101
|
Bazard P, Pineros J, Frisina RD, Bauer MA, Acosta AA, Paganella LR, Borakiewicz D, Thivierge M, Mannering FL, Zhu X, Ding B. Cochlear Inflammaging in Relation to Ion Channels and Mitochondrial Functions. Cells 2021; 10:2761. [PMID: 34685743 PMCID: PMC8534887 DOI: 10.3390/cells10102761] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/06/2021] [Accepted: 10/12/2021] [Indexed: 12/20/2022] Open
Abstract
The slow accumulation of inflammatory biomarker levels in the body-also known as inflammaging-has been linked to a myriad of age-related diseases. Some of these include neurodegenerative conditions such as Parkinson's disease, obesity, type II diabetes, cardiovascular disease, and many others. Though a direct correlation has not been established, research connecting age-related hearing loss (ARHL)-the number one communication disorder and one of the most prevalent neurodegenerative diseases of our aged population-and inflammaging has gained interest. Research, thus far, has found that inflammatory markers, such as IL-6 and white blood cells, are associated with ARHL in humans and animals. Moreover, studies investigating ion channels and mitochondrial involvement have shown promising relationships between their functions and inflammaging in the cochlea. In this review, we summarize key findings in inflammaging within the auditory system, the involvement of ion channels and mitochondrial functions, and lastly discuss potential treatment options focusing on controlling inflammation as we age.
Collapse
Affiliation(s)
- Parveen Bazard
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (J.P.); (M.A.B.); (A.A.A.); (L.R.P.); (D.B.); (M.T.); (X.Z.); (B.D.)
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA;
| | - Jennifer Pineros
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (J.P.); (M.A.B.); (A.A.A.); (L.R.P.); (D.B.); (M.T.); (X.Z.); (B.D.)
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA;
| | - Robert D. Frisina
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (J.P.); (M.A.B.); (A.A.A.); (L.R.P.); (D.B.); (M.T.); (X.Z.); (B.D.)
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA;
- Department Communication Sciences and Disorders, College of Behavioral & Communication Sciences, Tampa, FL 33620, USA
- Morsani College of Medicine, University of South Florida, Tampa, FL 33620, USA
| | - Mark A. Bauer
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (J.P.); (M.A.B.); (A.A.A.); (L.R.P.); (D.B.); (M.T.); (X.Z.); (B.D.)
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA;
| | - Alejandro A. Acosta
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (J.P.); (M.A.B.); (A.A.A.); (L.R.P.); (D.B.); (M.T.); (X.Z.); (B.D.)
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA;
| | - Lauren R. Paganella
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (J.P.); (M.A.B.); (A.A.A.); (L.R.P.); (D.B.); (M.T.); (X.Z.); (B.D.)
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA;
| | - Dominika Borakiewicz
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (J.P.); (M.A.B.); (A.A.A.); (L.R.P.); (D.B.); (M.T.); (X.Z.); (B.D.)
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA;
| | - Mark Thivierge
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (J.P.); (M.A.B.); (A.A.A.); (L.R.P.); (D.B.); (M.T.); (X.Z.); (B.D.)
- Morsani College of Medicine, University of South Florida, Tampa, FL 33620, USA
| | - Freyda L. Mannering
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA;
- Morsani College of Medicine, University of South Florida, Tampa, FL 33620, USA
| | - Xiaoxia Zhu
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (J.P.); (M.A.B.); (A.A.A.); (L.R.P.); (D.B.); (M.T.); (X.Z.); (B.D.)
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA;
| | - Bo Ding
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (J.P.); (M.A.B.); (A.A.A.); (L.R.P.); (D.B.); (M.T.); (X.Z.); (B.D.)
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA;
| |
Collapse
|
102
|
Ribeiro AB, da Silva TM, Santos-Júnior NN, Castania JA, Fazan R, Salgado HC. Short-term effect of ligature-induced periodontitis on cardiovascular variability and inflammatory response in spontaneously hypertensive rats. BMC Oral Health 2021; 21:515. [PMID: 34635094 PMCID: PMC8507371 DOI: 10.1186/s12903-021-01885-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 09/30/2021] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND We previously reported that periodontal disease (PD) induces high arterial pressure variability (APV) consistent with sympathetic overactivity and elicits myocardial inflammation in Balb/c mice. However, it is unknown whether PD can change APV and heart rate variability (HRV) in spontaneously hypertensive (SHR) and normotensive Wistar-Kyoto (WKY) rats. This study aimed to evaluate the hemodynamic level, HRV, and APV associating with myocardial inflammation and plasma concentrations of oxide nitric (NO) in SHR and WKY rats with PD. METHODS Three weeks after bilateral ligation of the first mandibular molar, or Sham operation, the rats received catheters into the femoral artery and had their arterial pressure (AP) recorded the following day. Subsequently, plasma, heart, and jaw were collected. The NO was quantified by the chemiluminescence method in plasma, and the myocardial IL-1β concentrations were evaluated by ELISA. In the jaw was evaluated linear alveolar bone loss induced by PD. RESULTS The linear alveolar bone loss in jaws of SHR with PD was higher than in all other groups. AP and heart rate were higher in SHR than in their WKY counterparts. SHR with PD showed lower AP than control SHR. HRV and APV were different between SHR and WKY rats; however, no differences in these parameters were found between the animals with PD and their control counterparts. Plasma NO and myocardial IL-1β concentrations were higher in SHR with PD as compared to control WKY. A significant correlation was found between linear alveolar bone loss and plasma NO and myocardial IL-1β concentrations. CONCLUSION Our results demonstrated that short-term PD lowered the AP in SHR, which might be due to the higher levels of plasma NO. Even though PD did not affect either HRV or APV, it did induce myocardial inflammation, which can determine cardiovascular dysfunction in long-term PD.
Collapse
Affiliation(s)
- Aline Barbosa Ribeiro
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Avenida dos Bandeirantes, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Thais Marques da Silva
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Avenida dos Bandeirantes, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Nilton Nascimento Santos-Júnior
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Avenida dos Bandeirantes, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Jaci Airton Castania
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Avenida dos Bandeirantes, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Rubens Fazan
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Avenida dos Bandeirantes, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Helio Cesar Salgado
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Avenida dos Bandeirantes, Ribeirão Preto, São Paulo, 14049-900, Brazil.
| |
Collapse
|
103
|
Anthoulakis C, Mamopoulos A, Rousso D, Karagiannis A, Athanasiadis A, Grimbizis G, Athyros V. Arterial Stiffness as a Cardiovascular Risk Factor for the Development of Preeclampsia and Pharmacopreventive Options. Curr Vasc Pharmacol 2021; 20:52-61. [PMID: 34615450 DOI: 10.2174/1570161119666211006114258] [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: 05/20/2021] [Revised: 08/08/2021] [Accepted: 08/18/2021] [Indexed: 11/22/2022]
Abstract
Arterial stiffness (AS) describes the rigidity of the arterial walls. Epidemiological studies have shown that increased AS is an independent predictive marker of cardiovascular (CV) morbidity and mortality in both pregnant and non-pregnant women. Preeclampsia (PE), a form of pregnancy-induced hypertension, affects approximately 5% of pregnancies worldwide. Preeclamptic women have a higher risk of CV disease (CVD), mainly because PE damages the heart's ability to relax between contractions. Different pharmacological approaches for the prevention of PE have been tested in clinical trials (e.g. aspirin, enoxaparin, metformin, pravastatin, and sildenafil citrate). In current clinical practice, only low-dose aspirin is used for PE pharmacoprevention. However, low-dose aspirin does not prevent term PE, which is the most common form of PE. Compromised vascular integrity precedes the onset of PE and therefore, AS assessment may constitute a promising predictive marker of PE. Several non-invasive techniques have been developed to assess AS. Compared with normotensive pregnancies, both carotid-femoral pulse wave velocity (cfPWV) and augmentation index (AIx) are increased in PE. In view of simplicity, reliability, and reproducibility, there is an interest in oscillometric AS measurements in pregnancies complicated by PE.
Collapse
Affiliation(s)
- Christos Anthoulakis
- First Department of Obstetrics & Gynecology, "Papageorgiou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki. Greece
| | - Apostolos Mamopoulos
- Third Department of Obstetrics & Gynecology, Hippokration General Hospital, Aristotle University of Thessaloniki, Thessaloniki. Greece
| | - David Rousso
- Third Department of Obstetrics & Gynecology, Hippokration General Hospital, Aristotle University of Thessaloniki, Thessaloniki. Greece
| | - Asterios Karagiannis
- Second Propaedeutic Department of Internal Medicine, Hippokration General Hospital, Aristotle University of Thessaloniki, Thessaloniki. Greece
| | - Apostolos Athanasiadis
- Third Department of Obstetrics & Gynecology, Hippokration General Hospital, Aristotle University of Thessaloniki, Thessaloniki. Greece
| | - Grigoris Grimbizis
- First Department of Obstetrics & Gynecology, "Papageorgiou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki. Greece
| | - Vasilios Athyros
- Second Propaedeutic Department of Internal Medicine, Hippokration General Hospital, Aristotle University of Thessaloniki, Thessaloniki. Greece
| |
Collapse
|
104
|
Wu XW, Li G, Cheng XB, Wang M, Wang LL, Wang HH, Yang JY, Hu XJ. Association of Angiotensin II Type 1 Receptor Agonistic Autoantibodies With Outcomes in Patients With Acute Aortic Dissection. JAMA Netw Open 2021; 4:e2127587. [PMID: 34596673 PMCID: PMC8486983 DOI: 10.1001/jamanetworkopen.2021.27587] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
IMPORTANCE Angiotensin II is significantly associated with the pathogenesis of acute aortic dissection. Angiotensin II type 1 receptor agonistic autoantibodies (AT1-AAs) can mimic the effect of angiotensin II. OBJECTIVE To investigate the association between AT1-AAs and all-cause and cause-specific mortality risk in patients with acute aortic dissection. DESIGN, SETTING, AND PARTICIPANTS A total of 662 patients with clinically suspected aortic dissection from 3 medical centers in Wuhan, China, were enrolled in this cohort study from August 1, 2014, to July 31, 2016. Of these, 315 patients were included in the 3-year follow-up study. Follow-up was mainly performed via telephone interviews and outpatient clinic visits. Data analysis was conducted from March 1 to May 31, 2020. MAIN OUTCOMES AND MEASURES The primary outcomes of interest were all-cause mortality, death due to aortic dissection, and late aortic-related adverse events. RESULTS The full study cohort included 315 patients with AAD (mean [SD] age, 56.2 [12.7] years; 230 men [73.0%]). Ninety-two patients (29.2%) were positive for AT1-AAs. The mortality of AT1-AA-positive patients was significantly higher than that of AT1-AA-negative patients (40 [43.5%] vs 37 [16.6%]; P < .001). The mortality risk in AT1-AA-positive patients was always significantly higher than that in AT1-AA-negative patients in patients with both type A and type B dissection. Multivariable analysis showed that the risk of AT1-AA-positive patients for type A dissection was significantly higher than that of AT1-AA-negative patients (odds ratio [OR], 1.88; 95% CI, 1.12-3.13; P = .02). The Cox proportional hazards regression model showed a significant increase of all-cause mortality risk (OR, 2.27; 95% CI, 1.44-3.57; P < .001) and late aortic-related adverse events (OR, 1.58; 95% CI, 1.06-2.36; P = .03) among AT1-AA-positive patients during the follow-up period compared with AT1-AA-negative patients. CONCLUSIONS AND RELEVANCE This cohort study first detected AT1-AAs in patients with acute aortic dissection. The presence of AT1-AAs was associated with significantly higher all-cause and cause-specific mortality during a follow-up period of 3 years. The antibodies may be a risk factor for aortic dissection.
Collapse
Affiliation(s)
- Xiao-wei Wu
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Li
- Emergency Department, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
- Hubei Province Academy of Traditional Chinese Medicine, Wuhan, China
| | - Xiao-bin Cheng
- Emergency Department, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Min Wang
- Emergency Department, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Liu-lin Wang
- Emergency Department, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Hai-hao Wang
- Department of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-ye Yang
- Department of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing-jian Hu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
105
|
Podzolkov VI, Nebieridze NN, Safronova TA. Transforming Growth Factor-β1, Arterial Stiffness and Vascular Age in Patients With Uncontrolled Arterial Hypertension. Heart Lung Circ 2021; 30:1769-1777. [PMID: 34389253 DOI: 10.1016/j.hlc.2021.06.524] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/26/2021] [Accepted: 06/02/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Lack of blood pressure control leads to a higher incidence of hypertension-mediated target organ damage (HMOD). One of the markers of HMOD is an increased arterial stiffness, an independent predictor of cardiovascular complications. However, abstract numbers showing the level of arterial stiffness do not give patients a clear understanding of the risk of their condition. In order to increase patient compliance, the term "vascular age" (VA) was introduced. Arteriosclerosis plays the main role in increasing VA. The greatest interest, according to the literature, in the study of this issue is in arteriosclerosis caused by transforming growth factor β1 (TGF-β1)-the effect of TGF-β1 on the culture of smooth muscle cells leads to their proliferation and growth; also, TGF-β1 increases the amount of collagen and accelerates the degradation of elastin. METHODS We included 140 people in the study: 80 patients with controlled arterial hypertension (CAH), 30 with uncontrolled arterial hypertension (UAH), and 30 patients who formed the control group. All patients underwent a determination of arterial stiffness and VA using the cardio-ankle vascular index (CAVI), a corrected (blood-pressure independent) cardio-ankle vascular index (CAVI0) and the concentration of TGF-β1 was measured. RESULTS The TGF-β1 value in the UAH group was 22.6 (25th percentile=20.6; 75th percentile=25.6) ng/mL, and in the control group it was 17.4 (25th percentile=11.8; 75th percentile=19.3) ng/mL. In the CAH group, an intermediate value was noted-19.2 (25th percentile=17.2; 75th percentile=24.7) ng/mL. The CAVI in the UAH group was 9.2 (25th percentile=8.5; 75th percentile=9.9), in the control group-7 (25th percentile=6.5; 75th percentile=7.5). In the CAH group, the average CAVI was 7.8 (25th percentile=7.0; 75th percentile=8.5). The CAVI 0 in the UAH group was 14.8 (25th percentile=12.0; 75th percentile=15.6), in the control group - 9.7 (25th percentile=8.8; 75th percentile=9.7). In the CAH group, the average CAVI was 11.1 (25th percentile=10.1; 75th percentile=13.6). Vascular age in the UAH group was 71.5 (25th percentile=64; 75th percentile=74) years, in the CAH group 59 (25th percentile=49; 75th percentile=69) years, and in both groups (UAH, CAH), VA was significantly higher than the chronological age (p<0.05). In the control group, the VA did not significantly differ from the chronological age (p>0.05) and it was 54 (25th percentile=44; 75th percentile=59) years. A significant relationship was found between the TGF-β1 level and CAVI (CAH r=0.777; UAH r=0.753; p<0.05), CAVI 0 (CAH r=0.625; UAH r=0.502; p<0.05) and VA in patients with AH (CAH r=0.649; UAH r=0.753; p<0.05). CONCLUSION In patients in the UAH group, there was an increase in the concentration of TGF-β1, an increase in the arterial stiffness and in VA in comparison with patients in the CAH group and the control group. The relationship between TGF-β1 and the arterial stiffness and VA was revealed in patients with hypertension.
Collapse
Affiliation(s)
- Valery Ivanovich Podzolkov
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russian Federation
| | - Natia Nugzarovna Nebieridze
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russian Federation.
| | - Tatiana Arkadyevna Safronova
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russian Federation
| |
Collapse
|
106
|
Majid DSA, Mahaffey E, Castillo A, Prieto MC, Navar LG. Angiotensin II-induced renal angiotensinogen formation is enhanced in mice lacking tumor necrosis factor-alpha type 1 receptor. Physiol Rep 2021; 9:e14990. [PMID: 34427402 PMCID: PMC8383705 DOI: 10.14814/phy2.14990] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/30/2021] [Accepted: 07/11/2021] [Indexed: 01/11/2023] Open
Abstract
In hypertension induced by angiotensin II (AngII) administration with high salt (HS) intake, intrarenal angiotensinogen (AGT) and tumor necrosis factor-alpha (TNF-α) levels increase. However, TNF-α has been shown to suppress AGT formation in cultured renal proximal tubular cells. We examined the hypothesis that elevated AngII levels during HS intake reduces TNF-α receptor type 1 (TNFR1) activity in the kidneys, thus facilitating increased intrarenal AGT formation. The responses to HS diet (4% NaCl) with chronic infusion of AngII (25 ng/min) via implanted minipump for 4 weeks were assessed in wild-type (WT) and knockout (KO) mice lacking TNFR1 or TNFR2 receptors. Blood pressure was measured by tail-cuff plethysmography, and 24-h urine samples were collected using metabolic cages prior to start (0 day) and at the end of 2nd and 4th week periods. The urinary excretion rate of AGT (uAGT; marker for intrarenal AGT) was measured using ELISA. HS +AngII treatment for 4 weeks increased mean arterial pressure (MAP) in all strains of mice. However, the increase in MAP in TNFR1KO (77 ± 2 to 115 ± 3 mmHg; n = 7) was significantly greater (p < 0.01) than in WT (76 ± 1 to 102 ± 2 mmHg; n = 7) or in TNFR2KO (78 ± 2 to 99 ± 5 mmHg; n = 6). The increase in uAGT at 4th week was also greater (p < 0.05) in TNFR1KO mice (6 ± 2 to 167 ± 75 ng/24 h) than that in WT (6 ± 3 to 46 ± 16 ng/24 h) or in TNFR2KO mice (8 ± 7 to 65 ± 44 ng/24 h). The results indicate that TNFR1 exerts a protective role by mitigating intrarenal AGT formation induced by elevated AngII and HS intake.
Collapse
MESH Headings
- Angiotensin II/toxicity
- Angiotensinogen/metabolism
- Animals
- Blood Pressure
- Hypertension, Renal/etiology
- Hypertension, Renal/metabolism
- Kidney/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Receptors, Tumor Necrosis Factor, Type I/deficiency
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Receptors, Tumor Necrosis Factor, Type II/genetics
- Receptors, Tumor Necrosis Factor, Type II/metabolism
- Sodium Chloride, Dietary/toxicity
Collapse
Affiliation(s)
- Dewan S. A. Majid
- Department of PhysiologyHypertension & Renal Center of ExcellenceTulane University School of MedicineNew OrleansLouisianaUSA
| | - Eamonn Mahaffey
- Department of PhysiologyHypertension & Renal Center of ExcellenceTulane University School of MedicineNew OrleansLouisianaUSA
| | - Alexander Castillo
- Department of PhysiologyHypertension & Renal Center of ExcellenceTulane University School of MedicineNew OrleansLouisianaUSA
| | - Minolfa C. Prieto
- Department of PhysiologyHypertension & Renal Center of ExcellenceTulane University School of MedicineNew OrleansLouisianaUSA
| | - L. Gabriel Navar
- Department of PhysiologyHypertension & Renal Center of ExcellenceTulane University School of MedicineNew OrleansLouisianaUSA
| |
Collapse
|
107
|
Gunay S, Çalışkan S, Sigirli D. Inflammation and Nocturnal Pattern of Blood Pressure in Normotensives. INTERNATIONAL JOURNAL OF CARDIOVASCULAR SCIENCES 2021. [DOI: 10.36660/ijcs.20200298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
108
|
Shima Y, Kumasaka S, Negishi Y. Sustained sterile inflammation is related to pulmonary morbidities in premature infants. J Matern Fetal Neonatal Med 2021; 35:6928-6932. [PMID: 34266359 DOI: 10.1080/14767058.2021.1931102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Sterile inflammation, initiated by endogenous molecules such as high-mobility group box-1 (HMGB1), has come to be recognized as a critical mechanism in a variety of chronic diseases. To elucidate the involvement of sterile inflammation in neonatal disease, the association between serum HMGB1 levels and the development of bronchopulmonary dysplasia (BPD) was evaluated. STUDY DESIGN Serum HMGB1 levels were measured in 25 premature infants born before 33 weeks of gestation, excluding any infection cases. Samples were collected at birth, two, and four weeks of age and compared according to BPD status. RESULTS The serum HMGB1 levels in infants with BPD were maintained up to 4 weeks of age, while those without BPD declined with time. Postnatal cardiopulmonary and nutritional transition was delayed in infants with BPD. CONCLUSION Sustained elevation of serum HMGB1 levels was associated with the development of BPD, suggesting that prolonged sterile inflammation may contribute to lung injury.
Collapse
Affiliation(s)
- Yoshio Shima
- Department of Neonatal Medicine, Nippon Medical School, Musashikosugi Hospital, Kanagawa, Japan.,Department of Pediatrics, Japanese Red Cross Maternity Hospital, Tokyo, Japan
| | - Sakae Kumasaka
- Department of Pediatrics, Japanese Red Cross Maternity Hospital, Tokyo, Japan
| | - Yasuyuki Negishi
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
109
|
Ranaweera SS, Dissanayake CY, Natraj P, Lee YJ, Han CH. Anti-inflammatory effect of sulforaphane on LPS-stimulated RAW 264.7 cells and ob/ob mice. J Vet Sci 2021; 21:e91. [PMID: 33263238 PMCID: PMC7710464 DOI: 10.4142/jvs.2020.21.e91] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/22/2020] [Accepted: 10/28/2020] [Indexed: 12/28/2022] Open
Abstract
Background Sulforaphane (SFN) is an isothiocyanate compound present in cruciferous vegetables. Although the anti-inflammatory effects of SFN have been reported, the precise mechanism related to the inflammatory genes is poorly understood. Objectives This study examined the relationship between the anti-inflammatory effects of SFN and the differential gene expression pattern in SFN treated ob/ob mice. Methods Nitric oxide (NO) level was measured using a Griess assay. The inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) expression levels were analyzed by Western blot analysis. Pro-inflammatory cytokines (tumor necrosis factor [TNF]-α, interleukin [IL]-1β, and IL-6) were measured by enzyme-linked immunosorbent assay (ELISA). RNA sequencing analysis was performed to evaluate the differential gene expression in the liver of ob/ob mice. Results The SFN treatment significantly attenuated the iNOS and COX-2 expression levels and inhibited NO, TNF-α, IL-1β, and IL-6 production in lipopolysaccharide (LPS)-stimulated RAW 264.7 cells. RNA sequencing analysis showed that the expression levels of 28 genes related to inflammation were up-regulated (> 2-fold), and six genes were down-regulated (< 0.6-fold) in the control ob/ob mice compared to normal mice. In contrast, the gene expression levels were restored to the normal level by SFN. The protein-protein interaction (PPI) network showed that chemokine ligand (Cxcl14, Ccl1, Ccl3, Ccl4, Ccl17) and chemokine receptor (Ccr3, Cxcr1, Ccr10) were located in close proximity and formed a “functional cluster” in the middle of the network. Conclusions The overall results suggest that SFN has a potent anti-inflammatory effect by normalizing the expression levels of the genes related to inflammation that were perturbed in ob/ob mice.
Collapse
Affiliation(s)
| | | | - Premkumar Natraj
- College of Veterinary Medicine, Jeju National University, Jeju 63243, Korea
| | - Young Jae Lee
- College of Veterinary Medicine, Jeju National University, Jeju 63243, Korea
| | - Chang Hoon Han
- College of Veterinary Medicine, Jeju National University, Jeju 63243, Korea.
| |
Collapse
|
110
|
Meynard C, Mansuet-Lupo A, Giraud N, Boulle G, Imbault P, Guénégou-Arnoux A, Bobbio A, Durdux C, Damotte D, Giraud P. Size and Predictive Factors of Microscopic Tumor Extension in Locally Advanced Non-Small Cell Lung Cancer. Pract Radiat Oncol 2021; 11:491-501. [PMID: 34126295 DOI: 10.1016/j.prro.2021.05.006] [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: 12/28/2020] [Revised: 05/21/2021] [Accepted: 05/27/2021] [Indexed: 12/25/2022]
Abstract
PURPOSE Radiation therapy for locally advanced non-small cell lung cancer (NSCLC) should treat the whole tumor, including its microscopic extensions, and protect adjacent organs at risk as much as possible. The aim of our study is to evaluate the size of microscopic tumor extension (MEmax) in NSCLC, and search for potential predictive factors. METHODS AND MATERIALS We retrospectively selected 70 patients treated with postoperative radiation therapy for a NSCLC with N2 nodal status, then 34 additional patients operated for a squamous cell lung cancer with N1 or N2 nodal status. On the digitized slides originating from the resected tumors of these 104 patients, we outlined the border of the tumor, as seen with the naked eye. We then searched for microscopic tumor extension outside of these borders with a magnification as high as 40 × and measured the maximum size of MEmax. RESULTS The median MEmax in the whole cohort was 0.85 mm (0-9.95). The MEmax was <5.3 mm in 95% of adenocarcinomas (6.5 mm in the subgroup without neoadjuvant chemotherapy) and <3.5 mm in 95% of squamous cell carcinomas (3.7 mm in the subgroup without neoadjuvant chemotherapy). After multivariate analysis, the factors associated with the size of MEmax were vascular invasion (P = .0002), histologic type, with a wider MEmax for adenocarcinomas in comparison with squamous cell carcinomas (P = .002), tumor size, which was inversely related with the size of MEmax (P = .024), and high blood pressure (P = .03). Macroscopic histologic tumor size was well correlated with both radiologic tumor size on a mediastinal setting computed tomography (correlation coefficient of 0.845) and on a parenchymal setting computed tomography (correlation coefficient of 0.836). CONCLUSIONS The clinical target volume margin, accounting for microscopic tumoral extension, could be reduced to 7 mm for adenocarcinomas and 4 mm for squamous cell carcinomas.
Collapse
Affiliation(s)
- Claire Meynard
- Radiation Oncology unit, Assistance Publique-Hôpitaux de Paris, Georges Pompidou European Hospital, Paris University, Paris, France.
| | - Audrey Mansuet-Lupo
- Department of Pathology, Assistance Publique-Hôpitaux de Paris, Cochin Hospital, Paris University, Paris, France; Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, Paris, France
| | - Nicolas Giraud
- Radiation Oncology Department, Hôpital Haut-Lévêque, CHU Bordeaux, Pessac, France
| | - Geoffroy Boulle
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, Paris, France
| | - Paul Imbault
- Clinical Research Unit, Assistance Publique-Hôpitaux de Paris, Georges Pompidou European Hospital, Paris University, Paris, France
| | - Armelle Guénégou-Arnoux
- Clinical Research Unit, Assistance Publique-Hôpitaux de Paris, Georges Pompidou European Hospital, Paris University, Paris, France; INSERM CIC1418-EC, Georges Pompidou European Hospital, Paris, France
| | - Antonio Bobbio
- Department of Thoracic Surgery, Assistance Publique - Hôpitaux de Paris, Cochin Hospital, Paris University, Paris, France
| | - Catherine Durdux
- Radiation Oncology unit, Assistance Publique-Hôpitaux de Paris, Georges Pompidou European Hospital, Paris University, Paris, France
| | - Diane Damotte
- Department of Pathology, Assistance Publique-Hôpitaux de Paris, Cochin Hospital, Paris University, Paris, France; Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, Paris, France
| | - Philippe Giraud
- Radiation Oncology unit, Assistance Publique-Hôpitaux de Paris, Georges Pompidou European Hospital, Paris University, Paris, France
| |
Collapse
|
111
|
Choi MR, Fernández BE. Protective Renal Effects of Atrial Natriuretic Peptide: Where Are We Now? Front Physiol 2021; 12:680213. [PMID: 34135773 PMCID: PMC8202499 DOI: 10.3389/fphys.2021.680213] [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: 03/13/2021] [Accepted: 04/29/2021] [Indexed: 12/11/2022] Open
Abstract
Atrial natriuretic peptide belongs to the family of natriuretic peptides, a system with natriuretic, diuretic, and vasodilator effects that opposes to renin-angiotensin system. In addition to its classic actions, atrial natriuretic peptide exerts a nephroprotective effect given its antioxidant and anti-inflammatory properties, turning it as a beneficial agent against acute and chronic kidney diseases. This minireview describes the most relevant aspects of atrial natriuretic peptide in the kidney, including its renal synthesis, physiological actions through specific receptors, the importance of its metabolism, and its potential use in different pathological scenarios.
Collapse
Affiliation(s)
- Marcelo Roberto Choi
- Instituto Alberto C. Taquini de Investigaciones en Medicina Traslacional (IATIMET), CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Ciencias Biológicas, Facultad de Farmacia y Bioquímica, Cátedra de Anatomía e Histología, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto Universitario de Ciencias de la Salud, Fundación H.A. Barceló, Buenos Aires, Argentina
| | | |
Collapse
|
112
|
Liberale L, Montecucco F, Tardif JC, Libby P, Camici GG. Inflamm-ageing: the role of inflammation in age-dependent cardiovascular disease. Eur Heart J 2021; 41:2974-2982. [PMID: 32006431 DOI: 10.1093/eurheartj/ehz961] [Citation(s) in RCA: 189] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 09/10/2019] [Accepted: 01/09/2020] [Indexed: 12/12/2022] Open
Abstract
The ongoing worldwide increase in life expectancy portends a rising prevalence of age-related cardiovascular (CV) diseases in the coming decades that demands a deeper understanding of their molecular mechanisms. Inflammation has recently emerged as an important contributor for CV disease development. Indeed, a state of chronic sterile low-grade inflammation characterizes older organisms (also known as inflamm-ageing) and participates pivotally in the development of frailty, disability, and most chronic degenerative diseases including age-related CV and cerebrovascular afflictions. Due to chronic activation of inflammasomes and to reduced endogenous anti-inflammatory mechanisms, inflamm-ageing contributes to the activation of leucocytes, endothelial, and vascular smooth muscle cells, thus accelerating vascular ageing and atherosclerosis. Furthermore, inflamm-ageing promotes the development of catastrophic athero-thrombotic complications by enhancing platelet reactivity and predisposing to plaque rupture and erosion. Thus, inflamm-ageing and its contributors or molecular mediators might furnish targets for novel therapeutic strategies that could promote healthy ageing and conserve resources for health care systems worldwide. Here, we discuss recent findings in the pathophysiology of inflamm-ageing, the impact of these processes on the development of age-related CV diseases, results from clinical trials targeting its components and the potential implementation of these advances into daily clinical practice.
Collapse
Affiliation(s)
- Luca Liberale
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren CH-8952, Switzerland.,Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa, v.le Benedetto XV 10, 16132 Genoa, Italy
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, L.go Rosanna Benzi 10, 16132 Genoa, Italy.,First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, v.le Benedetto XV 10, 16132 Genoa, Italy
| | - Jean-Claude Tardif
- Montreal Heart Institute, Université de Montreal, Rue Bélanger 5000, Montreal, QC H1T 1C8, Canada
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Francis Street 75, Boston, MA 02115, USA
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren CH-8952, Switzerland.,Department of Cardiology, University Heart Center, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland.,Department of Research and Education, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| |
Collapse
|
113
|
Modulations of Cardiac Functions and Pathogenesis by Reactive Oxygen Species and Natural Antioxidants. Antioxidants (Basel) 2021; 10:antiox10050760. [PMID: 34064823 PMCID: PMC8150787 DOI: 10.3390/antiox10050760] [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] [Received: 04/11/2021] [Revised: 05/03/2021] [Accepted: 05/08/2021] [Indexed: 01/11/2023] Open
Abstract
Homeostasis in the level of reactive oxygen species (ROS) in cardiac myocytes plays a critical role in regulating their physiological functions. Disturbance of balance between generation and removal of ROS is a major cause of cardiac myocyte remodeling, dysfunction, and failure. Cardiac myocytes possess several ROS-producing pathways, such as mitochondrial electron transport chain, NADPH oxidases, and nitric oxide synthases, and have endogenous antioxidation mechanisms. Cardiac Ca2+-signaling toolkit proteins, as well as mitochondrial functions, are largely modulated by ROS under physiological and pathological conditions, thereby producing alterations in contraction, membrane conductivity, cell metabolism and cell growth and death. Mechanical stresses under hypertension, post-myocardial infarction, heart failure, and valve diseases are the main causes for stress-induced cardiac remodeling and functional failure, which are associated with ROS-induced pathogenesis. Experimental evidence demonstrates that many cardioprotective natural antioxidants, enriched in foods or herbs, exert beneficial effects on cardiac functions (Ca2+ signal, contractility and rhythm), myocytes remodeling, inflammation and death in pathological hearts. The review may provide knowledge and insight into the modulation of cardiac pathogenesis by ROS and natural antioxidants.
Collapse
|
114
|
Periodontitis, Blood Pressure, and the Risk and Control of Arterial Hypertension: Epidemiological, Clinical, and Pathophysiological Aspects-Review of the Literature and Clinical Trials. Curr Hypertens Rep 2021; 23:27. [PMID: 33961166 PMCID: PMC8105217 DOI: 10.1007/s11906-021-01140-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2021] [Indexed: 12/11/2022]
Abstract
Purpose of Review Arterial hypertension is an important risk factor for cardiovascular disease. In the world, about 45% of people suffer from arterial hypertension, while good blood pressure control is achieved by only approximately 50% of all hypertensive patients treated. The reason for the high prevalence of arterial hypertension and its poor control is low knowledge of hypertensinogenic factors. One such factor is periodontitis, which is a disease of social importance. Recent Findings It has been shown that the occurrence of periodontitis leads to an increase in blood pressure, increasing the risk of arterial hypertension. Periodontitis can also lead to ineffectiveness of antihypertensive treatment. Some interventional studies have shown that treatment of periodontitis reduced blood pressure in patients with arterial hypertension. The pathogenesis of arterial hypertension in periodontitis is complex and concerns mainly the impairment of the vasodilatation properties of the endothelium. Summary Hygiene and periodontitis treatment should be a method of preventing arterial hypertension and a method of increasing the effectiveness of antihypertensive treatment.
Collapse
|
115
|
Moreno JM, Martinez CM, de Jodar C, Reverte V, Bernabé A, Salazar FJ, Llinás MT. Gender differences in the renal changes induced by a prolonged high-fat diet in rats with altered renal development. J Physiol Biochem 2021; 77:431-441. [PMID: 33851366 DOI: 10.1007/s13105-021-00815-y] [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: 11/04/2020] [Accepted: 04/03/2021] [Indexed: 10/21/2022]
Abstract
The mechanisms involved in renal dysfunction induced by high-fat diet (HFD) in subjects with altered renal development (ARDev) are understudied. The objective of this study is to examine whether there are sex-dependent differences in the mechanisms involved in the hypertension and deterioration of renal function in SD rats with prolonged HFD and ARDev. The role of angiotensin II (Ang II) in the arterial pressure (AP) increments, the renal hemodynamic sensitivity to Ang II, glomerular damage and changes in fat abdominal volume, plasma adipokine levels, renal NADPHp67phox expression, and renal infiltration of immune cells were examined. Hypertension and deterioration of renal function were enhanced (P < 0.05) in both sexes of rats with HFD and ARDev. The decrease (P < 0.05) of AP elicited by candesartan in hypertensive rats was similar to that induced by the simultaneous administration of candesartan and apocynin. The greater (P < 0.05) renal vasoconstriction induced by Ang II in both sexes of rats with HFD and ARDev was accompanied by an enhanced (P < 0.05) infiltration of CD-3 cells and macrophages in the renal cortex and renal medulla. The increments (P < 0.05) in the renal expression of NADPHp67phox and glomeruloesclerosis were greater (P < 0.05) in males than in females with HFD and ARDev. Our results suggest that the hypertension and deterioration of renal function induced by HFD in rats with ARDev are Ang II-dependent and mediated by increments in oxidative stress and immune system activation. Sex-dependent increments in oxidative stress and glomerular damage may contribute to the deterioration of renal function in these rats.
Collapse
Affiliation(s)
- Juan M Moreno
- Department of Physiology, School of Medicine, University of Murcia, 30100, Murcia, Spain.,Biomedical Research Institute in Murcia, Murcia, Spain
| | | | - Carlos de Jodar
- Department of Pathology, School of Veterinary, University of Murcia, Murcia, Spain
| | - Virginia Reverte
- Department of Physiology, School of Medicine, University of Murcia, 30100, Murcia, Spain.,Biomedical Research Institute in Murcia, Murcia, Spain
| | - Antonio Bernabé
- Department of Pathology, School of Veterinary, University of Murcia, Murcia, Spain
| | - F Javier Salazar
- Department of Physiology, School of Medicine, University of Murcia, 30100, Murcia, Spain. .,Biomedical Research Institute in Murcia, Murcia, Spain.
| | - María T Llinás
- Department of Physiology, School of Medicine, University of Murcia, 30100, Murcia, Spain.,Biomedical Research Institute in Murcia, Murcia, Spain
| |
Collapse
|
116
|
van Dorst DC, Dobbin SJ, Neves KB, Herrmann J, Herrmann SM, Versmissen J, Mathijssen RH, Danser AJ, Lang NN. Hypertension and Prohypertensive Antineoplastic Therapies in Cancer Patients. Circ Res 2021; 128:1040-1061. [PMID: 33793337 PMCID: PMC8011349 DOI: 10.1161/circresaha.121.318051] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The development of a wide range of novel antineoplastic therapies has improved the prognosis for patients with a wide range of malignancies, which has increased the number of cancer survivors substantially. Despite the oncological benefit, cancer survivors are exposed to short- and long-term adverse cardiovascular toxicities associated with anticancer therapies. Systemic hypertension, the most common comorbidity among cancer patients, is a major contributor to the increased risk for developing these adverse cardiovascular events. Cancer and hypertension have common risk factors, have overlapping pathophysiological mechanisms and hypertension may also be a risk factor for some tumor types. Many cancer therapies have prohypertensive effects. Although some of the mechanisms by which these antineoplastic agents lead to hypertension have been characterized, further preclinical and clinical studies are required to investigate the exact pathophysiology and the optimal management of hypertension associated with anticancer therapy. In this way, monitoring and management of hypertension before, during, and after cancer treatment can be improved to minimize cardiovascular risks. This is vital to optimize cardiovascular health in patients with cancer and survivors, and to ensure that advances in terms of cancer survivorship do not come at the expense of increased cardiovascular toxicities.
Collapse
Affiliation(s)
- Daan C.H. van Dorst
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (D.C.H.v.D., J.V., A.H.J.D.), Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Medical Oncology, Erasmus MC Cancer Institute (D.C.H.v.D., R.H.J.M.), Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Stephen J.H. Dobbin
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (S.J.H.D., K.B.N., N.N.L.)
| | - Karla B. Neves
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (S.J.H.D., K.B.N., N.N.L.)
| | - Joerg Herrmann
- Department of Cardiovascular Medicine (J.H.), Mayo Clinic, Rochester, MN
| | - Sandra M. Herrmann
- Division of Nephrology and Hypertension (S.M.H.), Mayo Clinic, Rochester, MN
| | - Jorie Versmissen
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (D.C.H.v.D., J.V., A.H.J.D.), Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Hospital Pharmacy (J.V.), Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Ron H.J. Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute (D.C.H.v.D., R.H.J.M.), Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - A.H. Jan Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (D.C.H.v.D., J.V., A.H.J.D.), Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Ninian N. Lang
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (S.J.H.D., K.B.N., N.N.L.)
| |
Collapse
|
117
|
Regulatory T-cell subset distribution in children with primary hypertension is associated with hypertension severity and hypertensive target organ damage. J Hypertens 2021; 38:692-700. [PMID: 31834124 DOI: 10.1097/hjh.0000000000002328] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND The relationship between circulating regulatory T-cell (Tregs) subset distribution and hypertension severity in children with primary hypertension is not known. We aimed to find out if target organ damage (TOD) in children with primary hypertension is related to defects in Tregs distribution reflected by their phenotype characteristics. METHODS The study constituted 33 nontreated hypertensive children and 35 sex-matched and age-matched controls. Using multicolor flow cytometry technique, we assessed a distribution of the total Tregs (CD4CD25CD127) and their subsets (CD45RA-naive Tregs, CD45RA memory/activated Tregs, CD45RACD31 recent thymic emigrants Tregs and mature naive CD45RACD31 Tregs) in the whole blood. RESULTS Hypertensive children showed decreased percentage of the total Tregs, the CD45RA-naive Tregs, the total CD31 Tregs and the recent thymic emigrants Tregs but elevation of the CD45RA memory/activated Treg and mature naive CD45RACD31 Tregs. Decreased frequency of the total Tregs, naive Tregs and CD31-bearing Treg cell subsets (CD31 total Tregs, CD45RACD31 recent thymic emigrants Tregs) negatively correlated to TOD markers, arterial stiffness and blood pressure elevation. In contrast, increased percentage of memory Tregs and CD31 Tregs subsets positively correlated to organ damage markers, arterial stiffness and blood pressure values. These changes were independent of BMI, age, sex and hsCRP. CONCLUSION Both diagnosis of hypertension, TOD and arterial stiffness in hypertensive children were associated with decreased population of total CD4 Tregs, limited output of recent thymic emigrants Tregs, and increased pool of activated/memory Tregs. Hypertension was an independent predictor of the circulating Treg subsets distribution irrespective of hsCRP.
Collapse
|
118
|
The association of novel inflammatory marker GlycA and incident atrial fibrillation in the Multi-Ethnic Study of Atherosclerosis (MESA). PLoS One 2021; 16:e0248644. [PMID: 33765041 PMCID: PMC7993599 DOI: 10.1371/journal.pone.0248644] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 02/11/2021] [Indexed: 01/07/2023] Open
Abstract
Background Emerging evidence has implicated that inflammation contributes to the pathogenesis of atrial fibrillation (AF). GlycA is a novel marker of systemic inflammation with low intra-individual variability and high analytic precision. GlycA has been associated with incident cardiovascular disease (CVD) independent of other inflammatory markers. However, whether GlycA is associated with AF, specifically, has yet to be established. We examined the association between GlycA and AF in a multi-ethnic cohort. Methods We studied 6,602 MESA participants aged 45–85, with no clinical CVD at baseline, with data on GlycA and incident AF. We used multivariable-adjusted Cox models to evaluate the association between GlycA and incident AF. We also examined other inflammatory markers [high-sensitivity C-reactive protein (hsCRP), interleukin-6 (IL6) and fibrinogen] and incident AF for comparison. Results The mean (SD) age was 62 (10) years, 53% women. The mean plasma GlycA was 381 (62) μmol/L. Over median follow-up of 12.9 years, 869 participants experienced AF. There was no statistically significant association between GlycA and incident AF after adjusting for sociodemographics, CVD risk factors, and other inflammatory markers [Hazard Ratio (95% CI) per 1 SD increment in GlycA: 0.97 (0.88–1.06)]. Neither hsCRP nor fibrinogen was associated with incident AF in same model. In contrast, IL-6 was independently associated with incident AF [HR 1.12 per 1 SD increment (1.05–1.19)]. Conclusions Although GlycA has been associated with other CVD types, we found that GlycA was not associated with AF. More research will be required to understand why IL-6 was associated with AF but not GlycA. Clinical trial registration MESA is not a clinical trial. However, the cohort is registered at: URL: https://clinicaltrials.gov/ct2/show/NCT00005487 Unique identifier: NCT00005487.
Collapse
|
119
|
Jung R, Wild J, Ringen J, Karbach S, Wenzel P. Innate Immune Mechanisms of Arterial Hypertension and Autoimmune Disease. Am J Hypertens 2021; 34:143-153. [PMID: 32930786 DOI: 10.1093/ajh/hpaa145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/15/2020] [Accepted: 09/14/2020] [Indexed: 12/16/2022] Open
Abstract
The immune system is indispensable in the development of vascular dysfunction and hypertension. The interplay between immune cells and the vasculature, kidneys, heart, and blood pressure regulating nuclei in the central nervous system results in a complex and closely interwoven relationship of the immune system with arterial hypertension. A better understanding of this interplay is necessary for optimized and individualized antihypertensive therapy. Our review article focuses on innate cells in hypertension and to what extent they impact on development and preservation of elevated blood pressure. Moreover, we address the association of hypertension with chronic autoimmune diseases. The latter are ideally suited to learn about immune-mediated mechanisms in cardiovascular disease leading to high blood pressure.
Collapse
Affiliation(s)
- Rebecca Jung
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Johannes Wild
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
- Department of Cardiology, University Medical Center Mainz, Mainz, Germany
| | - Julia Ringen
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Susanne Karbach
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
- Department of Cardiology, University Medical Center Mainz, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), partner site Rhine-Main, Germany
| | - Philip Wenzel
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
- Department of Cardiology, University Medical Center Mainz, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), partner site Rhine-Main, Germany
| |
Collapse
|
120
|
Venkatakrishnan K, Chiu HF, Wang CK. Impact of functional foods and nutraceuticals on high blood pressure with a special focus on meta-analysis: review from a public health perspective. Food Funct 2021; 11:2792-2804. [PMID: 32248209 DOI: 10.1039/d0fo00357c] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In recent times many researchers are expressing immense interest in nutraceuticals and functional foods for combating various diseases or abnormal conditions, especially against hypertension (HT). Persistent HT is medically referred to as chronic high blood pressure (BP) and considered to be one of the major risk factors for the deadliest diseases including cardiovascular disease (CVD) and cerebrovascular diseases. Hence HT poses a serious socio-economic burden worldwide, particularly to developing countries. The current treatment strategy for HT includes standard anti-hypertensive drugs, which are associated with many adverse effects and lower drug adherence rates. Therefore, an alternative or complementary natural therapy (functional foods or nutraceuticals or dietary supplements) would be the alternate choice along with a modified lifestyle pattern that might help to manage or combat HT and its related complications. During this review, the author would like to shed light on the basic science behind HT including pathophysiology and the impact of dietary salt on HT and the impact of various functional foods or nutraceuticals against HT in humans (meta-analysis and systemic review). This contribution gives a better idea (public health perspective) for choosing the best functional foods/nutraceuticals for the prevention, management or delaying the onset of HT and its associated conditions along with modified lifestyle patterns and standard anti-hypertensive drugs.
Collapse
Affiliation(s)
- Kamesh Venkatakrishnan
- School of Nutrition, Chung Shan Medical University, 110, Sec. 1, Jianguo North Road, Taichung City-40201, Taiwan, Republic of China.
| | - Hui-Fang Chiu
- Department of Chinese Medicine, Taichung Hospital Ministry of Health and Welfare, Taichung-40301, Taiwan, Republic of China
| | - Chin-Kun Wang
- School of Nutrition, Chung Shan Medical University, 110, Sec. 1, Jianguo North Road, Taichung City-40201, Taiwan, Republic of China.
| |
Collapse
|
121
|
Chen XJ, Zhang H, Yang F, Liu Y, Chen G. DNA Methylation Sustains "Inflamed" Memory of Peripheral Immune Cells Aggravating Kidney Inflammatory Response in Chronic Kidney Disease. Front Physiol 2021; 12:637480. [PMID: 33737884 PMCID: PMC7962671 DOI: 10.3389/fphys.2021.637480] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 02/04/2021] [Indexed: 01/19/2023] Open
Abstract
The incidence of chronic kidney disease (CKD) has rapidly increased in the past decades. A progressive loss of kidney function characterizes a part of CKD even with intensive supportive treatment. Irrespective of its etiology, CKD progression is generally accompanied with the development of chronic kidney inflammation that is pathologically featured by the low-grade but chronic activation of recruited immune cells. Cumulative evidence support that aberrant DNA methylation pattern of diverse peripheral immune cells, including T cells and monocytes, is closely associated with CKD development in many chronic disease settings. The change of DNA methylation profile can sustain for a long time and affect the future genes expression in the circulating immune cells even after they migrate from the circulation into the involved kidney. It is of clinical interest to reveal the underlying mechanism of how altered DNA methylation regulates the intensity and the time length of the inflammatory response in the recruited effector cells. We and others recently demonstrated that altered DNA methylation occurs in peripheral immune cells and profoundly contributes to CKD development in systemic chronic diseases, such as diabetes and hypertension. This review will summarize the current findings about the influence of aberrant DNA methylation on circulating immune cells and how it potentially determines the outcome of CKD.
Collapse
Affiliation(s)
- Xiao-Jun Chen
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Hong Zhang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Fei Yang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Yu Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Guochun Chen
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| |
Collapse
|
122
|
Martin JH, Head R. A pharmacological framework for integrating treating the host, drug repurposing and the damage response framework in COVID-19. Br J Clin Pharmacol 2021; 87:875-885. [PMID: 32959913 PMCID: PMC7646655 DOI: 10.1111/bcp.14551] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 08/27/2020] [Accepted: 09/02/2020] [Indexed: 12/19/2022] Open
Abstract
With any new disease a framework for the development of preventative or treatment therapeutics is key; the absence of such in COVID-19 has enabled ineffective and potentially unsafe treatments to be taken up by governments and clinicians desperate to have options for patients. As we still have few therapies and nil vaccines yet available, the void of a clear framework for research and practice is increasingly clear. We describe a framework that has been used to prioritise therapeutic research in previous pandemics which could be used to progress clinical pharmacology and therapeutics research in COVID-19. This is particularly relevant as discussion has already moved on from antiviral therapeutics to delineating the treatment of the host from treatment and elimination of the infective agent. Focussing on the host brings together three concepts: host treatment, the damage response framework and therapeutic repurposing. The integration of these three areas plays to the traditional strength of pharmaceuticals in providing a period of stabilization to permit time for the development of novel antiviral drugs and vaccines. In integrating approaches to repurposing, host treatment and damage response we identified three key properties that a potentially effective repurposed drug must possess by way of a framework. There must be homology, i.e., the same or similar relation with the pathogenesis of the disease, ideally targeted to the conserved pathophysiological outcomes of the viral attack; there must be a defined locus within the spectrum to prevention to severe disease and the framework must draw upon the historical dose and safety experience of the repurposed drug. To illustrate, we have mapped therapeutics that impact upon a key dysregulated pathway in COVID-19 - the renin angiotensin system - using this approach. Collectively this type of analysis reveals the importance of existing data (repurposed information and administrative observational data) and the importance of the details of the known pathophysiological response to viruses in approaches to treating the host.
Collapse
Affiliation(s)
- Jennifer H. Martin
- Centre for Human Drug Repurposing and Medicines ResearchUniversity of NewcastleNSW2305Australia
| | - Richard Head
- University of South AustraliaSouth AustraliaAustralia
| |
Collapse
|
123
|
Basile DP, Abais-Battad JM, Mattson DL. Contribution of Th17 cells to tissue injury in hypertension. Curr Opin Nephrol Hypertens 2021; 30:151-158. [PMID: 33394732 PMCID: PMC8221512 DOI: 10.1097/mnh.0000000000000680] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Hypertension has been demonstrated to be a chief contributor to morbidity and mortality throughout the world. Although the cause of hypertension is multifactorial, emerging evidence, obtained in experimental studies, as well as observational studies in humans, points to the role of inflammation and immunity. Many aspects of immune function have now been implicated in hypertension and end-organ injury; this review will focus upon the recently-described role of Th17 cells in this pathophysiological response. RECENT FINDINGS Studies in animal models and human genetic studies point to a role in the adaptive immune system as playing a contributory role in hypertension and renal tissue damage. Th17 cells, which produce the cytokine IL17, are strongly pro-inflammatory cells, which may contribute to tissue damage if expressed in chronic disease conditions. The activity of these cells may be enhanced by physiological factors associated with hypertension such as dietary salt or Ang II. This activity may culminate in the increased sodium retaining activity and exacerbation of inflammation and renal fibrosis via multiple cellular mechanisms. SUMMARY Th17 cells are a distinct component of the adaptive immune system that may strongly enhance pathways leading to increased sodium reabsorption, elevated vascular tone and end-organ damage. Moreover, this pathway may lend itself towards specific targeting for treatment of kidney disease and hypertension.
Collapse
Affiliation(s)
- David P Basile
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | | | - David L Mattson
- Department of Physiology, Medical College of Georgia, Augusta, Georgia, USA
| |
Collapse
|
124
|
Stanley CP, Stocker R. Regulation of vascular tone and blood pressure by singlet molecular oxygen in inflammation. Curr Opin Nephrol Hypertens 2021; 30:145-150. [PMID: 33427761 DOI: 10.1097/mnh.0000000000000679] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW The principle aim of this review is to prompt vascular researchers interested in vascular inflammation and oxidative stress to consider singlet molecular oxygen (1O2) as a potentially relevant contributor. A secondary goal is to propose novel treatment strategies to address haemodynamic complications associated with septic shock. RECENT FINDINGS Increased inflammation and oxidative stress are hallmarks of a range of vascular diseases. We recently showed that in systemic inflammation and oxidative stress associated with models of inflammation including sepsis, the tryptophan catabolizing enzyme indoleamine 2,3-dioxygenase-1 (Ido1) contributes to hypotension and decreased blood pressure through production of singlet molecular oxygen (1O2). Once formed, 1O2 converts tryptophan bound to Ido1 to a vasoactive hydroperoxide which decreases arterial tone and blood pressure via oxidation of a specific cysteine residue of protein kinase G1α. SUMMARY These works show, for the first time, that 1O2 contributes to arterial redox signalling and that Ido1 contributes to the regulation of blood pressure through production of a novel tryptophan-derived hydroperoxide, thus presenting a new signalling pathway as novel target in the treatment of blood pressure disorders such as sepsis.
Collapse
Affiliation(s)
- Christopher P Stanley
- Heart Research Institute, Newtown
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Roland Stocker
- Heart Research Institute, Newtown
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
125
|
Santos WA, Dourado KMC, Araújo FA, Jesus RLC, Moraes RA, Oliveira SCDS, Alves QL, Simões LO, Casais-E-Silva LL, Costa RS, Velozo ES, Silva DF. Braylin induces a potent vasorelaxation, involving distinct mechanisms in superior mesenteric and iliac arteries of rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2021; 394:437-446. [PMID: 33034715 DOI: 10.1007/s00210-020-01985-0] [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] [Received: 05/12/2020] [Accepted: 10/04/2020] [Indexed: 11/28/2022]
Abstract
Arterial hypertension is a risk factor for various cardiovascular and renal diseases, representing a major public health challenge. Although a wide range of treatment options are available for blood pressure control, many hypertensive individuals remain with uncontrolled hypertension. Thus, the search for new substances with antihypertensive potential becomes necessary. Coumarins, a group of polyphenolic compounds derived from plants, have attracted intense interest due to their diverse pharmacological properties, like potent antihypertensive activities. Braylin (6-methoxyseselin) is a coumarin identified in the Zanthoxylum tingoassuiba species, described as a phosphodiesterase-4 (PDE4) inhibitor. Although different coumarin compounds have been described as potent antihypertensive agents, the activity of braylin on the cardiovascular system has yet to be investigated. To investigate the vasorelaxation properties of braylin and its possible mechanisms of action, we performed in vitro studies using superior mesenteric arteries and the iliac arteries isolated from rats. In this study, we demonstrated, for the first time, that braylin induces potent vasorelaxation, involving distinct mechanisms from two different arteries, isolated from rats. A possible inhibition of phosphodiesterase, altering the cyclic adenosine monophosphate (cAMP)/cAMP-dependent protein kinase (PKA) pathway, may be correlated with the biological action of braylin in the mesenteric vessel, while in the iliac artery, the biological action of braylin may be correlated with increase of cyclic guanosine monophosphate (cGMP), followed by BKCa, Kir, and Kv channel activation. Together, these results provide evidence that braylin can represent a potential therapeutic use in preventing and treating cardiovascular diseases.
Collapse
Affiliation(s)
- W A Santos
- Laboratory of Cardiovascular Physiology and Pharmacology, Federal University of Bahia, Avenida Reitor Miguel Calmon, Vale do Canela, Salvador, Bahia, 40110-902, Brazil
| | - K M C Dourado
- Laboratory of Cardiovascular Physiology and Pharmacology, Federal University of Bahia, Avenida Reitor Miguel Calmon, Vale do Canela, Salvador, Bahia, 40110-902, Brazil
| | - F A Araújo
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation - FIOCRUZ, Rio de Janeiro, Bahia, Brazil
| | - R L C Jesus
- Laboratory of Cardiovascular Physiology and Pharmacology, Federal University of Bahia, Avenida Reitor Miguel Calmon, Vale do Canela, Salvador, Bahia, 40110-902, Brazil
| | - R A Moraes
- Laboratory of Cardiovascular Physiology and Pharmacology, Federal University of Bahia, Avenida Reitor Miguel Calmon, Vale do Canela, Salvador, Bahia, 40110-902, Brazil
| | - S C D S Oliveira
- Laboratory of Cardiovascular Physiology and Pharmacology, Federal University of Bahia, Avenida Reitor Miguel Calmon, Vale do Canela, Salvador, Bahia, 40110-902, Brazil
| | - Q L Alves
- Laboratory of Cardiovascular Physiology and Pharmacology, Federal University of Bahia, Avenida Reitor Miguel Calmon, Vale do Canela, Salvador, Bahia, 40110-902, Brazil
| | - L O Simões
- Laboratory of Cardiovascular Physiology and Pharmacology, Federal University of Bahia, Avenida Reitor Miguel Calmon, Vale do Canela, Salvador, Bahia, 40110-902, Brazil
| | - L L Casais-E-Silva
- Laboratory of Cardiovascular Physiology and Pharmacology, Federal University of Bahia, Avenida Reitor Miguel Calmon, Vale do Canela, Salvador, Bahia, 40110-902, Brazil
| | - R S Costa
- Faculty of Pharmacy, Federal University of Bahia, Salvador, Bahia, Brazil
| | - E S Velozo
- Faculty of Pharmacy, Federal University of Bahia, Salvador, Bahia, Brazil
| | - D F Silva
- Laboratory of Cardiovascular Physiology and Pharmacology, Federal University of Bahia, Avenida Reitor Miguel Calmon, Vale do Canela, Salvador, Bahia, 40110-902, Brazil.
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation - FIOCRUZ, Rio de Janeiro, Bahia, Brazil.
| |
Collapse
|
126
|
Periodontal therapy and treatment of hypertension-alternative to the pharmacological approach. A systematic review and meta-analysis. Pharmacol Res 2021; 166:105511. [PMID: 33617973 DOI: 10.1016/j.phrs.2021.105511] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/17/2021] [Accepted: 02/17/2021] [Indexed: 01/02/2023]
Abstract
AIM Quantitative comparison of the effects of intensive (IPT) or conventional (CPT) periodontal treatment on arterial blood pressure, endothelial function and inflammatory/metabolic biomarkers. MATERIALS AND METHODS A systematic search was conducted to identify randomized controlled trials (RCT) of IPT (supra and subgingival instrumentation). Eight RCTs were included in the meta-analysis. Difference in change of systolic blood pressure (SBP) and diastolic blood pressure (DBP) before and after IPT or CPT were the primary outcomes. The secondary outcomes included: endothelial function and selected inflammatory/anti-inflammatory (CRP, IL-6, IL-10, IFN-γ) and metabolic biomarkers (HDL, LDL, TGs). RESULTS The overall effect estimates (pooled Weighted Mean Difference (WMD)) of the primary outcome for SBP and DBP was -4.3 mmHg [95%CI: -9.10-0.48], p = 0.08 and -3.16 mmHg [95%CI: -6.51-0.19], p = 0.06 respectively. These studies were characterized by high heterogeneity. Therefore, random effects model for meta-analysis was performed. Sub-group analyses confirmed statistically significant reduction in SBP [WMD = -11.41 mmHg (95%CI: -13.66, -9.15) P < 0.00001] and DBP [WMD = -8.43 mmHg (95%CI: -10.96,-5.91)P < 0.00001] after IPT vs CPT among prehypertensive/hypertensive patients, while this was not observed in normotensive individuals. The meta-analyses showed significant reductions in CRP and improvement of endothelial function following IPT at all analysed timepoints. CONCLUSIONS IPT leads to improvement of the cardiovascular health in hypertensive and prehypertensive individuals.
Collapse
|
127
|
Mattson DL, Dasinger JH, Abais-Battad JM. Amplification of Salt-Sensitive Hypertension and Kidney Damage by Immune Mechanisms. Am J Hypertens 2021; 34:3-14. [PMID: 32725162 PMCID: PMC7891248 DOI: 10.1093/ajh/hpaa124] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/27/2020] [Accepted: 07/23/2020] [Indexed: 12/13/2022] Open
Abstract
Humans with salt-sensitive (SS) hypertension demonstrate increased morbidity, increased mortality, and renal end-organ damage when compared with normotensive subjects or those with salt-resistant hypertension. Increasing evidence indicates that immune mechanisms play an important role in the full development of SS hypertension and associated renal damage. Recent experimental advances and studies in animal models have permitted a greater understanding of the mechanisms of activation and action of immunity in this disease process. Evidence favors a role of both innate and adaptive immune mechanisms that are triggered by initial, immune-independent alterations in blood pressure, sympathetic activity, or tissue damage. Activation of immunity, which can be enhanced by a high-salt intake or by alterations in other components of the diet, leads to the release of cytokines, free radicals, or other factors that amplify renal damage and hypertension and mediate malignant disease.
Collapse
Affiliation(s)
- David L Mattson
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - John Henry Dasinger
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Justine M Abais-Battad
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| |
Collapse
|
128
|
Osborn JW, Tyshynsky R, Vulchanova L. Function of Renal Nerves in Kidney Physiology and Pathophysiology. Annu Rev Physiol 2021; 83:429-450. [PMID: 33566672 DOI: 10.1146/annurev-physiol-031620-091656] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Renal sympathetic (efferent) nerves play an important role in the regulation of renal function, including glomerular filtration, sodium reabsorption, and renin release. The kidney is also innervated by sensory (afferent) nerves that relay information to the brain to modulate sympathetic outflow. Hypertension and other cardiometabolic diseases are linked to overactivity of renal sympathetic and sensory nerves, but our mechanistic understanding of these relationships is limited. Clinical trials of catheter-based renal nerve ablation to treat hypertension have yielded promising results. Therefore, a greater understanding of how renal nerves control the kidney under physiological and pathophysiological conditions is needed. In this review, we provide an overview of the current knowledge of the anatomy of efferent and afferent renal nerves and their functions in normal and pathophysiological conditions. We also suggest further avenues of research for development of novel therapies targeting the renal nerves.
Collapse
Affiliation(s)
- John W Osborn
- Department of Surgery, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA;
| | - Roman Tyshynsky
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Lucy Vulchanova
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, USA
| |
Collapse
|
129
|
Teklu M, Zhou W, Kapoor P, Patel N, Dey AK, Sorokin AV, Manyak GA, Teague HL, Erb-Alvarez JA, Sajja A, Abdelrahman KM, Reddy AS, Uceda DE, Lateef SS, Shanbhag SM, Scott C, Prakash N, Svirydava M, Parel P, Rodante JA, Keel A, Siegel EL, Chen MY, Bluemke DA, Playford MP, Gelfand JM, Mehta NN. Metabolic syndrome and its factors are associated with noncalcified coronary burden in psoriasis: An observational cohort study. J Am Acad Dermatol 2021; 84:1329-1338. [PMID: 33383084 DOI: 10.1016/j.jaad.2020.12.044] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/10/2020] [Accepted: 12/16/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Psoriasis is associated with a heightened risk of cardiovascular disease and higher prevalence of metabolic syndrome. OBJECTIVE Investigate the effect of metabolic syndrome and its factors on early coronary artery disease assessed as noncalcified coronary burden by coronary computed tomography angiography in psoriasis. METHODS This cross-sectional study consisted of 260 participants with psoriasis and coronary computed tomography angiography characterization. Metabolic syndrome was defined according to the harmonized International Diabetes Federation criteria. RESULTS Of the 260 participants, 80 had metabolic syndrome (31%). The metabolic syndrome group had a higher burden of cardiometabolic disease, systemic inflammation, noncalcified coronary burden, and high-risk coronary plaque. After adjusting for Framingham risk score, lipid-lowering therapy, and biologic use, metabolic syndrome (β = .31; P < .001) and its individual factors of waist circumference (β = .33; P < .001), triglyceride levels (β = .17; P = .005), blood pressure (β = .18; P = .005), and fasting glucose (β = .17; P = .009) were significantly associated with noncalcified coronary burden. After adjusting for all other metabolic syndrome factors, blood pressure and waist circumference remained significantly associated with noncalcified coronary burden. LIMITATIONS Observational nature with limited ability to control for confounders. CONCLUSIONS In psoriasis, individuals with metabolic syndrome had more cardiovascular disease risk factors, systemic inflammation, and noncalcified coronary burden. Efforts to increase metabolic syndrome awareness in psoriasis should be undertaken to reduce the heightened cardiovascular disease risk.
Collapse
Affiliation(s)
- Meron Teklu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Wunan Zhou
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Promita Kapoor
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Nidhi Patel
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Amit K Dey
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Alexander V Sorokin
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Grigory A Manyak
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Heather L Teague
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Julie A Erb-Alvarez
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Aparna Sajja
- Department of Internal Medicine, Johns Hopkins University Medical Center, Baltimore, Maryland
| | - Khaled M Abdelrahman
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Aarthi S Reddy
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Domingo E Uceda
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Sundus S Lateef
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Sujata M Shanbhag
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Colin Scott
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Nina Prakash
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Maryia Svirydava
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Philip Parel
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Justin A Rodante
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Andrew Keel
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Evan L Siegel
- Department of Rheumatology, Arthritis and Rheumatism Associates, Rockville, Maryland
| | - Marcus Y Chen
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - David A Bluemke
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Martin P Playford
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Joel M Gelfand
- Department of Dermatology, Perelman School of Medicine, Philadelphia, Pennsylvania; Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Nehal N Mehta
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
130
|
Pagano PJ, Cifuentes-Pagano E. The Enigmatic Vascular NOX: From Artifact to Double Agent of Change: Arthur C. Corcoran Memorial Lecture - 2019. Hypertension 2021; 77:275-283. [PMID: 33390049 DOI: 10.1161/hypertensionaha.120.13897] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
NOXs (NADPH oxidases) comprise a family of proteins whose primary function is the production of reactive oxygen species, namely, superoxide anion and hydrogen peroxide. The prototype first being discovered and characterized in neutrophils, multiple NOXs are now known to be broadly expressed in cell and organ systems and whose phylogeny spans countless life forms beginning with prokaryotes. This long-enduring evolutionary conservation underscores the importance of fundamental NOX functions. This review chronicles a personal perspective of the field beginning with the discovery of NOXs in the vasculature and the advances achieved through the years as to our understanding of their mechanisms of action and role in oxidative stress and disease. Furthermore, applications of isoform-selective inhibitors to dissect the role of NOX isozymes in vascular biology, focusing on inflammation, pulmonary hypertension, and aging are described.
Collapse
Affiliation(s)
- Patrick J Pagano
- Department of Pharmacology and Chemical Biology, Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, PA
| | - Eugenia Cifuentes-Pagano
- Department of Pharmacology and Chemical Biology, Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, PA
| |
Collapse
|
131
|
Eskandari M, Asghari H, Saghebjoo M, Kazemi T. Short duration moderate resistance training reduces blood pressure and plasma TNF-α in hypertensive men: The importance role of upper and lower body training. Sci Sports 2021. [DOI: 10.1016/j.scispo.2019.12.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
132
|
Wei SG, Yu Y, Felder RB. TNF-α-induced sympathetic excitation requires EGFR and ERK1/2 signaling in cardiovascular regulatory regions of the forebrain. Am J Physiol Heart Circ Physiol 2021; 320:H772-H786. [PMID: 33337962 PMCID: PMC8082799 DOI: 10.1152/ajpheart.00606.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 12/09/2020] [Accepted: 12/09/2020] [Indexed: 12/13/2022]
Abstract
Peripherally or centrally administered TNF-α elicits a prolonged sympathetically mediated pressor response, but the underlying molecular mechanisms are unknown. Activation of extracellular signal-regulated kinases 1 and 2 (ERK1/2) in cardiovascular regions of the brain has recently been recognized as a key mediator of sympathetic excitation, and ERK1/2 signaling is induced by activation of epidermal growth factor receptor (EGFR) tyrosine kinase activity. The present study examined the role of EGFR and ERK1/2 signaling in the sympathetic response to TNF-α. In urethane-anesthetized rats, intracarotid artery injection of TNF-α increased phosphorylation of EGFR and ERK1/2 in the subfornical organ (SFO) and the hypothalamic paraventricular nucleus (PVN); upregulated the gene expression of excitatory mediators in SFO and PVN; and increased blood pressure (BP), heart rate (HR), and renal sympathetic nerve activity (RSNA). A continuous intracerebroventricular infusion of the selective EGFR tyrosine kinase inhibitor AG1478 or the ERK1/2 inhibitor PD98059 significantly attenuated these responses. Bilateral PVN microinjections of TNF-α also increased phosphorylated ERK1/2 and the gene expression of excitatory mediators in PVN, along with increases in BP, HR, and RSNA, and these responses were substantially reduced by prior bilateral PVN microinjections of AG1478. These results identify activation of EGFR in cardiovascular regulatory regions of the forebrain as an important molecular mediator of TNF-α-driven sympatho-excitatory responses and suggest that EGFR activation of the ERK1/2 signaling pathway plays an essential role. These mechanisms likely contribute to sympathetic excitation in pathophysiological states like heart failure and hypertension, in which circulating and brain TNF-α levels are increased.NEW & NOTEWORTHY Proinflammatory cytokines contribute to the augmented sympathetic nerve activity in hypertension and heart failure, but the central mechanisms involved are largely unknown. The present study reveals that TNF-α transactivates EGFR in the subfornical organ and the hypothalamic paraventricular nucleus to initiate ERK1/2 signaling, upregulate the gene expression of excitatory mediators, and increase sympathetic nerve activity. These findings identify EGFR as a gateway to sympathetic excitation and a potential target for intervention in cardiovascular disease states.
Collapse
Affiliation(s)
- Shun-Guang Wei
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Yang Yu
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Robert B Felder
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Veterans Affairs Medical Center, Iowa City, Iowa
| |
Collapse
|
133
|
Zuk AM, Liberda EN, Tsuji LJS. Examining chronic inflammatory markers on blood pressure measures in the presence of vitamin D insufficiency among indigenous cree adults: results from the cross-sectional Multi-Community Environment-and-Health Study in Eeyou Istchee, Quebec, Canada. BMJ Open 2021; 11:e043166. [PMID: 33504558 PMCID: PMC7843349 DOI: 10.1136/bmjopen-2020-043166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 12/14/2020] [Accepted: 12/18/2020] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE High blood pressure (BP) is a risk factor for cardiovascular disease. Examining the role of inflammatory mediators on BP is important since vitamin D (VD) is a modifiable risk factor, which possibly modulates inflammatory cytokines. This study simulated what are known as average 'controlled direct effects (CDE)' of inflammatory markers, C reactive protein (CRP), tumour necrosis factor-α (TNF-α), and interlukin-6 (IL-6) on continuous BP measures, while fixing VD, an intermediate variable to specific level. DESIGN Cross-sectional study. SETTING We analysed data from the Multi-Community Environment-and-Health Study, 2005-2009, conducted in Eeyou Istchee, Quebec, Canada. PARTICIPANTS This study recruited 1425 study Indigenous Cree participants from seven Cree communities. Only adults with serum VD levels, inflammatory markers and BP measures were included in this data analysis. PRIMARY AND SECONDARY OUTCOMES MEASURES Inflammatory markers examined the top 25th exposure percentiles. VD 'insufficiency' (ie, 25-hydroxyvitamin-D levels<50 nmol/L) defined by the Institute of Medicine. CDE for each inflammatory marker in the presence and absence of population VD insufficiency simulated the average direct effect change for systolic and diastolic BP (SBP and DBP) measures. All models were adjusted for exposure-and-mediator outcome relationship. RESULTS Among 161 participants, 97 (60 %) were female. The prevalence of VD insufficiency was 32%. CDE estimates show in the presence and absence of population vitamin D insufficiency, inflammatory markers have a slightly different association on BP. TNF-α significantly and inversely associated with SBP in the presence of vitamin D insufficiency, fully adjusted model β = -13.61 (95% CI -24.42 to -2.80); however, TNF-α was not associated with SBP in the absence of vitamin D insufficiency. CRP, IL-6 were also not significantly associated with BP measures, although the magnitude of association was greater for those with elevated inflammation and VD insufficiency. CONCLUSION This novel analysis shows in the presence of VD insufficiency, inflammation (particularly TNF-α) may affect SBP. Additional research is needed to elucidate these findings, and the temporal relationship between these variables.
Collapse
Affiliation(s)
- Aleksandra M Zuk
- Department of Physical and Environmental Sciences, University of Toronto, Toronto, Ontario, Canada
- School of Nursing, Queen's University, Kingston, Ontario, Canada
| | - Eric N Liberda
- School of Occupational and Public Health, Ryerson University, Toronto, Ontario, Canada
| | - Leonard J S Tsuji
- Department of Physical and Environmental Sciences, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
134
|
Kuk M, Ward NC, Dwivedi G. Extrinsic and Intrinsic Responses in the Development and Progression of Atherosclerosis. Heart Lung Circ 2021; 30:807-816. [PMID: 33468387 DOI: 10.1016/j.hlc.2020.12.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 11/10/2020] [Accepted: 12/02/2020] [Indexed: 11/25/2022]
Abstract
Atherosclerosis is a multifactorial disease that is thought to be primarily inflammatory in origin. Given the contribution of inflammation to the development and progression of atherosclerosis, other conditions that are characterised by a dysregulated inflammatory response have also been proposed to play a role. The purpose of this review is to organise and present the various inflammatory processes that can affect atherosclerosis into two broad categories: extrinsic or host-independent and intrinsic or host-dependent. Within these two categories, we will discuss various processes that may contribute to the development and progression of atherosclerosis and the clinical studies describing these associations. Although the clinical trials investigating anti-inflammatory therapies have to date provided mixed results, further studies, particularly in conjunction with lipid-lowering and blood pressure lowering therapies should be considered.
Collapse
Affiliation(s)
- Mariya Kuk
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, Canada; McGill University Health Centre, McGill University, Montreal, Canada
| | - Natalie C Ward
- School of Public Health, Curtin University, Perth, WA, Australia; Medical School, University of Western Australia, Perth, WA, Australia
| | - Girish Dwivedi
- Medical School, University of Western Australia, Perth, WA, Australia; Harry Perkins Institute for Medical Research, Fiona Stanley Hospital, Perth, WA, Australia.
| |
Collapse
|
135
|
Pellegrini C, Martelli A, Antonioli L, Fornai M, Blandizzi C, Calderone V. NLRP3 inflammasome in cardiovascular diseases: Pathophysiological and pharmacological implications. Med Res Rev 2021; 41:1890-1926. [PMID: 33460162 DOI: 10.1002/med.21781] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/30/2020] [Accepted: 12/15/2020] [Indexed: 12/11/2022]
Abstract
Growing evidence points out the importance of nucleotide-binding oligomerization domain leucine-rich repeat and pyrin domain-containing protein 3 (NLRP3) inflammasome in the pathogenesis of cardiovascular diseases (CVDs), including hypertension, myocardial infarct (MI), ischemia, cardiomyopathies (CMs), heart failure (HF), and atherosclerosis. In this regard, intensive research efforts both in humans and in animal models of CVDs are being focused on the characterization of the pathophysiological role of NLRP3 inflammasome signaling in CVDs. In addition, clinical and preclinical evidence is coming to light that the pharmacological blockade of NLRP3 pathways with drugs, including novel chemical entities as well as drugs currently employed in the clinical practice, biologics and phytochemicals, could represent a suitable therapeutic approach for prevention and management of CVDs. On these bases, the present review article provides a comprehensive overview of clinical and preclinical studies about the role of NLRP3 inflammasome in the pathophysiology of CVDs, including hypertension, MI, ischemic injury, CMs, HF and atherosclerosis. In addition, particular attention has been focused on current evidence on the effects of drugs, biologics, and phytochemicals, targeting different steps of inflammasome signaling, in CVDs.
Collapse
Affiliation(s)
| | - Alma Martelli
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | - Luca Antonioli
- Department of Clinical and Experimental Medicine, Unit of Pharmacology and Pharmacovigilance, University of Pisa, Pisa, Italy
| | - Matteo Fornai
- Department of Clinical and Experimental Medicine, Unit of Pharmacology and Pharmacovigilance, University of Pisa, Pisa, Italy
| | - Corrado Blandizzi
- Department of Clinical and Experimental Medicine, Unit of Pharmacology and Pharmacovigilance, University of Pisa, Pisa, Italy
| | | |
Collapse
|
136
|
Landman TR, Thijssen DH, Tuladhar AM, de Leeuw FE. Relation between physical activity and cerebral small vessel disease: A nine-year prospective cohort study. Int J Stroke 2021; 16:962-971. [PMID: 33413019 DOI: 10.1177/1747493020984090] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND AND AIMS Given the unexplored potential of physical activity to reduce the progression of cerebral small vessel disease (cSVD, the purpose of this study was to prospectively (across nine-year follow-up) examine the relation between (baseline) physical activity and the (clinical and imaging) consequences of the whole spectrum of cerebral small vessel disease. METHODS Five hundred and three patients with cerebral small vessel disease from the RUNDMC study were followed for nine years. Physical activity was assessed using a questionnaire in 2006, 2011, and 2015. Clinical events (i.e. all-cause mortality, cerebrovascular events (by stroke subtype)) were collected with a structured questionnaire. Patients underwent magnetic resonance imaging scanning for the assessment of magnetic resonance imaging markers of cerebral small vessel disease (i.e. white matter hyperintensities, lacunes, and microbleeds) and microstructural integrity of the white matter at three timepoints. RESULTS The mean age at baseline was 66 (SD 9.0) years; 44% were women. A higher baseline physical activity level was independently associated with a lower all-cause mortality (HR: 0.69, 95%CI: 0.49-0.98, p = 0.03) and incidence of cerebrovascular disease (HR: 0.58, 95%CI: 0.36-0.96, p = 0.03). However, we found no relation between physical activity and incident lacunar stroke or progression of magnetic resonance imaging markers of cerebral small vessel disease. CONCLUSIONS Whilst regular physical activity was not related to the progression of magnetic resonance imaging markers of cerebral small vessel disease across a nine-year follow-up, results from our study prove that high levels of physical activity in patients with cerebral small vessel disease are associated with a lower all-cause mortality and lower incidence of cerebrovascular events.
Collapse
Affiliation(s)
- Thijs Rj Landman
- Department of Physiology, Radboud Institute for Health Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Dick Hj Thijssen
- Department of Physiology, Radboud Institute for Health Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Anil M Tuladhar
- Department of Neurology, Centre for Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Frank-Erik de Leeuw
- Department of Neurology, Centre for Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
137
|
Hypertension and Periodontitis: An Upcoming Joint Report by the Italian Society of Hypertension (SIIA) and the Italian Society of Periodontology and Implantology (SIdP). High Blood Press Cardiovasc Prev 2021; 28:1-3. [PMID: 33400213 DOI: 10.1007/s40292-020-00430-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 12/23/2020] [Indexed: 01/03/2023] Open
|
138
|
Cañas CA, Cañas F, Bautista-Vargas M, Bonilla-Abadía F. Role of Tissue Factor in the Pathogenesis of COVID-19 and the Possible Ways to Inhibit It. Clin Appl Thromb Hemost 2021; 27:10760296211003983. [PMID: 33784877 PMCID: PMC8020089 DOI: 10.1177/10760296211003983] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 02/06/2023] Open
Abstract
COVID-19 (Coronavirus Disease 2019) is a highly contagious infection and associated with high mortality rates, primarily in elderly; patients with heart failure; high blood pressure; diabetes mellitus; and those who are smokers. These conditions are associated to increase in the level of the pulmonary epithelium expression of angiotensin-converting enzyme 2 (ACE-2), which is a recognized receptor of the S protein of the causative agent SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2). Severe cases are manifested by parenchymal lung involvement with a significant inflammatory response and the development of microvascular thrombosis. Several factors have been involved in developing this prothrombotic state, including the inflammatory reaction itself with the participation of proinflammatory cytokines, endothelial dysfunction/endotheliitis, the presence of antiphospholipid antibodies, and possibly the tissue factor (TF) overexpression. ARS-Cov-19 ACE-2 down-regulation has been associated with an increase in angiotensin 2 (AT2). The action of proinflammatory cytokines, the increase in AT2 and the presence of antiphospholipid antibodies are known factors for TF activation and overexpression. It is very likely that the overexpression of TF in COVID-19 may be related to the pathogenesis of the disease, hence the importance of knowing the aspects related to this protein and the therapeutic strategies that can be derived. Different therapeutic strategies are being built to curb the expression of TF as a therapeutic target for various prothrombotic events; therefore, analyzing this treatment strategy for COVID-19-associated coagulopathy is rational. Medications such as celecoxib, cyclosporine or colchicine can impact on COVID-19, in addition to its anti-inflammatory effect, through inhibition of TF.
Collapse
Affiliation(s)
- Carlos A. Cañas
- Unit of Rheumatology, Fundación Valle del Lili, Universidad Icesi, Cali, Colombia
| | - Felipe Cañas
- Unit of Cardiology, Clínica Medellín, Medellín, Colombia
| | | | - Fabio Bonilla-Abadía
- Unit of Rheumatology, Fundación Valle del Lili, Universidad Icesi, Cali, Colombia
| |
Collapse
|
139
|
Latorre M, Spronck B, Humphrey JD. Complementary roles of mechanotransduction and inflammation in vascular homeostasis. Proc Math Phys Eng Sci 2021; 477:20200622. [PMID: 33642928 PMCID: PMC7897647 DOI: 10.1098/rspa.2020.0622] [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: 07/31/2020] [Accepted: 12/09/2020] [Indexed: 12/13/2022] Open
Abstract
Arteries are exposed to relentless pulsatile haemodynamic loads, but via mechanical homeostasis they tend to maintain near optimal structure, properties and function over long periods in maturity in health. Numerous insults can compromise such homeostatic tendencies, however, resulting in maladaptations or disease. Chronic inflammation can be counted among the detrimental insults experienced by arteries, yet inflammation can also play important homeostatic roles. In this paper, we present a new theoretical model of complementary mechanobiological and immunobiological control of vascular geometry and composition, and thus properties and function. We motivate and illustrate the model using data for aortic remodelling in a common mouse model of induced hypertension. Predictions match the available data well, noting a need for increased data for further parameter refinement. The overall approach and conclusions are general, however, and help to unify two previously disparate literatures, thus leading to deeper insight into the separate and overlapping roles of mechanobiology and immunobiology in vascular health and disease.
Collapse
Affiliation(s)
- Marcos Latorre
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Bart Spronck
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA,Department of Biomedical Engineering, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - Jay D. Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA,Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA,e-mail:
| |
Collapse
|
140
|
Chen Y, Qin Z, Wang Y, Li X, Zheng Y, Liu Y. Role of Inflammation in Vascular Disease-Related Perivascular Adipose Tissue Dysfunction. Front Endocrinol (Lausanne) 2021; 12:710842. [PMID: 34456867 PMCID: PMC8385491 DOI: 10.3389/fendo.2021.710842] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 07/21/2021] [Indexed: 12/12/2022] Open
Abstract
Perivascular adipose tissue (PVAT) is the connective tissue around most blood vessels throughout the body. It provides mechanical support and maintains vascular homeostasis in a paracrine/endocrine manner. Under physiological conditions, PVAT has anti-inflammatory effects, improves free fatty acid metabolism, and regulates vasodilation. In pathological conditions, PVAT is dysfunctional, secretes many anti-vasodilator factors, and participates in vascular inflammation through various cells and mediators; thus, it causes dysfunction involving vascular smooth muscle cells and endothelial cells. Inflammation is an important pathophysiological event in many vascular diseases, such as vascular aging, atherosclerosis, and hypertension. Therefore, the pro-inflammatory crosstalk between PVAT and blood vessels may comprise a novel therapeutic target for the prevention and treatment of vascular diseases. In this review, we summarize findings concerning PVAT function and inflammation in different pathophysiological backgrounds, focusing on the secretory functions of PVAT and the crosstalk between PVAT and vascular inflammation in terms of vascular aging, atherosclerosis, hypertension, diabetes mellitus, and other diseases. We also discuss anti-inflammatory treatment for potential vascular diseases involving PVAT.
Collapse
Affiliation(s)
- Yaozhi Chen
- Center for Cardiovascular Medicine, First Hospital of Jilin University, Changchun, China
| | - Zeyu Qin
- Department of Respiratory Medicine, First Hospital of Jilin University, Changchun, China
| | - Yaqiong Wang
- Department of Endocrinology and Metabolism, First Hospital of Jilin University, Changchun, China
| | - Xin Li
- Center for Cardiovascular Medicine, First Hospital of Jilin University, Changchun, China
| | - Yang Zheng
- Center for Cardiovascular Medicine, First Hospital of Jilin University, Changchun, China
- *Correspondence: Yunxia Liu, ; Yang Zheng,
| | - Yunxia Liu
- Center for Cardiovascular Medicine, First Hospital of Jilin University, Changchun, China
- *Correspondence: Yunxia Liu, ; Yang Zheng,
| |
Collapse
|
141
|
Pan XX, Wu F, Chen XH, Chen DR, Chen HJ, Kong LR, Ruan CC, Gao PJ. T-cell senescence accelerates angiotensin II-induced target organ damage. Cardiovasc Res 2021; 117:271-283. [PMID: 32049355 DOI: 10.1093/cvr/cvaa032] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 01/04/2020] [Accepted: 02/05/2020] [Indexed: 01/08/2023] Open
Abstract
AIMS Aging is a risk factor for cardiovascular diseases and adaptive immunity has been implicated in angiotensin (Ang) II-induced target organ dysfunction. Herein, we sought to determine the role of T-cell senescence in Ang II-induced target organ impairment and to explore the underlying mechanisms. METHODS AND RESULTS Flow cytometric analysis revealed that T cell derived from aged mice exhibited immunosenescence. Adoptive transfer of aged T cells to immunodeficient RAG1 KO mice accelerates Ang II-induced cardiovascular and renal fibrosis compared with young T-cell transfer. Aged T cells also promote inflammatory factor expression and superoxide production in these target organs. In vivo and in vitro studies revealed that Ang II promotes interferon-gamma (IFN-γ) production in the aged T cells comparing to young T cells. Importantly, transfer of senescent T cell that IFN-γ KO mitigates the impairment. Aged T-cell-conditioned medium stimulates inflammatory factor expression and oxidative stress in Ang II-treated renal epithelial cells compared with young T cells, and these effects of aged T-cell-conditioned medium are blunted after IFN-γ-neutralizing antibody pre-treatment. CONCLUSION These results provide a significant insight into the contribution of senescent T cells to Ang II-induced cardiovascular dysfunction and provide an attractive possibility that targeting T cell specifically might be a potential strategy to treat elderly hypertensive patients with end-organ dysfunction.
Collapse
Affiliation(s)
- Xiao-Xi Pan
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Fang Wu
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiao-Hui Chen
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Dong-Rui Chen
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Hong-Jin Chen
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Ling-Ran Kong
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Cheng-Chao Ruan
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Ping-Jin Gao
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| |
Collapse
|
142
|
von Degenfeld G, Truebel H. Cardiovascular translational biomarkers: translational aspects of hypertension, atherosclerosis, and heart failure in drug development in the digital era. PRINCIPLES OF TRANSLATIONAL SCIENCE IN MEDICINE 2021:177-193. [DOI: 10.1016/b978-0-12-820493-1.00017-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
143
|
Kang KW, Ok M, Lee SK. Leptin as a Key between Obesity and Cardiovascular Disease. J Obes Metab Syndr 2020; 29:248-259. [PMID: 33342767 PMCID: PMC7789022 DOI: 10.7570/jomes20120] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/03/2020] [Accepted: 12/13/2020] [Indexed: 12/14/2022] Open
Abstract
Obesity increases the risk of cardiovascular disease through various influencing factors. Leptin, which is predominantly secreted by adipose tissue, regulates satiety homeostasis and energy balance, and influences cardiovascular functions directly and indirectly. Leptin appears to play a role in heart protection in leptin-deficient and leptin-receptor-deficient rodent model experiments. Hyperleptinemia or leptin resistance in human obesity influences the vascular endothelium, cardiovascular structure and functions, inflammation, and sympathetic activity, which may lead to cardiovascular disease. Leptin is involved in many processes, including signal transduction, vascular endothelial function, and cardiac structural remodeling. However, the dual (positive and negative) regulator effect of leptin and its receptor on cardiovascular disease has not been completely understood. The protective role of leptin signaling in cardiovascular disease could be a promising target for cardiovascular disease prevention in obese patients.
Collapse
Affiliation(s)
- Ki-Woon Kang
- Division of Cardiology, Department of Internal Medicine, Eulji University School of Medicine, Daejeon, Korea
| | - Minho Ok
- Department of Cardiovascular Pharmacology, Mokpo National University, Mokpo, Korea
| | - Seong-Kyu Lee
- Division of Endocrinology, Department of Internal Medicine, Daejeon, Korea.,Department of Biochemistry-Molecular Biology, Eulji University School of Medicine, Daejeon, Korea
| |
Collapse
|
144
|
Bisaria S, Terrigno V, Hunter K, Roy S. Association of Elevated Levels of Inflammatory Marker High-Sensitivity C-Reactive Protein and Hypertension. J Prim Care Community Health 2020; 11:2150132720984426. [PMID: 33356789 PMCID: PMC7768830 DOI: 10.1177/2150132720984426] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Introduction The correlation between inflammation and vascular disease is widely accepted. High levels of C-reactive protein (CRP) have been shown to play a role in the process of endothelial dysfunction. Hypertension is described as an inflammatory vascular disease, and is 1 of the most commonly encountered diseases in the outpatient setting. We studied the association between the elevated high sensitivity-CRP (hs-CRP) level and hypertension, as well as other comorbid conditions. Methods Electronic medical records of 169 adult patients in our internal medicine office were reviewed for hs-CRP levels, and divided into 2 groups: elevated hs-CRP (≥2 mg/L; n = 110) and normal hs-CRP (<2 mg/L; n = 59). Independent T-Test was used to compare the means of continuous variables between the groups if they were normally distributed. Mann Whitney U-Test was used to compare the continuous variables that were non-parametric. Logistic regression was used to compare the dependent and independent variables. Results Among subjects with elevated hs-CRP, 58.2% had hypertension while 47.5% of subjects with normal hs-CRP levels had hypertension (P = .182). There were higher frequencies of association of coronary artery disease (CAD), cerebrovascular disease and hypothyroidism in elevated hs-CRP group but the differences were not statistically significant. Mean white blood cell count was statistically higher in elevated hs-CRP group (P < .05), while alcohol use was significantly higher (P < .05) and statin use was higher in the normal hs-CRP group. There was an inverse relationship between HDL-C and hs-CRP. Conclusions There was no statistically significant correlation between hs-CRP level and hypertension. Hs-CRP has statistically significant associations between alcohol use, dementia, white blood cell count, and HDL levels. Promising but not statistically significant correlations were observed between hs-CRP and statin therapy, hypothyroidism, coronary artery disease, and cerebrovascular disease.
Collapse
Affiliation(s)
- Sharmila Bisaria
- Cooper Medical School of Rowan University, Cooper University Health Care, Department of Medicine, Camden, New Jersey, USA
| | - Vittorio Terrigno
- Cooper Medical School of Rowan University, Cooper University Health Care, Department of Medicine, Camden, New Jersey, USA
| | - Krystal Hunter
- Cooper Research Institute, Cooper Medical School of Rowan University, Camden, New Jersey, USA
| | - Satyajeet Roy
- Cooper Medical School of Rowan University, Cooper University Health Care, Department of Medicine, Camden, New Jersey, USA
| |
Collapse
|
145
|
Jackson IL, Okonta JM, Ukwe CV. HIV- and hypertension-related knowledge and medication adherence in HIV seropositive persons with hypertension. J Public Health (Oxf) 2020; 44:e79-e87. [DOI: 10.1093/pubmed/fdaa221] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 11/07/2020] [Indexed: 11/13/2022] Open
Abstract
AbstractBackgroundThere is increasing prevalence of diseases associated with ageing such as hypertension, among people living with HIV (PLWHV). This study sought to assess: (1) knowledge related to HIV infection and hypertension, (2) adherence to prescribed medications and (3) clinical outcomes among HIV-positive persons with hypertension at a Nigerian hospital.MethodsThe Patient’s HIV Knowledge Questionnaire (PHKQ), Hypertension Knowledge-Level Scale (HK-LS) and Adherence to Refills and Medication Scale (ARMS) were administered to 220 eligible patients through the HIV clinic. Demographic and clinical data were also obtained.ResultsParticipants were predominantly females (57.3%), with a median (IQR) age of 46 (38–58) years; majority were married (67.8%) and employed (60.8%). Participants reported a higher hypertension-related knowledge compared with HIV-related knowledge (63.6% versus 33.3%, Z = −10.263, P < 0.001), but better adherence to antiretroviral medications compared to antihypertensives (100.0% versus 89.3%, Z = −9.118, P < 0.001). Of the 98 participants with documented viral load, 55 (56.1%) had undetectable (<40 copies/ml) values; however, only four (2.0%) of the entire sample had controlled (<140/90 mmHg) blood pressure.ConclusionsDespite having a higher hypertension-related knowledge, adherence to antihypertensive medications and blood pressure control were poor. There is a need for increased attention to HIV education and comorbidities in PLWHV.
Collapse
Affiliation(s)
- I L Jackson
- Department of Clinical Pharmacy and Biopharmacy, University of Uyo, Uyo 520103, Nigeria
| | - J M Okonta
- Department of Clinical Pharmacy and Pharmacy Management, University of Nigeria, Nsukka 410001, Nigeria
| | - C V Ukwe
- Department of Clinical Pharmacy and Pharmacy Management, University of Nigeria, Nsukka 410001, Nigeria
| |
Collapse
|
146
|
Eslamifar Z, Behzadifard M, Soleimani M, Behzadifard S. Coagulation abnormalities in SARS-CoV-2 infection: overexpression tissue factor. Thromb J 2020; 18:38. [PMID: 33323111 PMCID: PMC7737414 DOI: 10.1186/s12959-020-00250-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 11/12/2020] [Indexed: 12/15/2022] Open
Abstract
Among the pathways and mediators that may be dysregulated in COVID-19 infection, there are proinflammatory cytokines, lymphocyte apoptosis, and the coagulation cascade. Venous and arterial thromboembolisms also are frequent in COVID-19 patients with the increased risk of some life-threatening complications such as pulmonary embolism, myocardial infarction, and ischemic stroke. In this regard, overproduction of proinflammatory cytokines such as IL-6, IL-1β, and TNF-α induce cytokine storms, increase the risk of clot formation, platelet activation, and multiorgan failure that may eventually lead to death among these patients. Surface S protein of SARS-CoV-2 binds to its target transmembrane receptor, named as angiotensin converting enzyme 2 (ACE2(, on various cells such as lymphocyte, alveolar cells, monocytes/macrophages, and platelets. Notably, the activation of the coagulation cascade occurs through tissue factor (TF)/FVIIa-initiated hemostasis. Accordingly, TF plays the major role in the activation of coagulation system during viral infection. In viral infections, the related coagulopathy multiple factors such as inflammatory cytokines and viral specific TLRs are involved, which consequently induce TF expression aberrantly. SARS-COV-2 may directly infect monocytes/ macrophages. In addition, TF expression/release from these cells may play a critical role in the development of COVID-19 coagulopathy. In this regard, the use of TF- VIIa complex inhibitor may reduce the cytokine storm and mortality among COVID-19 patients.
Collapse
Affiliation(s)
| | | | - Masoud Soleimani
- Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Saba Behzadifard
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
147
|
Crouch SH, Botha-Le Roux S, Delles C, Graham LA, Schutte AE. Inflammation and hypertension development: A longitudinal analysis of the African-PREDICT study. INTERNATIONAL JOURNAL CARDIOLOGY HYPERTENSION 2020; 7:100067. [PMID: 33392493 PMCID: PMC7768897 DOI: 10.1016/j.ijchy.2020.100067] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/31/2020] [Accepted: 11/06/2020] [Indexed: 12/20/2022]
Abstract
Background The role of inflammation in the development of hypertension remains incompletely understood. While single inflammatory mediators have been shown to associate with changes in blood pressure (ΔBP), the role of clusters of inflammatory mediators has been less comprehensively explored. We therefore determined whether individual or clusters of inflammatory mediators from a large biomarker panel were associated with ΔBP over 4.5 years, in young healthy adults. Methods We included 358 adults (white, n = 156; black, n = 202) with detailed information on ambulatory blood pressure (BP) at baseline and follow-up. Baseline blood samples were analysed for 22 inflammatory mediators using multiplexing technology. Principal component analysis was used to study associations between clusters of inflammatory mediators and ΔBP. Results In the total cohort in multivariable-adjusted regression analyses, percentage change in 24hr systolic BP associated positively with Factors 1 (Interferon-gamma, interleukin (IL)-4, IL-7, IL-10, IL-12, IL-17A, IL-21, IL-23, macrophage inflammatory protein (MIP)-1α, MIP-1β, TNF-α, granulocyte-macrophage colony-stimulating factor (GM-CSF)) and 2 (IL-5, IL-6, IL-8, IL-13). Change in daytime systolic BP associated positively with Factors 1, 2 and 3 (C-Reactive protein, IL-1β, IL-2, MIP-3α). Subgroup analysis found these findings were limited to white study participants. Numerous associations were present between individual inflammatory mediators (Interferon-gamma, GM-CSF, IL-4, IL-6, IL-7, IL-8, IL-10, IL-12, IL-13, IL-17A, IL-21, IL-23, MIP-1α and MIP-1β) and ΔBP in the white but not black subgroups. Conclusion We found independent relationships between numerous inflammatory mediators (individual and clusters) and ΔBP over 4.5 years. The relationship between inflammatory markers and ΔBP was only found in white participants. ClinicalTrials.gov (Identifier: NCT03292094)..
Collapse
Affiliation(s)
- Simone H. Crouch
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa
| | - Shani Botha-Le Roux
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa
- MRC Research Unit: Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| | - Christian Delles
- The British Heart Foundation Centre of Excellence, Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Lesley A. Graham
- The British Heart Foundation Centre of Excellence, Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Aletta E. Schutte
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa
- MRC Research Unit: Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
- School of Population Health, University of New South Wales; The George Institute for Global Health, Sydney, Australia
- Corresponding author. School of Population Health, UNSW Medicine, University of New South Wales, NSW 2052, Australia.
| |
Collapse
|
148
|
Ghantous CM, Kamareddine L, Farhat R, Zouein FA, Mondello S, Kobeissy F, Zeidan A. Advances in Cardiovascular Biomarker Discovery. Biomedicines 2020; 8:biomedicines8120552. [PMID: 33265898 PMCID: PMC7759775 DOI: 10.3390/biomedicines8120552] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases are the leading causes of mortality worldwide. Among them, hypertension and its pathological complications pose a major risk for the development of other cardiovascular diseases, including heart failure and stroke. Identifying novel and early stage biomarkers of hypertension and other cardiovascular diseases is of paramount importance in predicting and preventing the major morbidity and mortality associated with these diseases. Biomarkers of such diseases or predisposition to their development are identified by changes in a specific indicator’s expression between healthy individuals and patients. These include changes in protein and microRNA (miRNA) levels. Protein profiling using mass spectrometry and miRNA screening utilizing microarray and sequencing have facilitated the discovery of proteins and miRNA as biomarker candidates. In this review, we summarized some of the different, promising early stage protein and miRNA biomarker candidates as well as the currently used biomarkers for hypertension and other cardiovascular diseases. Although a number of promising markers have been identified, it is unlikely that a single biomarker will unambiguously aid in the classification of these diseases. A multi-marker panel-strategy appears useful and promising for classifying and refining risk stratification among patients with cardiovascular disease.
Collapse
Affiliation(s)
- Crystal M. Ghantous
- Department of Nursing and Health Sciences, Faculty of Nursing and Health Sciences, Notre Dame University-Louaize, Keserwan 72, Lebanon;
| | - Layla Kamareddine
- Biomedical Sciences Department, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar;
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha 2713, Qatar
| | - Rima Farhat
- Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon;
| | - Fouad A. Zouein
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon;
| | - Stefania Mondello
- Oasi Research Institute-IRCCS, 94018 Troina, Italy;
- Department of Biomedical and Dental Sciences and Morpho-functional Imaging, University of Messina, 98125 Messina, Italy
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon;
| | - Asad Zeidan
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha 2713, Qatar
- Department of Basic Medical Science, Faculty of Medicine, QU Health, Qatar University, Doha 2713, Qatar
- Correspondence: ; Tel.: +97-431-309-19
| |
Collapse
|
149
|
Humphrey JD. Mechanisms of Vascular Remodeling in Hypertension. Am J Hypertens 2020; 34:432-441. [PMID: 33245319 PMCID: PMC8140657 DOI: 10.1093/ajh/hpaa195] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/09/2020] [Accepted: 11/19/2020] [Indexed: 12/19/2022] Open
Abstract
Hypertension is both a cause and a consequence of central artery stiffening, which in turn is an initiator and indicator of myriad disease conditions and thus all-cause mortality. Such stiffening results from a remodeling of the arterial wall that is driven by mechanical stimuli and mediated by inflammatory signals, which together lead to differential gene expression and concomitant changes in extracellular matrix composition and organization. This review focuses on biomechanical mechanisms by which central arteries remodel in hypertension within the context of homeostasis-what promotes it, what prevents it. It is suggested that the vasoactive capacity of the wall and inflammatory burden strongly influence the ability of homeostatic mechanisms to adapt the arterial wall to high blood pressure or not. Maladaptation, often reflected by inflammation-driven adventitial fibrosis, not just excessive intimal-medial thickening, significantly diminishes central artery function and disturbs hemodynamics, ultimately compromising end organ perfusion and thus driving the associated morbidity and mortality. It is thus suggested that there is a need for increased attention to controlling both smooth muscle phenotype and inflammation in hypertensive remodeling of central arteries, with future studies of the often adaptive response of medium-sized muscular arteries promising to provide additional guidance.
Collapse
Affiliation(s)
- Jay D Humphrey
- Department of Biomedical Engineering, Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut, USA,Correspondence: Jay D. Humphrey ()
| |
Collapse
|
150
|
Müller-Wieland D, Marx N, Dreher M, Fritzen K, Schnell O. COVID-19 and Cardiovascular Comorbidities. Exp Clin Endocrinol Diabetes 2020; 130:178-189. [PMID: 33157558 DOI: 10.1055/a-1269-1405] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The emergence of a new coronavirus - severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) - has resulted in a global pandemic. The associated coronavirus disease 2019 (COVID-19) has resulted in a high number of death worldwide. Observational studies and case reports have provided insights that older age and the presence of chronic diseases is frequently associated with a higher COVID-19 severity. These individuals also seem to have a higher risk of mortality due to COVID-19. In this review we provide insights into the impact chronic diseases associated with the cardiovascular system, such as obesity, diabetes mellitus, hypertension and cardiovascular disease might have on SARS-CoV-2 infection and COVID-19. Additionally we review recommendations and guidance's of international scientific associations and discuss which key learnings might be of importance for the future.
Collapse
Affiliation(s)
- Dirk Müller-Wieland
- Clinic for Cardiology, Angiology and Internal Care Medicine (Medical Clinic I) Universitatsklinikum Aachen, Aachen
| | - Nikolaus Marx
- Clinic for Cardiology, Angiology and Internal Care Medicine (Medical Clinic I) Universitatsklinikum Aachen, Aachen
| | - Michael Dreher
- Clinic for Pneumology and Internal Intensive Care Medicine (Medical Clinic V, Uniklinik RWTH Aachen, Aachen
| | | | - Oliver Schnell
- Sciarc GmbH, Baierbrunn.,Forschergruppe Diabetes e.V., Munich Neuherberg
| |
Collapse
|