z.); (H.L.)
| | - Qinkun Zhang
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama Birmingham School of Medicine, Birmingham, AL 35233, USA; (A.P.S.); (P.U.); (Q.Z.); (H.L.)
| | - Presley L. Cannon
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (L.Z.); (W.A.); (N.L.); (C.P.A.); (P.L.C.); (J.T.R.); (E.J.C.); (A.K.H.)
| | - Maciej Buchowski
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Joseph T. Roland
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (L.Z.); (W.A.); (N.L.); (C.P.A.); (P.L.C.); (J.T.R.); (E.J.C.); (A.K.H.)
| | - Erica J. Carrier
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (L.Z.); (W.A.); (N.L.); (C.P.A.); (P.L.C.); (J.T.R.); (E.J.C.); (A.K.H.)
| | - William B. Burnette
- Division of Pediatric Neurology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Antonis K. Hatzopoulos
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (L.Z.); (W.A.); (N.L.); (C.P.A.); (P.L.C.); (J.T.R.); (E.J.C.); (A.K.H.)
| | - Hind Lal
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama Birmingham School of Medicine, Birmingham, AL 35233, USA; (A.P.S.); (P.U.); (Q.Z.); (H.L.)
| | - Cristi L. Galindo
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (L.Z.); (W.A.); (N.L.); (C.P.A.); (P.L.C.); (J.T.R.); (E.J.C.); (A.K.H.)
- Department of Biology, Western Kentucky University, Bowling Green, KY 42101, USA
- Correspondence: ; Tel.: +1-270-745-3696
| |
Collapse
|
44
|
Monisha KG, Prabu P, Chokkalingam M, Murugesan R, Milenkovic D, Ahmed SSSJ. Clinical utility of brain-derived neurotrophic factor as a biomarker with left ventricular echocardiographic indices for potential diagnosis of coronary artery disease. Sci Rep 2020; 10:16359. [PMID: 33004884 PMCID: PMC7530751 DOI: 10.1038/s41598-020-73296-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 08/26/2020] [Indexed: 11/12/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) plays a central pivotal role in the development of the cardiovascular system. Recent evidence suggests that BDNF has adverse subclinical cardiac remodeling in participants with cardiovascular disease risk factors. Relating serum BDNF levels with two-dimensional echocardiographic indices will provide insights into the BDNF mediated pathophysiology in coronary artery disease (CAD) that may shed light upon potential diagnostic biomarkers. For the study, 221 participants were recruited and classified based on coronary angiogram examination as control (n = 105) and CAD (n = 116). All participants underwent routine blood investigation, two-dimensional echocardiography, and serum BDNF estimation. As a result, total cholesterol, triglyceride, low-density lipid, high-density lipid, HbA1c (glycosylated hemoglobin), serum creatinine, eosinophils, lymphocyte, monocytes, neutrophils, and platelets were significantly elevated in CAD individuals compared to controls. Notably, the serum BDNF was significantly lower in individuals with CAD (30.69 ± 5.45 ng/ml) than controls (46.58 ± 7.95 ng/ml). Multivariate regression analysis showed neutrophils, total cholesterol, left ventricular mass index, mitral inflow E/A ratio, and pulmonary vein AR duration were associated with low BDNF in CAD. Four independent support vector machine (SVM) models performed to ensure the BDNF level in the classification of CAD from healthy controls. Particularly, the model with serum BDNF concentration and blood parameters of CAD achieved significant improvement from 90.95 to 98.19% in detecting CAD from healthy controls. Overall, our analysis provides a significant molecular linkage between the serum BDNF level and cardiovascular function. Our results contribute to the emerging evidence of BDNF as a potential diagnostic value in CAD that might lead to clinical application.
Collapse
Affiliation(s)
- K G Monisha
- Department of Cardiology, Chettinad Hospital and Research Institute, Chettinad Health City, Kelambakkam, Tamil Nadu, 603103, India
| | - Paramasivam Prabu
- School of Medicine, Department of Neurology, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, USA
| | - M Chokkalingam
- Department of Cardiology, Chettinad Hospital and Research Institute, Chettinad Health City, Kelambakkam, Tamil Nadu, 603103, India
| | - Ram Murugesan
- Drug Discovery and Multi-Omics Laboratory, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu, 603103, India
| | | | - Shiek S S J Ahmed
- Drug Discovery and Multi-Omics Laboratory, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu, 603103, India.
| |
Collapse
|
45
|
Wang T, Maltez MT, Lee HW, Ahmad M, Wang HW, Leenen FHH. Effect of exercise training on the FNDC5/BDNF pathway in spontaneously hypertensive rats. Physiol Rep 2020; 7:e14323. [PMID: 31883222 PMCID: PMC6934876 DOI: 10.14814/phy2.14323] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Increased sympathetic activity contributes to the development of cardiovascular diseases such as hypertension. Exercise training lowers sympathetic activity and is beneficial for the prevention and treatment of hypertension and associated cognitive impairment. Increased BDNF expression in skeletal muscle, heart, and brain may contribute to these actions of exercise, but the mechanisms by which this occurs are unknown. We postulated that hypertension is associated with decreased hippocampal BDNF, which can be restored by exercise‐mediated upregulation of fibronectin type‐II domain‐containing 5 (FNDC5). Spontaneously hypertensive rats (SHR) and normotensive Wistar–Kyoto rats (WKY) were subjected to 5 weeks of motorized treadmill training. BDNF and FNDC5 expressions were measured in the left ventricle (LV), quadriceps, soleus muscle, and brain areas. Exercise training reduced blood pressure (BP) in both strains. BDNF and FNDC5 protein in the LV were increased in SHR, but exercise increased only BDNF protein in both strains. BDNF mRNA, but not protein, was increased in the quadriceps of SHR, and BDNF mRNA and protein were decreased by exercise in both groups. FNDC5 protein was higher in SHR in both the quadriceps and soleus muscle, whereas exercise increased FNDC5 protein only in the quadriceps in both strains. BDNF mRNA was lower in the dentate gyrus (DG) of SHR, which was normalized by exercise. BDNF mRNA expression in the DG negatively correlated with BP. No differences in FNDC5 expression were observed in the brain, suggesting that enhanced BDNF signaling may contribute to the cardiovascular and neurological benefits of exercise training, and these processes involve peripheral, but not central, FNDC5.
Collapse
Affiliation(s)
- Tao Wang
- Brain and Heart Research Group, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Melissa T Maltez
- Brain and Heart Research Group, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Heow Won Lee
- Brain and Heart Research Group, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Monir Ahmad
- Brain and Heart Research Group, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Hong-Wei Wang
- Brain and Heart Research Group, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Frans H H Leenen
- Brain and Heart Research Group, University of Ottawa Heart Institute, Ottawa, ON, Canada
| |
Collapse
|
46
|
Vita GL, Aguennouz M, Polito F, Oteri R, Russo M, Gentile L, Barbagallo C, Ragusa M, Rodolico C, Di Giorgio RM, Toscano A, Vita G, Mazzeo A. Circulating microRNAs Profile in Patients With Transthyretin Variant Amyloidosis. Front Mol Neurosci 2020; 13:102. [PMID: 32655365 PMCID: PMC7325132 DOI: 10.3389/fnmol.2020.00102] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 05/12/2020] [Indexed: 12/11/2022] Open
Abstract
Transthyretin variant amyloidosis (ATTRv) is a rare autosomal dominant disease characterized by the accumulation of amyloid in many organs, mostly causing a sensory-motor neuropathy, cardiomyopathy, and dysautonomia. The aim of the study was to report microRNAs (miRNAs) expression profile identified in the blood of ATTRv patients. Ten ATTRv patients, 10 asymptomatic carriers of transthyretin variant (TTRv), 10 patients with Charcot-Marie-Tooth (CMT) disease, and 10 healthy controls were studied. Human Schwann cells cultures were used to study the regulatory effects of miR-150-5p on the expression of cAMP response element-binding protein (CREB), brain-derived neurotrophic factor (BDNF), and nerve growth factor (NGF). ATTRv patients had 33 miRNAs up-regulated and 48 down-regulated versus healthy controls; 9 miRNAs were up-regulated and 30 down-regulated versus CMT patients; 19 miRNAs were up-regulated and 38 down-regulated versus asymptomatic TTRv carriers. Twelve out of the 19 upregulated miRNAs had a fold increase higher than 100. The validation experiment indicated miR-150-5p as a valuable biomarker to differentiate ATTRv patients from asymptomatic TTRv carriers (AUC: 0.9728; p < 0.0001). Schwann cells culture model demonstrated that miR-150-5p is a powerful negative regulator of CREB, BDNF, and NGF genes. Identification of deregulated miRNAs can help in understanding the complex pathomechamism underlying the development of ATTRv and related multisystemic pathology. Further investigations are needed on the role of circulating miR-150-5p to predict the shift of TTRv carriers from an asymptomatic status to symptoms appearance.
Collapse
Affiliation(s)
- Gian Luca Vita
- Nemo Sud Clinical Centre for Neuromuscular Disorders, Messina, Italy
| | - M'Hammed Aguennouz
- Unit of Neurology and Neuromuscular Diseases, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy.,Mohammed VI University of Health Sciences, Casablanca, Morocco
| | - Francesca Polito
- Unit of Neurology and Neuromuscular Diseases, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Rosaria Oteri
- Unit of Neurology and Neuromuscular Diseases, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Massimo Russo
- Unit of Neurology and Neuromuscular Diseases, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Luca Gentile
- Unit of Neurology and Neuromuscular Diseases, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Cristina Barbagallo
- Molecular, Genome and Complex Systems BioMedicine Unit, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Marco Ragusa
- Molecular, Genome and Complex Systems BioMedicine Unit, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.,Oasi Research Institute IRCCS, Troina, Italy
| | - Carmelo Rodolico
- Unit of Neurology and Neuromuscular Diseases, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Rosa Maria Di Giorgio
- Unit of Neurology and Neuromuscular Diseases, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Antonio Toscano
- Unit of Neurology and Neuromuscular Diseases, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Giuseppe Vita
- Nemo Sud Clinical Centre for Neuromuscular Disorders, Messina, Italy.,Unit of Neurology and Neuromuscular Diseases, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Anna Mazzeo
- Unit of Neurology and Neuromuscular Diseases, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| |
Collapse
|
47
|
Impact of BDNF Val66Met Polymorphism on Myocardial Infarction: Exploring the Macrophage Phenotype. Cells 2020; 9:cells9051084. [PMID: 32349267 PMCID: PMC7290372 DOI: 10.3390/cells9051084] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/22/2020] [Accepted: 04/24/2020] [Indexed: 12/13/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a member of the neurotrophin growth factor family, well known for its role in the homeostasis of the cardiovascular system. Recently, the human BDNF Val66Met single nucleotide polymorphism has been associated with the increased propensity for arterial thrombosis related to acute myocardial infarction (AMI). Using cardiac magnetic resonance imaging and immunohistochemistry analyses, we showed that homozygous mice carrying the human BDNF Val66Met polymorphism (BDNFMet/Met) undergoing left anterior descending (LAD) coronary artery ligation display an adverse cardiac remodeling compared to wild-type (BDNFVal/Val). Interestingly, we observed a persistent presence of pro-inflammatory M1-like macrophages and a reduced accumulation of reparative-like phenotype macrophages (M2-like) in the infarcted heart of mutant mice. Further qPCR analyses showed that BDNFMet/Met peritoneal macrophages are more pro-inflammatory and have a higher migratory ability compared to BDNFVal/Val ones. Finally, macrophages differentiated from circulating monocytes isolated from BDNFMet/Met patients with coronary heart disease displayed the same pro-inflammatory characteristics of the murine ones. In conclusion, the BDNF Val66Met polymorphism predisposes to adverse cardiac remodeling after myocardial infarction in a mouse model and affects macrophage phenotype in both humans and mice. These results provide a new cellular mechanism by which this human BDNF genetic variant could influence cardiovascular disease.
Collapse
|
48
|
Elsworthy RJ, Aldred S. The effect of age and obesity on platelet amyloid precursor protein processing and plasma markers of oxidative stress and inflammation. Exp Gerontol 2020; 132:110838. [PMID: 31981682 DOI: 10.1016/j.exger.2020.110838] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 12/06/2019] [Accepted: 01/10/2020] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Advancing age is a major risk factor for a range of diseases such as, cardiovascular disease, diabetes, cancer and neurodegenerative diseases. In addition, over a third of the population are overweight and obesity is becoming more prevalent in younger people. Ageing and obesity are both linked to a chronic proinflammatory state and elevated oxidative stress, which have both been implicated in cardiovascular and neurodegenerative diseases. Platelets contain all the necessary machinery to process the Amyloid precursor protein AβPP, a pathway thought to be perturbed in Alzheimer's Disease (AD). The ratio of AβPP isoforms present in platelets, and the amount of alpha secretase ADAM10, that works to process AβPP, appear to be associated with cognitive decline and a diagnosis of Alzheimer's disease. The aim of this study was to assess changes in AβPP ratio, ADAM10 and markers of inflammation and oxidative stress with ageing and obesity. MATERIALS AND METHODS Ninety participants were recruited to this study to provide one blood sample. Platelet-rich plasma and platelet lysates were collected and the expression of AβPPr, proADAM10 and mADAM10 was assessed by Western blotting. In addition, markers of inflammation (IL-6) and oxidative stress (8-Isoprostane) were assessed in plasma. RESULTS Participants were placed into one of four groups based on their age and body mass index (Young/Lean, Young/obese, Old/Lean and Old/Obese). IL-6 was able to significantly distinguish obese from lean participants (AUC of 0.80, SE = 0.05, P < 0.001). Plasma isoprostanes were able to distinguish between both young/old (AUC of 0.73, SE = 0.05, P < 0.01), and obese/non-obese participants (AUC of 0.66, SE = 0.01, P < 0.01). Plasma protein carbonyls could distinguish young and old participants (AUC of 0.69, SE = 0.07 P < 0.02). Old Lean participants had significantly lower mADAM10 expression than both Young Lean and Young Obese participants (p < 0.05). Old obese participants had significantly lower proADAM10 expression compared to both Young Lean and Young Obese (p < 0.05). Both mADAM10 and proADAM10 were significantly decreased with advancing age (p < 0.05). CONCLUSIONS The findings presented in this study provide evidence that blood-based biomarkers related to the pathology of AD are altered with age and obesity in otherwise healthy adults. Ageing was more strongly associated with elevated markers of oxidative stress whereas obesity was associated with elevated inflammatory IL-6.
Collapse
Affiliation(s)
- Richard J Elsworthy
- School of Sport, Exercise and Rehabilitation Sciences, College of Life and Environmental Sciences, University of Birmingham, UK
| | - Sarah Aldred
- School of Sport, Exercise and Rehabilitation Sciences, College of Life and Environmental Sciences, University of Birmingham, UK.
| |
Collapse
|
49
|
Dux M, Rosta J, Messlinger K. TRP Channels in the Focus of Trigeminal Nociceptor Sensitization Contributing to Primary Headaches. Int J Mol Sci 2020; 21:ijms21010342. [PMID: 31948011 PMCID: PMC6981722 DOI: 10.3390/ijms21010342] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 12/31/2019] [Accepted: 01/02/2020] [Indexed: 12/12/2022] Open
Abstract
Pain in trigeminal areas is driven by nociceptive trigeminal afferents. Transduction molecules, among them the nonspecific cation channels transient receptor potential vanilloid 1 (TRPV1) and ankyrin 1 (TRPA1), which are activated by endogenous and exogenous ligands, are expressed by a significant population of trigeminal nociceptors innervating meningeal tissues. Many of these nociceptors also contain vasoactive neuropeptides such as calcitonin gene-related peptide (CGRP) and substance P. Release of neuropeptides and other functional properties are frequently examined using the cell bodies of trigeminal neurons as models of their sensory endings. Pathophysiological conditions cause phosphorylation, increased expression and trafficking of transient receptor potential (TRP) channels, neuropeptides and other mediators, which accelerate activation of nociceptive pathways. Since nociceptor activation may be a significant pathophysiological mechanism involved in both peripheral and central sensitization of the trigeminal nociceptive pathway, its contribution to the pathophysiology of primary headaches is more than likely. Metabolic disorders and medication-induced painful states are frequently associated with TRP receptor activation and may increase the risk for primary headaches.
Collapse
Affiliation(s)
- Mária Dux
- Department of Physiology, University of Szeged, Dóm tér 10, H-6720 Szeged, Hungary;
- Correspondence: ; Tel.: +36-62-545-374; Fax: +36-62-545-842
| | - Judit Rosta
- Department of Physiology, University of Szeged, Dóm tér 10, H-6720 Szeged, Hungary;
| | - Karl Messlinger
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-University Erlangen-Nürnberg, Universitätsstr. 17, D-91054 Erlangen, Germany;
| |
Collapse
|
50
|
Benicky J, Sanda M, Brnakova Kennedy Z, Goldman R. N-Glycosylation is required for secretion of the precursor to brain-derived neurotrophic factor (proBDNF) carrying sulfated LacdiNAc structures. J Biol Chem 2019; 294:16816-16830. [PMID: 31558607 DOI: 10.1074/jbc.ra119.009989] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/18/2019] [Indexed: 01/17/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is generated by proteolytic cleavage of a prodomain from the proBDNF precursor either intracellularly by furin-like proteases or extracellularly by plasmin or matrix metalloproteinases. ProBDNF carries a single N-glycosylation sequon (Asn-127) that remains virtually unstudied despite being located in a highly conserved region proximal to the proteolytic site. To study the proBDNF structure and function, here we expressed the protein and its nonglycosylated N127Q mutant in HEK293F cells. We found that mutation of the Asn-127 prevents intracellular maturation and secretion, an effect reproduced in WT proBDNF by tunicamycin-induced inhibition of N-glycosylation. Absence of the N-glycan did not affect the kinetics of proBDNF cleavage by furin in vitro, indicating that effects other than a direct furin-proBDNF interaction may regulate proBDNF maturation. Using an optimized LC-MS/MS workflow, we demonstrate that secreted proBDNF is fully glycosylated and carries rare N-glycans terminated by GalNAcβ1-4GlcNAcβ1-R (LacdiNAc) extensively modified by terminal sulfation. We and others noted that this type of glycosylation is protein-specific, extends to proBDNF expressed in PC12 cells, and implies the presence of interacting partners that recognize this glycan epitope. The findings of our study reveal that proBDNF carries an unusual type of N-glycans important for its processing and secretion. Our results open new opportunities for functional studies of these protein glycoforms in different cells and tissues.
Collapse
Affiliation(s)
- Julius Benicky
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, D. C. 20057.,Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, D. C. 20057
| | - Miloslav Sanda
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, D. C. 20057.,Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, D. C. 20057
| | - Zuzana Brnakova Kennedy
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, D. C. 20057.,Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, D. C. 20057
| | - Radoslav Goldman
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, D. C. 20057 .,Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, D. C. 20057.,Clinical and Translational Glycoscience Research Center, Georgetown University, Washington, D. C. 20057
| |
Collapse
|