Abstract
Cardiovascular diseases (CVDs) have been the top-ranked cause of human death in the world for years, according to the World Health Organization. Accumulating evidence from epidemiological data supports the view that the risk of CVDs is higher in northern China than in southern area. There is no doubt that living environment has become a crucial factor contributing to the occurrence and progression of CVDs in northern region. However, there have not been any clinical guidelines for the prevention strategy of environment-related CVDs, especially for cold exposure. Thus, there is an urgent need for better understanding of the clinical characteristics and underlying mechanisms of cold-induced CVDs in order to formulate and implement proper and effective measures for minimizing the risk of CVDs for people residing in low-temperature area. Cold exposure, air pollution, lack of sunlight and irrational diet are believed to be crucial factors responsible for environment-related CVDs, and preventive measures might be carried out accordingly to decrease the high risk of CVDs in northern China.
A large number of epidemiological studies confirm that individuals living in frigid zone have a higher risk of multiple cardiovascular diseases (CVDs), such as ischemic heart disease (IHD), hypertension, cardiac arrhythmia and heart failure, which lead to increased death[1]. The differences of living environment have been considered one of the most important factors responsible for the increase in CVDs[2–3]. However, there have not been any authoritative guidelines for the prevention and treatment of CVDs for these patients in cold environment. Thus, geographically targeted interventions are urgently needed to mitigate the CVD-related health and social burden in frigid zone.
It has been recognized that there is a wide geographical distribution of CVDs risk factors and their clusters throughout China. In a nationwide population-based cohort study in China, 983 476 individuals from 31 provinces were included, and after standardizing age and gender, the overall high CVD risk was 10.3%, with a range of 3.1%–24.9% among all the provinces. Moreover, the results showed that northeast China (12.6%) and northern China (11.4%) had relatively higher prevalence of high risk of CVDs, whereas southern China (8.0%) was lower[4]. Various factors contribute to the high prevalence of CVDs in northern China, including cold exposure, air pollution, inadequate sunlight, and unhealthy diet.
1 Cold exposure and CVDs
Compared with other regions, northern China experiences a longer and colder winter, with far lower average annual temperature. People in northern China expose to cold for a long time. A study comparing the cold tolerance of people from north with those from south reached a surprising conclusion that the northern residents are more sensitive to cold[5]. Immense amount of evidence from epidemiological data supports that cold exposure is closely correlated to various CVDs, including hypertension, myocardial infarction (MI), atrial fibrillation (AF), and heart failure (HF), resulting in adverse prognosis and high mortality. A cross-sectional study including 506 673 adults (aged 30–79 years) recruited from 10 diverse urban or rural regions in China showed that blood pressure was strongly inversely associated with outdoor temperature, and the systolic blood pressure peaked in cold seasons[6]. Moreover, ambient temperature is closely related to hypertensive disorders in pregnancy[7], and maternal exposure to cold spells during pregnancy is associated with hypertension in offspring later in life[8]. When temperature is lower than 15.6°C, the risk of myocardial infarction increases by 1.6% for every 1°C decrease[9]. Moreover, it has been demonstrated that HF admissions and mortality are closely associated with cold temperatures, especially in the older population[10]. A prospective study including 200 patients with dual chamber implantable cardioverter-defibrillators found that lower temperatures within the initial 48 hours were positively associated with the new onset episodes of AF and that the risk of AF increased by 3% for every 1°C decrease in ambient temperature[11]. More importantly, temperature is closely related to the prognosis of patients with AF, and the risk of cardiovascular events and all-cause mortality are higher during colder months and seasons[12].
The activation of sympathetic nervous system (SNS) and reninangiotensin-aldosterone system (RAAS), inflammatory response, gut microbiota dysbiosis and endothelial dysfunction are thought to be responsible for the high risk of CVDs in residents under cold exposure.
1.1 Cold activates SNS and RAAS
Continuous hypothermia stimulation can increase SNS activity. SNS is an important regulatory mechanism for maintaining body homeostasis. Excessive activation of SNS can increase arterial blood pressure, secondary inflammation, and plaque formation, which results in atherosclerosis. Healthy people with extreme cold exposure showed an increase in the muscle sympathetic activity, which is associated with increased blood pressure[13], and changes in the ratio of low-frequency to high-frequency of heart rate variability (HRV) can be observed while sleeping in low temperature surroundings[14]. Moreover, cold exposure increases the angiotensin II (Ang II) level in patients with major cardiovascular events[15–16]. These results indicate that cold exposure causes the activation of SNS and RAAS[15,17–18]. Chen et al.[19] found that adult offspring of parent rats that had exposed to cold had increased activities of the peripheral and central sympathetic nervous systems. In addition, administration of clonidine, a centrally acting α2 adrenergic receptor agonist, lowered blood pressure to a greater degree in the prenatal cold-exposed offspring than control ones, indicating that the increase in peripheral sympathetic nerve activity can be ascribed to the activation of central nervous system.
1.2 Cold stress exacerbates systemic inflammation
One study aimed to investigate the effects of cold stress on the quail cecum revealed that cold stress caused inflammatory injury in cecal tissues[20]. Bao et al.[21] found that low temperature (18°C) upregulated the expressions of inflammatory cytokines including interleukin (IL)-1β, IL-4, IL-6, IL-8, IL-10, IL-13 and granulocyte macrophage colony-stimulating factor (GM-CSF). Furthermore, tumor necrosis factor (TNF)-α in airway epithelial cells was isolated from asthma model mice, highlighting the important role of cold exposure in activation of inflammation. Zhang et al.[22] revealed that long-term (21 d) cold exposure enhanced inflammatory response in mice with elevated TNF-α and IL-6 concentrations in peripheral blood samples. In an experimental study, male Sprague-Dawley rats were exposed to cold environment (5°C) after knockdown of IL-6[23]. It was found that IL-6 small hairpin RNA (shRNA) prevented cold-induced inflammation by inhibiting leukocyte infiltration, intravascular superoxide synthesis and cardiac collagen deposition, as well as decreasing systolic blood pressure[23]. Yu et al.[24] discovered that in addition to stimulating cytokines, cold exposure (4±1)°C significantly raised blood pressure, NLRP3 inflammasome level and fibrosis in the aorta of Sprague-Dawley rats, which could be attenuated by tranilast, an inhibitor of NLRP3. In northern China, people who are exposed to low ambient temperature for a longer time have an enhanced defensive response to low temperature stimulation, thereby increasing circulating inflammation. Clearly, cold exposure contributes to CVDs partly via activating systemic inflammation.
1.3 Cold induces gut microbiota dysbiosis
The gut microbiota and its metabolites have been implicated in the pathogenesis of many diseases, such as CVDs and diabetes[25]. Cold exposure has been reported to reshape the composition-correlated metabolic pattern of the gut microbiota, ultimately resulting in cardiovascular alterations[26]. Compared with the mice raised at room temperature, the diversity of gut microbiota significantly decreased, and the abundance of Bacteroides increased in mice exposed to cold temperature. Zhang et al.[27] demonstrated that cold exposure decreased the expression of phosphorylated endothelial NO synthase (eNOS) protein and triggered the production of gut-derived inflammatory cytokines, TNF-α, and IL-6 in aorta, which caused vascular dysfunction. Moreover, cold exposure also increased gut permeability, inhibited tight junction protein expression in proximal colon, leading to gut barrier dysfunction, which allowed lipopolysaccharide, a harmful metabolite, to activate vascular inflammatory reaction.
1.4 Cold exposure leads to endothelial dysfunction
Cold exposure can cause endothelial dysfunction through endoplasmic reticulum stress, inhibition of adiponectin and up-regulation of uncoupling protein (UCP), thus promoting the growth of atherosclerotic plaque and increasing plaque instability[28,29,30]. Persistent cold stress induces activation of SNS and RAAS, producing continuous stress of endoplasmic reticulum in endothelial cells[28–29]. Noticeably, in cold areas, people are more likely to have a high level of plasma homocysteine due to smoking, high-fat diet and folic acid deficiency, which promote endoplasmic reticulum stress and endothelial cell apoptosis[31]. Cold exposure decreases the expression of adiponectin in vascular system, which in turn inhibits eNOS and weakens its protective effect on endothelial cells. Moreover, cold exposure enhances the activity of endothelin-1 (ET-1), a key factor regulating blood pressure and endothelial function, upregulates ETA-R and downregulates ETB-R, which in turn lead to vasoconstriction and sodium retention, eventually causing hypertension[18,32].
As for other possible mechanisms, studies in blood flow rheology indicated an association between “cavitation”, a term in the field of ultrasonography and the progression of intracoronary atherosclerotic plaque and thrombosis[33]. However, it remains to be elucidated whether cold exposure could contribute to the pathogenesis of MI via hemodynamic changes as predicted by computer simulation[34]. Cold exposure might also cause dysfunction of energy metabolism by dampening mitochondrial function, giving rise to vascular calcification (VC), a possible end-stage pathological process of CVDs, and VSMC phenotype switch modulated by hypoxia-induced autophagy[35–36].
2 Air pollution and CVDs
Air pollution is a crucial environmental risk factor contributing to global cardiovascular (CV) mortality and disability, especially the fine particulate matter ≤2.5 μm (PM 2.5). It has been established that PM 2.5 and inhalable particulate matter ≤10 μm (PM 10) are significantly higher in northern China than in other regions. The ESCAPE study confirmed that nonfatal acute coronary events increased by 13% with a 5 mg/m3 elevation following long-term exposure to PM 2.5[37]. Another study reported that short-term PM 2.5 exposure increased the relative risk of acute CV events by 1% to 3% and the relative risk for acute MI by 2.5% per 10 mg/m3[38]. More importantly, short-term exposure to PM 2.5 accounts for up to 5% of MI incidence worldwide[39]. Recently, one study conducted in China confirmed that the risks for CV morbidity and mortality are markedly increased with increasing exposure to polluted air, with the relative risk for CV mortality being increased by 9% per 10 mg/m3 elevation[40]. Furthermore, a meta-analysis showed that a short-term increase in gaseous components and PM (both PM 10 and PM 2.5) increased the risk for heart failure hospitalization or death[41]. Substantial evidence supports a link between air pollution and the development of cardiometabolic risk factors, such as hypertension, insulin resistance and cardiac arrhythmias[42,43,44]. However, the mechanism underlying PM-induced CVD remains largely unknown. PM may cause vascular dysfunction such as increased blood pressure and exacerbation of atherosclerosis, increased susceptibility of the heart to ischemic damage, and increased risk of thrombosis through direct and indirect effects[42, 45,46,47].
PM may increase the risk of cardiovascular events by the following indirect actions. The pulmonary and systematic oxidative stress as an early response to PM inhalation initiates many secondary processes[48–49]. First, the inflammatory mediators induced by PM entering the alveoli penetrate into the circulation[50]. Second, the activation of alveolar sensory receptors affects cardiac autonomic nerve function and humoral endocrine function through afferent nerves[51]. Finally, other unidentified blood-borne mediators may exert cardiovascular effects[52]. Oxidative stress and inflammation are more critical pathogenic mechanisms than the direct action of PM[45,53–54]. Long-term exposure to environment rich in PM activates the NLRP3 (Nacht, LRR and PYD domain signaling protein 3) inflammasome pathway and systemic inflammation in mice[55]. Reactive oxygen species (ROS) can directly or indirectly activate membrane receptors, such as Toll-like receptors2/4 (TLR2/TLR4) and the nucleotide-binding domain leucine-rich repeats of Nod-like receptors, etc., causing and exacerbating downstream inflammatory cascades[56,57,58]. A study about HECT E3 ubiquitin ligases provides a novel insight that living environment may be responsible for the occurrence of oxidative stress by affecting the expression of HECT E3 ubiquitin ligases, eventually causing alterations in vascular function and fundamental pathways such as the NF-κB and NLRP3 signaling pathway[59].
3 Sunlight and CVDs
Epidemiological data from clinical research showed that people in northern region were more likely to suffer from vitamin D deficiency compared with those in southern region, because the ultraviolet light decreased with increasing latitude[60]. Vitamin D deficiency has been identified as an important risk factor of CVDs. Mounting studies have shown that vitamin D deficiency significantly increases CVDs events. Studies uncovered that the majority of patients with acute MI present vitamin D deficiency or insufficiency and low serum vitamin D concentration is associated with the high risk of major adverse CVDs events (MACE) and increasing incidence of HF[61,62,63,64]. As the latitude increases, the gradual decline of UVB in the environment causes progressive increase in blood pressure proportional to latitude, which can be alleviated by exposing to sunlight and taking high-dose vitamin D2 supplementation[65,66,67].
3.1 The activation of RAAS
Vitamin D inhibits the synthesis of renin and the activation of RAAS by acting on vitamin D receptors (VDR). It has been reported that the levels of renin, angiotensin II and aldosterone were significantly higher in mice with vitamin D deficiency than that in wild type mice, with obvious myocardial hypertrophy and collagen aggregation, which could be reversed by vitamin D supplementation[68]. Given that decreased sunlight in northern China hinders the generation of vitamin D, RAAS is likely to be hyperactivated, thereby inducing CVDs. Intriguingly, aldosteronism owing to RAAS activation during HF and vitamin D deficiency due to sunscreen effect of melanin could contribute synergically to hyperthyroidism in African-Americans[69]. Except for their separate roles in developing hyperthyroidism, there might be crosstalk between RAAS and vitamin D deficiency, indicating that the activation of RAAS due partly to vitamin D deficiency triggers the elevation of circulating parathyroid hormone, ultimately causing oxidative stress and proinflammatory condition which in turn exacerbate HF[69].
3.2 Inflammatory response
Vitamin D deficiency upregulates karyopherin α4 (KPNA4) followed by subsequent activation of NF-κB, a transcription factor promoting the release of inflammatory cytokines (IL-6, IL-8, TNF-α, etc.) and motivating ox-LDL phagocytization of macrophages to form early atherosclerotic lipid streak[70,71,72]. Such inflammatory response can be reduced by adequate intracellular 1, 25-(OH)2-D3 and inhibition of NF-κB activation can alleviate cardiac dysfunction and ventricular remodeling in mice, further suggesting that vitamin D protects cardiovascular system via repressing the expression of the proinflammatory molecule NF-κB[70,73].
3.3 Cardiac hypertrophy
Vitamin D plays a critical role in maintaining the systolic and diastolic functions of cardiomyocytes by regulating calcium uptake. Vitamin D deficiency can trigger cardiac dysfunction and myocardial energy metabolic disorders, leading to myocardial hypertrophy. Moreover, cardiomyocyte-specific deletion of the VDR gene results in cardiac hypertrophy by activating the pro-hypertrophic calcineurin/NFAT/MCIP1 pathway[74]. Recently, vitamin D deficiency was confirmed to cause cardiac dysfunction by inducing myocardial insulin resistance[75]. In addition, Kong et al.[76] revealed that the beneficial effect of vitamin D was mediated by the cyclin-dependent kinase inhibitor p21.
3.4 Endothelial dysfunction
Multiple enzymes are involved in vitamin D metabolism and VDRs are expressed in endothelial cells in the blood vessel wall. The concentration of 25(OH)D in the serum is negatively correlated with endothelial dysfunction[77]. Lacking of endothelial VDR, the vasodilator effect in endothelial cells is significantly weakened under the stimulation of acetylcholine perfusion[78]. The above studies demonstrated the crucial role of vitamin D and its receptor VDRs in modulating NO synthesis and endothelial function[79]. It is demonstrated that vitamin D stimulates the synthesis of NO in endothelial cells but inhibits the production of vasoconstrictors such as cyclooxygenase-1[80–81]. UVA can induce the conversion of cutaneous unstable NO derivatives to NO, which diffuses into deep tissues to increase the level of local S-nitrosoglutathione that then redistributes in the blood circulation and eventually decreases blood pressure[82]. The following studies have confirmed the relationships among UVA, NO and blood pressure: short-term whole-body UVA irradiation can cause rapid and long-lasting blood pressure decrease in humans[83], NO scavengers block the inhibitory effects of UV irradiation on weight and metabolic syndrome[84], and UVA irradiation combined with oral nitrite supplementation improves the training performance of athletes[85]. Moreover, vitamin D inhibits endothelial cell apoptosis by inhibiting oxidative stress, the release of cytochrome C from mitochondria, the activity of cysteine proteases, and the expression of apoptosis/autophagy genes, thereby improving endothelial cell function[86].
4 Irrational diet and CVDs in northern China
A large amount of data showed that unhealthy diet is an important risk factor for CVD. Residents in northern China usually consume high-salt, high-fat, high alcohol, and less vegetables, which contributes to the north-south difference in CVD mortality rate[87]. In 2013, 12 million Chinese died of high-salt diets, among which 31.5% was caused by cardiovascular and cerebrovascular diseases[88]. More importantly, the burden of diseases due to a high-salt diet in northern China is significantly higher than that in other provinces in China and limiting daily salt intake can reduce the occurrence of CVD[89–90]. As we know, high-fat diet puts human at a high risk of obesity, an independent risk factor for CVDs. Epidemiological data showed that the prevalence of overweight and obesity, particularly central obesity, was higher among residents in northern China than their counterparts in southern China[91–92]. The CHARLS study revealed that higher prevalence of metabolic syndrome was observed in middle-aged and elderly women residing in northern China[93]. One study including 820 older adults in northern China showed that excessive sodium intake and alcohol drinking were both independently related to higher risk of hypertension[94]. Another study analyzed 16 100 CVD deaths among adults aged 25 to 69 in Shandong province and found that high sodium intake accounted for 19.9% of total CVD deaths. A considerable drop in CVD death can well be achieved with reduced sodium intake[95].
It is currently believed that a high-salt diet can affect the cardiovascular system through multiple mechanisms[96,97,98,99]. First, high-salt diet can cause sodium and water retention and increase circulating blood volume. Second, high-salt diet increases plasma sodium chloride concentration followed by the activation of sodium/osmotic pressure receptors to excite the sympathetic nerves. Third, high-salt diet damages the vascular endothelium and mitigate the release and utilization of NO, resulting in peripheral vasodilation dysfunction. In addition, high-salt diet can also increase arterial stiffness, with the pro-fibrotic effect of transforming growth factor-β (TGF-β) being one of the key factors. On the other hand, high-fat diet increases the prevalence of obesity and metabolic syndrome, contributing to CVDs.
5 Preventive measures of cold-related CVDs
At present, there is a lack of authoritative guideline for formulating practical prevention and control measures for CVDs in northern China. Such measures could greatly reduce the morbidity and mortality of CVDs, thereby improving citizens physical condition in northern China[100].
First, the direct exposure to cold environment can be avoided by keeping warm. A simple and effective method is to add clothes such as hats, gloves, thermal insulation jackets, etc.[101]. Long-term measures such as wall and pipe insulation of houses and improvement of the heating facilities should be implemented[102–103]. During the follow-up of 1 840 families with thermal insulation reconstruction, it was found that increasing indoor temperature could significantly reduce the incidence of hypertension[104]. Secondly, the government should urge communities to include the elderly, people with chronic diseases, alcohol and drug users, as well as specific occupations (custodians of freezer lockers, meat packers, ice cream manufacturers, etc.) into high-risk groups of cold-related CVDs, by providing health education for them and implementing programs against cold environments[18,105,106,107,108]. Moreover, increasing outdoor activities moderately, supplementing vitamin D and reducing salt intake can effectively prevent the occurrence of CVDs[102,109]. Interestingly, researchers have proposed that TRPV1 might be used as a potential therapeutic target for cold stress-related comorbidities[110–111]. Lu et al. demonstrated that a TRPV1 antagonist ameliorated cold stress-induced myocardial hypertrophy under pressure overload, while Zhang et al. suggested that cold exposure-induced cardiac contractile and geometric dysfunction could be reversed by a TRPV1 agonist[110–111]. Therefore, further studies are needed to elucidate the specific role of TRPV1 in the regulation of cold exposure-induced CVDs.
Considering the large reaction surface area and the recognized toxicity of PM, as well as mixed pollutants composed of nitrogen dioxide and PM, reducing automobile exhaust emissions is an effective measure to reduce the incidence of CVDs. In addition, the use of masks and indoor air purifiers can protect the cardiovascular system to a certain extent[112].
Taking balanced diet is the probably most cost-effective and simple way to avoid the detrimental effects of a high-salt diet on the cardiovascular system. Moderate servings of egg (up to <1 egg/d), dairy products and whole grain may help reduce the risk of CVDs by attenuating oxidative stress, enhancing endothelial function, and alleviating systemic inflammation[113,114,115,116]. Moreover, daily consumption of fruits containing dietary fiber, potassium, vitamins and antioxidants is also a plausible way of decreasing the incident of CVDs[117]. In recent years, several dietary patterns, such as Mediterranean diet and Jiangnan diet, have proven benefits to the primary prevention of CVDs and diabetes and the management of metabolic syndrome as well[118–119]. In addition, ketogenic diet and intermittent fasting are beneficial to obese and diabetic populations, while it remains largely unknown whether they can effectively decrease cardiovascular risk or improve outcomes[120].
6 Conclusions
The high prevalence of CVDs in northern China has provoked the exploration of the underlying mechanisms of environment-related CVDs. Cold exposure, air pollution, inadequate sunlight and irrational diet might be the key factors contributing to the pathogenesis and progression of CVDs in frigid zones. Various measures can be taken to reduce CVDs, such as individual and domestic protection against cold weather and air pollution, vitamin D supplementation, a balanced diet and so on. With the precise mechanisms to be elucidated in the future, more targeted measures would be implemented to alleviate the burden of environment-related CVDs in northern China.
Conflicts of interests
Yue Li is an Editorial Board Member of the journal. The article was subject to the journal's standard procedures, with peer review handled independently of this member and his research groups.
References
[1] Duan B. Concise Review: Harnessing iPSC-derived Cells for Ischemic Heart Disease Treatment. J Transl Int Med, 2020; 8(1): 20–25.10.2478/jtim-2020-0004Search in Google Scholar
[2] Sun D, Wang J, Shao W, et al. Pathogenesis and Damage Targets of Hypertensive Kidney Injury. J Transl Int Med, 2020; 8(4): 205–209.10.2478/jtim-2020-0033Search in Google Scholar
[3] Redant S, Honoré P M, De Bels D. Fifty Shades of Central Venous Pressure in the Cardiorenal Syndrome. J Transl Int Med, 2020; 8(1): 1–2.10.2478/jtim-2020-0001Search in Google Scholar
[4] Li X, Wu C, Lu J, et al. Cardiovascular risk factors in China: a nationwide population-based cohort study. Lancet Public Health, 2020; 5(12): e672–e681.10.1016/S2468-2667(20)30191-2Search in Google Scholar
[5] Yufan L, Liu Y, Wu X Z, et al. Physiological adaptability and subjective feelings of male college students in South and North China in an acceptable cold environment. J Civil, Archi Environ Eng. Journal of Chongqing Jianzhu University, 2018; 40(04): 55–62. (Chinese)Search in Google Scholar
[6] Lewington S, Li L, Sherliker P, et al. Seasonal variation in blood pressure and its relationship with outdoor temperature in 10 diverse regions of China: the China Kadoorie Biobank. J Hypertens, 2012; 30(7): 1383–1391.10.1097/HJH.0b013e32835465b5Search in Google Scholar PubMed PubMed Central
[7] Xiong T, Chen P, Mu Y, et al. Association between ambient temperature and hypertensive disorders in pregnancy in China. Nat Commun, 2020; 11(1): 2925.10.1038/s41467-020-16775-8Search in Google Scholar PubMed PubMed Central
[8] Li N, Cai L, Heizhati M, et al. Maternal exposure to cold spells during pregnancy is associated with higher blood pressure and hypertension in offspring later in life. J Clin Hypertens (Greenwich), 2020; 22(10): 1884–1891.10.1111/jch.14015Search in Google Scholar PubMed PubMed Central
[9] Cheng J, Bambrick H, Tong S, et al. Winter temperature and myocardial infarction in Brisbane, Australia: Spatial and temporal analyses. Sci Total Environ, 2020; 715: 136860.10.1016/j.scitotenv.2020.136860Search in Google Scholar PubMed
[10] Goggins W B, Chan E Y. A study of the short-term associations between hospital admissions and mortality from heart failure and meteorological variables in Hong Kong: Weather and heart failure in Hong Kong. Int J Cardiol, 2017; 228: 537–542.10.1016/j.ijcard.2016.11.106Search in Google Scholar PubMed
[11] Nguyen J L, Link M S, Luttmann-Gibson H, et al. Drier air, lower temperatures, and triggering of paroxysmal atrial fibrillation. Epidemiology, 2015; 26(3): 374–380.10.1097/EDE.0000000000000284Search in Google Scholar PubMed PubMed Central
[12] Rivera-Caravaca J M, Roldán V, Vicente V, et al. Particulate Matter and Temperature: Increased Risk of Adverse Clinical Outcomes in Patients With Atrial Fibrillation. Mayo Clin Proc, 2020; 95(11): 2360–2369.10.1016/j.mayocp.2020.05.046Search in Google Scholar PubMed
[13] Park J, Middlekauff H R, Campese V M. Abnormal sympathetic reactivity to the cold pressor test in overweight humans. Am J Hypertens, 2012; 25(12): 1236–1241.10.1038/ajh.2012.115Search in Google Scholar PubMed PubMed Central
[14] Okamoto-Mizuno K, Tsuzuki K, Mizuno K, et al. Effects of low ambient temperature on heart rate variability during sleep in humans. Eur J Appl Physiol, 2009; 105(2): 191–197.10.1007/s00421-008-0889-1Search in Google Scholar PubMed
[15] Sun Z. Cardiovascular responses to cold exposure. Front Biosci (Elite Ed), 2010; 2: 495–503.10.2741/e108Search in Google Scholar PubMed PubMed Central
[16] Zhang X, Zhang S, Wang C, et al. Effects of moderate strength cold air exposure on blood pressure and biochemical indicators among cardiovascular and cerebrovascular patients. Int J Environ Res Public Health, 2014; 11(3): 2472–2487.10.3390/ijerph110302472Search in Google Scholar PubMed PubMed Central
[17] Qingmei W, Hong G, Ye T. Cold exposure and hypertension. Chinese Journal of Hypertension, 2013; 21(01): 21–24. (Chinese)Search in Google Scholar
[18] Giorgini P, Di Giosia P, Petrarca M, et al. Climate changes and human health: a review of the effect of environmental stressors on cardiovascular diseases across epidemiology and biological mechanisms. Curr Pharm Des, 2017; 23(22): 3247–3261.10.2174/1381612823666170317143248Search in Google Scholar PubMed
[19] Chen K, Sun D, Qu S, et al. Prenatal cold exposure causes hypertension in offspring by hyperactivity of the sympathetic nervous system. Clin Sci (Lond), 2019; 133(9): 1097–1113.10.1042/CS20190254Search in Google Scholar PubMed PubMed Central
[20] Liu C, Chaudhry M T, Zhao D, et al. Heat shock protein 70 protects the quail cecum against oxidant stress, inflammatory injury, and microbiota imbalance induced by cold stress. Poult Sci, 2019; 98(11): 5432–5445.10.3382/ps/pez327Search in Google Scholar PubMed
[21] Liu H, Hua L, Liu Q, et al. Cold stimuli facilitate inflammatory responses through transient receptor potential melastatin 8 (TRPM8) in primary airway epithelial cells of asthmatic mice. Inflammation, 2018; 41(4): 1266–1275.10.1007/s10753-018-0774-ySearch in Google Scholar PubMed
[22] Nie Y, Yan Z, Yan W, et al. Cold exposure stimulates lipid metabolism, induces inflammatory response in the adipose tissue of mice and promotes the osteogenic differentiation of BMMSCs via the p38 MAPK pathway in vitro. Int J Clin Exp Pathol, 2015; 8(9): 10875–10886.Search in Google Scholar
[23] Crosswhite P, Sun Z. Ribonucleic acid interference knockdown of interleukin 6 attenuates cold-induced hypertension. Hypertension, 2010; 55(6): 1484–1491.10.1161/HYPERTENSIONAHA.109.146902Search in Google Scholar PubMed PubMed Central
[24] Yu H, Zhou Y, Duan Y, et al. Tranilast treats cold-related hypertension by reducing the expression of NLRP3 inflammasome. Frigid Zone Medicine, 2021; 1(2): 95–101.10.2478/fzm-2021-0012Search in Google Scholar
[25] Huang Q, Fang Q, Hu Z. A P4 medicine perspective of gut microbiota and prediabetes: systems analysis and personalized intervention. J Transl Int Med, 2020; 8(3): 119–130.10.2478/jtim-2020-0020Search in Google Scholar PubMed PubMed Central
[26] Worthmann A, John C, Rühlemann M C, et al. Cold-induced conversion of cholesterol to bile acids in mice shapes the gut microbiome and promotes adaptive thermogenesis. Nat Med, 2017; 23(7): 839–849.10.1038/nm.4357Search in Google Scholar PubMed
[27] Zhang S, Zhang Y, Ahsan M Z, et al. Atorvastatin attenuates cold-induced hypertension by preventing gut barrier injury. J Cardiovasc Pharmacol, 2019; 74(2): 143–151.10.1097/FJC.0000000000000690Search in Google Scholar PubMed
[28] Zhang K, Kaufman R J. From endoplasmic-reticulum stress to the inflammatory response. Nature, 2008; 454(7203): 455–462.10.1038/nature07203Search in Google Scholar PubMed PubMed Central
[29] Castro A F, Rebhun J F, Quilliam L A. Measuring ras-family gtp levels in vivo--running hot and cold. Methods, 2005; 37(2): 190–196.10.1016/j.ymeth.2005.05.015Search in Google Scholar PubMed
[30] Xue Y, Petrovic N, Cao R, et al. Hypoxia-independent angiogenesis in adipose tissues during cold acclimation. Cell Metab, 2009; 9(1): 99–109.10.1016/j.cmet.2008.11.009Search in Google Scholar PubMed
[31] Hotamisligil G S. Endoplasmic reticulum stress and atherosclerosis. Nat Med, 2010; 16(4): 396–399.10.1038/nm0410-396Search in Google Scholar PubMed PubMed Central
[32] Chen G F, Sun Z. Effects of chronic cold exposure on the endothelin system. J Appl Physiol (1985), 2006; 100(5): 1719–1726.10.1152/japplphysiol.01407.2005Search in Google Scholar PubMed
[33] Rigatelli G, Zuin M, Ngo T T, et al. Intracoronary Cavitation as a cause of plaque rupture and thrombosis propagation in patients with acute myocardial infarction: a computational study. J Transl Int Med, 2019; 7(2): 69–75.10.2478/jtim-2019-0014Search in Google Scholar PubMed PubMed Central
[34] Rigatelli G, Zuin M, Fong A, et al. Left main stenting induced flow disturbances on ascending aorta and aortic arch. J Transl Int Med, 2019; 7(1): 22–28.10.2478/jtim-2019-0005Search in Google Scholar
[35] Peng X, Wei C, Li H Z, et al. NPS2390, a selective calcium-sensing receptor antagonist controls the phenotypic modulation of hypoxic human pulmonary arterial smooth muscle cells by regulating autophagy. J Transl Int Med, 2019; 7(2): 59–68.10.2478/jtim-2019-0013Search in Google Scholar
[36] Wang P, Zhang N, Wu B, et al. The role of mitochondria in vascular calcification. J Transl Int Med, 2020; 8(2): 80–90.10.2478/jtim-2020-0013Search in Google Scholar
[37] Cesaroni G, Forastiere F, Stafoggia M, et al. Long term exposure to ambient air pollution and incidence of acute coronary events: prospective cohort study and meta-analysis in 11 European cohorts from the ESCAPE Project. BMJ, 2014; 348: f7412.10.1136/bmj.f7412Search in Google Scholar
[38] Mustafic H, Jabre P, Caussin C, et al. Main air pollutants and myocardial infarction: a systematic review and meta-analysis. JAMA, 2012; 307(7): 713–721.10.1001/jama.2012.126Search in Google Scholar
[39] Nawrot T S, Perez L, Künzli N, et al. Public health importance of triggers of myocardial infarction: a comparative risk assessment. Lancet, 2011; 377(9767): 732–740.10.1016/S0140-6736(10)62296-9Search in Google Scholar
[40] Yin P, Brauer M, Cohen A, et al. Long-term fine particulate matter exposure and nonaccidental and cause-specific mortality in a large national cohort of chinese men. Environ Health Perspect, 2017; 125(11): 117002.10.1289/EHP1673Search in Google Scholar
[41] Shah A S, Langrish J P, Nair H, et al. Global association of air pollution and heart failure: a systematic review and meta-analysis. Lancet, 2013; 382(9897): 1039–1048.10.1016/S0140-6736(13)60898-3Search in Google Scholar
[42] Yang B Y, Qian Z, Howard S W, et al. Global association between ambient air pollution and blood pressure: A systematic review and meta-analysis. Environ Pollut, 2018; 235: 576–588.10.1016/j.envpol.2018.01.001Search in Google Scholar PubMed
[43] Eze I C, Hemkens L G, Bucher H C, et al. Association between ambient air pollution and diabetes mellitus in Europe and North America: systematic review and meta-analysis. Environ Health Perspect, 2015; 123(5): 381–389.10.1289/ehp.1307823Search in Google Scholar PubMed PubMed Central
[44] Shao Q, Liu T, Korantzopoulos P, et al. Association between air pollution and development of atrial fibrillation: A meta-analysis of observational studies. Heart Lung, 2016; 45(6): 557–562.10.1016/j.hrtlng.2016.08.001Search in Google Scholar
[45] Miller M R, Newby D E. Air pollution and cardiovascular disease: car sick. Cardiovasc Res, 2020; 116(2): 279–294.10.1093/cvr/cvz228Search in Google Scholar
[46] Provost E B, Madhloum N, Int Panis L, et al. Carotid intima-media thickness, a marker of subclinical atherosclerosis, and particulate air pollution exposure: the meta-analytical evidence. PLoS One, 2015; 10(5): e0127014.10.1371/journal.pone.0127014Search in Google Scholar
[47] Hoffmann B, Moebus S, Möhlenkamp S, et al. Residential exposure to traffic is associated with coronary atherosclerosis. Circulation, 2007; 116(5): 489–496.10.1161/CIRCULATIONAHA.107.693622Search in Google Scholar
[48] Rao X, Zhong J, Brook R D, et al. Effect of particulate matter air pollution on cardiovascular oxidative stress pathways. Antioxid Redox Signal, 2018; 28(9): 797–818.10.1089/ars.2017.7394Search in Google Scholar
[49] Calderón-Garcidueñas L, Vojdani A, Blaurock-Busch E, et al. Air pollution and children: neural and tight junction antibodies and combustion metals, the role of barrier breakdown and brain immunity in neurodegeneration. J Alzheimers Dis, 2015; 43(3): 1039–1058.10.3233/JAD-141365Search in Google Scholar
[50] Seaton A, MacNee W, Donaldson K, et al. Particulate air pollution and acute health effects. Lancet, 1995; 345(8943): 176–178.10.1016/S0140-6736(95)90173-6Search in Google Scholar
[51] Kodavanti U P. Stretching the stress boundary: Linking air pollution health effects to a neurohormonal stress response. Biochim Biophys Acta, 2016; 1860(12): 2880–2890.10.1016/j.bbagen.2016.05.010Search in Google Scholar PubMed
[52] Channell M M, Paffett M L, Devlin R B, et al. Circulating factors induce coronary endothelial cell activation following exposure to inhaled diesel exhaust and nitrogen dioxide in humans: evidence from a novel translational in vitro model. Toxicol Sci, 2012; 127(1): 179–186.10.1093/toxsci/kfs084Search in Google Scholar PubMed PubMed Central
[53] Miller M R. The role of oxidative stress in the cardiovascular actions of particulate air pollution. Biochem Soc Trans, 2014; 42(4): 1006–1011.10.1042/BST20140090Search in Google Scholar PubMed
[54] Fiordelisi A, Piscitelli P, Trimarco B, et al. The mechanisms of air pollution and particulate matter in cardiovascular diseases. Heart Fail Rev, 2017; 22(3): 337–347.10.1007/s10741-017-9606-7Search in Google Scholar PubMed
[55] Bai N, Kido T, Suzuki H, et al. Changes in atherosclerotic plaques induced by inhalation of diesel exhaust. Atherosclerosis, 2011; 216(2): 299–306.10.1016/j.atherosclerosis.2011.02.019Search in Google Scholar PubMed PubMed Central
[56] Miyata R, van Eeden S F. The innate and adaptive immune response induced by alveolar macrophages exposed to ambient particulate matter. Toxicol Appl Pharmacol, 2011; 257(2): 209–226.10.1016/j.taap.2011.09.007Search in Google Scholar PubMed
[57] Kampfrath T, Maiseyeu A, Ying Z, et al. Chronic fine particulate matter exposure induces systemic vascular dysfunction via NADPH oxidase and TLR4 pathways. Circ Res, 2011; 108(6): 716–726.10.1161/CIRCRESAHA.110.237560Search in Google Scholar PubMed PubMed Central
[58] Shoenfelt J, Mitkus R J, Zeisler R, et al. Involvement of TLR2 and TLR4 in inflammatory immune responses induced by fine and coarse ambient air particulate matter. J Leukoc Biol, 2009; 86(2): 303–312.10.1189/jlb.1008587Search in Google Scholar PubMed PubMed Central
[59] Qian H, Zhang Y, Wu B, et al. Structure and function of HECT E3 ubiquitin ligases and their role in oxidative stress. J Transl Int Med, 2020; 8(2): 71–79.10.2478/jtim-2020-0012Search in Google Scholar PubMed PubMed Central
[60] Dou R, Ng K, Giovannucci E L, et al. Vitamin D and colorectal cancer: molecular, epidemiological and clinical evidence. Br J Nutr, 2016; 115(9): 1643–1660.10.1017/S0007114516000696Search in Google Scholar PubMed PubMed Central
[61] Lee J H, Gadi R, Spertus J A, et al. Prevalence of vitamin D deficiency in patients with acute myocardial infarction. Am J Cardiol, 2011; 107(11): 1636–1638.10.1016/j.amjcard.2011.01.048Search in Google Scholar PubMed PubMed Central
[62] Siasos G, Tousoulis D, Oikonomou E, et al. Vitamin D3, D2 and arterial wall properties in coronary artery disease. Curr Pharm Des, 2014; 20(37): 5914–5918.10.2174/1381612820666140619122937Search in Google Scholar PubMed
[63] Giovannucci E, Liu Y, Hollis B W, et al. 25-hydroxyvitamin D and risk of myocardial infarction in men: a prospective study. Arch Intern Med, 2008; 168(11): 1174–1180.10.1001/archinte.168.11.1174Search in Google Scholar PubMed PubMed Central
[64] Kim D H, Sabour S, Sagar U N, et al. Prevalence of hypovitaminosis D in cardiovascular diseases (from the National Health and Nutrition Examination Survey 2001 to 2004). Am J Cardiol, 2008; 102(11): 1540–1544.10.1016/j.amjcard.2008.06.067Search in Google Scholar PubMed
[65] Rostand S G. Ultraviolet light may contribute to geographic and racial blood pressure differences. Hypertension, 1997; 30(2 Pt 1): 150–156.10.1161/01.HYP.30.2.150Search in Google Scholar PubMed
[66] Kokot F, Schmidt-Gayk H, Wiecek A, et al. Influence of ultraviolet irradiation on plasma vitamin D and calcitonin levels in humans. Kidney Int Suppl, 1989; 27: S143–S146.Search in Google Scholar
[67] Sugden A, Smith J, Pennisi E. The future of forests. Science, 2008; 320(5882): 1435.10.1126/science.320.5882.1435Search in Google Scholar PubMed
[68] Li Y C, Qiao G, Uskokovic M, et al. Vitamin D: a negative endocrine regulator of the renin-angiotensin system and blood pressure. J Steroid Biochem Mol Biol, 2004; 89–90(1–5): 387–392.10.1016/j.jsbmb.2004.03.004Search in Google Scholar PubMed
[69] Bhattacharya S K, Ahokas R A, Carbone L D, et al. Macro- and micronutrients in African-Americans with heart failure. Heart Fail Rev, 2006; 11(1): 45–55.10.1007/s10741-006-9192-6Search in Google Scholar PubMed
[70] Kutuk O, Basaga H. Inflammation meets oxidation: NF-kappaB as a mediator of initial lesion development in atherosclerosis. Trends Mol Med, 2003; 9(12): 549–557.10.1016/j.molmed.2003.10.007Search in Google Scholar PubMed
[71] Chen S, Swier V J, Boosani C S, et al. Vitamin D deficiency accelerates coronary artery disease progression in Swine. Arterioscler Thromb Vasc Biol, 2016; 36(8): 1651–1659.10.1161/ATVBAHA.116.307586Search in Google Scholar PubMed PubMed Central
[72] Sun X, Icli B, Wara A K, et al. MicroRNA-181b regulates NF-κB-mediated vascular inflammation. J Clin Invest, 2012; 122(6): 1973–1990.10.1172/JCI61495Search in Google Scholar PubMed PubMed Central
[73] Onai Y, Suzuki J, Maejima Y, et al. Inhibition of NF-{kappa} B improves left ventricular remodeling and cardiac dysfunction after myocardial infarction. Am J Physiol Heart Circ Physiol, 2007; 292(1): H530–H538.10.1152/ajpheart.00549.2006Search in Google Scholar PubMed
[74] Chen S, Law C S, Grigsby C L, et al. Cardiomyocyte-specific deletion of the vitamin D receptor gene results in cardiac hypertrophy. Circulation, 2011; 124(17): 1838–1847.10.1161/CIRCULATIONAHA.111.032680Search in Google Scholar PubMed PubMed Central
[75] Nizami H L, Katare P, Prabhakar P, et al. Vitamin D deficiency in rats causes cardiac dysfunction by inducing myocardial insulin resistance. Mol Nutr Food Res, 2019; 63(17): e1900109.10.1002/mnfr.201900109Search in Google Scholar PubMed
[76] Liu N, Su H, Zhang Y, et al. The protective effect of 1,25(OH)(2)D(3) against cardiac hypertrophy is mediated by the cyclin-dependent kinase inhibitor p21. Eur J Pharmacol, 2020; 888: 173510.10.1016/j.ejphar.2020.173510Search in Google Scholar PubMed
[77] Zhang Q Y, Jiang C M, Sun C, et al. Hypovitaminosis D is associated with endothelial dysfunction in patients with non-dialysis chronic kidney disease. J Nephrol, 2015; 28(4): 471–476.10.1007/s40620-014-0167-8Search in Google Scholar PubMed
[78] Napoli C, de Nigris F, Williams-Ignarro S, et al. Nitric oxide and atherosclerosis: an update. Nitric Oxide, 2006; 15(4): 265–279.10.1016/j.niox.2006.03.011Search in Google Scholar PubMed
[79] Legarth C, Grimm D, Wehland M, et al. The impact of Vitamin D in the treatment of essential hypertension. Int J Mol Sci, 2018; 19(2):455.10.3390/ijms19020455Search in Google Scholar PubMed PubMed Central
[80] Andrukhova O, Slavic S, Zeitz U, et al. Vitamin D is a regulator of endothelial nitric oxide synthase and arterial stiffness in mice. Mol Endocrinol, 2014; 28(1): 53–64.10.1210/me.2013-1252Search in Google Scholar PubMed PubMed Central
[81] Wong M S, Man R Y, Vanhoutte P M. Calcium-independent phospholipase A(2) plays a key role in the endothelium-dependent contractions to acetylcholine in the aorta of the spontaneously hypertensive rat. Am J Physiol Heart Circ Physiol, 2010; 298(4): H1260–H1266.10.1152/ajpheart.01068.2009Search in Google Scholar PubMed
[82] Bhatnagar A. Environmental determinants of cardiovascular disease. Circ Res, 2017; 121(2): 162–180.10.1161/CIRCRESAHA.117.306458Search in Google Scholar PubMed PubMed Central
[83] Opländer C, Volkmar C M, Paunel-Görgülü A, et al. Whole body UVA irradiation lowers systemic blood pressure by release of nitric oxide from intracutaneous photolabile nitric oxide derivates. Circ Res, 2009; 105(10): 1031–1040.10.1161/CIRCRESAHA.109.207019Search in Google Scholar PubMed
[84] Geldenhuys S, Hart P H, Endersby R, et al. Ultraviolet radiation suppresses obesity and symptoms of metabolic syndrome independently of vitamin D in mice fed a high-fat diet. Diabetes, 2014; 63(11): 3759–3769.10.2337/db13-1675Search in Google Scholar PubMed
[85] Muggeridge D J, Sculthorpe N, Grace F M, et al. Acute whole body UVA irradiation combined with nitrate ingestion enhances time trial performance in trained cyclists. Nitric Oxide, 2015; 48: 3–9.10.1016/j.niox.2014.09.158Search in Google Scholar
[86] Uberti F, Lattuada D, Morsanuto V, et al. Vitamin D protects human endothelial cells from oxidative stress through the autophagic and survival pathways. J Clin Endocrinol Metab, 2014; 99(4): 1367–1374.10.1210/jc.2013-2103Search in Google Scholar
[87] Wang M, Huang Y, Song Y, et al. Study on environmental and lifestyle factors for the north-south differential of cardiovascular disease in China. Front Public Health, 2021; 9: 615152.10.3389/fpubh.2021.615152Search in Google Scholar
[88] Liu S W, Cai Y, Zeng X Y, et al. Deaths and life expectancy losses attributable to diet high in sodium in China. Zhonghua Liu Xing Bing Xue Za Zhi, 2017; 38(8): 1022–1027.Search in Google Scholar
[89] Liu M, Li Y C, Liu S W, et al. Burden of disease attributable to high-sodium diets in China, 2013. Zhonghua Yu Fang Yi Xue Za Zhi, 2016; 50(9): 759–763.Search in Google Scholar
[90] He F J, Li J, Macgregor G A. Effect of longer term modest salt reduction on blood pressure: Cochrane systematic review and meta-analysis of randomised trials. BMJ, 2013; 346: f1325.10.1136/bmj.f1325Search in Google Scholar
[91] Reynolds K, Gu D, Whelton P K, et al. Prevalence and risk factors of overweight and obesity in China. Obesity (Silver Spring), 2007; 15(1): 10–18.10.1038/oby.2007.527Search in Google Scholar
[92] Li Z, Luo B, Du L, et al. Familial clustering of overweight and obesity among schoolchildren in northern China. Int J Clin Exp Med, 2014; 7(12): 5778–5783.Search in Google Scholar
[93] Liu B, Chen G, Zhao R, et al. Temporal trends in the prevalence of metabolic syndrome among middle-aged and elderly adults from 2011 to 2015 in China: the China health and retirement longitudinal study (CHARLS). BMC Public Health, 2021; 21(1): 1045.10.1186/s12889-021-11042-xSearch in Google Scholar
[94] Nan X, Lu H, Wu J, et al. The interactive association between sodium intake, alcohol consumption and hypertension among elderly in northern China: a cross-sectional study. BMC Geriatr, 2021; 21(1): 135.10.1186/s12877-021-02090-4Search in Google Scholar
[95] Qiu M S, Wang X W, Yao Y, et al. Protocol of jidong women health cohort study: rationale, design, and baseline characteristics. Biomed Environ Sci, 2019; 32(2): 144–152.Search in Google Scholar
[96] Iimura O, Shimamoto K. Salt and hypertension: water-sodium handling in essential hypertension. Ann N Y Acad Sci, 1993; 676: 105–121.10.1111/j.1749-6632.1993.tb38729.xSearch in Google Scholar
[97] DuPont J J, Greaney J L, Wenner M M, et al. High dietary sodium intake impairs endothelium-dependent dilation in healthy salt-resistant humans. J Hypertens, 2013; 31(3): 530–536.10.1097/HJH.0b013e32835c6ca8Search in Google Scholar
[98] Safar M E, Thuilliez C, Richard V, et al. Pressure-independent contribution of sodium to large artery structure and function in hypertension. Cardiovasc Res, 2000; 46(2): 269–276.10.1016/S0008-6363(99)00426-5Search in Google Scholar
[99] Sanders P W. Vascular consequences of dietary salt intake. Am J Physiol Renal Physiol, 2009; 297(2): F237–F243.10.1152/ajprenal.00027.2009Search in Google Scholar
[100] Cheng X, Su H. Effects of climatic temperature stress on cardiovascular diseases. Eur J Intern Med, 2010; 21(3): 164–167.10.1016/j.ejim.2010.03.001Search in Google Scholar
[101] McCullough L, Arora S. Diagnosis and treatment of hypothermia. Am Fam Physician, 2004; 70(12): 2325–2332.Search in Google Scholar
[102] Zhang J W, Zhang Y, Li Y. Effects and mechanisms of living environment on cardiovascular disease in northern China. J Clin Pathol Sci, 2021; 41(01): 190–194.Search in Google Scholar
[103] Howden-Chapman P, Matheson A, Crane J, et al. Effect of insulating existing houses on health inequality: cluster randomised study in the community. BMJ, 2007; 334(7591): 460.10.1136/bmj.39070.573032.80Search in Google Scholar
[104] Umishio W, Ikaga T, Kario K, et al. Cross-sectional analysis of the relationship between home blood pressure and indoor temperature in winter: a nationwide smart wellness housing survey in Japan. Hypertension, 2019; 74(4): 756–766.10.1161/HYPERTENSIONAHA.119.12914Search in Google Scholar
[105] Sun Z, Cade R, Zhang Z, et al. Angiotensinogen gene knockout delays and attenuates cold-induced hypertension. Hypertension, 2003; 41(2): 322–327.10.1161/01.HYP.0000050964.96018.FASearch in Google Scholar
[106] Monacelli F, Aramini I, Odetti P. For debate: The August sun and the December snow. J Am Med Dir Assoc, 2010; 11(6): 449–452.10.1016/j.jamda.2010.01.013Search in Google Scholar
[107] Preventing injuries associated with extreme cold. Int J Trauma Nurs, 2001; 7(1): 26–30.10.1067/mtn.2001.112376Search in Google Scholar
[108] Ebi K L, Semenza J C. Community-based adaptation to the health impacts of climate change. Am J Prev Med, 2008; 35(5): 501–507.10.1016/j.amepre.2008.08.018Search in Google Scholar
[109] Cold exposure and winter mortality from ischaemic heart disease, cerebrovascular disease, respiratory disease, and all causes in warm and cold regions of Europe. Lancet, 1997; 349(9062): 1341–1346.10.1016/S0140-6736(96)12338-2Search in Google Scholar
[110] Zhang Y, Li L, Hua Y, et al. Cardiac-specific knockout of ET(A) receptor mitigates low ambient temperature-induced cardiac hypertrophy and contractile dysfunction. J Mol Cell Biol, 2012; 4(2): 97–107.10.1093/jmcb/mjs002Search in Google Scholar PubMed PubMed Central
[111] Lu S, Xu D. Cold stress accentuates pressure overload-induced cardiac hypertrophy and contractile dysfunction: role of TRPV1/AMPK-mediated autophagy. Biochem Biophys Res Commun, 2013; 442(1–2): 8–15.10.1016/j.bbrc.2013.10.128Search in Google Scholar PubMed
[112] Münzel T, Sørensen M, Gori T, et al. Environmental stressors and cardio-metabolic disease: part I-epidemiologic evidence supporting a role for noise and air pollution and effects of mitigation strategies. Eur Heart J, 2017; 38(8): 550–556.10.1093/eurheartj/ehw269Search in Google Scholar PubMed
[113] Qin C, Lv J, Yu C, et al. Dietary patterns and cardiometabolic diseases in 0.5 million Chinese adults: a 10-year cohort study. Nutr J, 2021; 20(1): 74.10.1186/s12937-021-00730-4Search in Google Scholar PubMed PubMed Central
[114] Kahleova H, Levin S, Barnard N D. Vegetarian dietary patterns and cardiovascular disease. Prog Cardiovasc Dis, 2018; 61(1): 54–61.10.1016/j.pcad.2018.05.002Search in Google Scholar PubMed
[115] Roager H M, Vogt J K, Kristensen M, et al. Whole grain-rich diet reduces body weight and systemic low-grade inflammation without inducing major changes of the gut microbiome: a randomised cross-over trial. Gut, 2019; 68(1): 83–93.10.1136/gutjnl-2017-314786Search in Google Scholar PubMed PubMed Central
[116] Qin C, Lv J, Guo Y, et al. Associations of egg consumption with cardiovascular disease in a cohort study of 0.5 million Chinese adults. Heart, 2018; 104(21): 1756–1763.10.1136/heartjnl-2017-312651Search in Google Scholar PubMed PubMed Central
[117] Alissa E M, Ferns G A. Dietary fruits and vegetables and cardiovascular diseases risk. Crit Rev Food Sci Nutr, 2017; 57(9): 1950–1962.10.1080/10408398.2015.1040487Search in Google Scholar PubMed
[118] Kargin D, Tomaino L, Serra-Majem L. Experimental outcomes of the mediterranean diet: lessons learned from the predimed randomized controlled trial. Nutrients, 2019; 11(12): 2991.10.3390/nu11122991Search in Google Scholar PubMed PubMed Central
[119] Wang J, Lin X, Bloomgarden Z T, et al. The Jiangnan diet, a healthy diet pattern for Chinese. J Diabetes, 2020; 12(5): 365–371.10.1111/1753-0407.13015Search in Google Scholar PubMed PubMed Central
[120] D'Souza M S, Dong T A, Ragazzo G, et al. From fad to fact: evaluating the impact of emerging diets on the prevention of cardiovascular disease. Am J Med, 2020; 133(10): 1126–1134.10.1016/j.amjmed.2020.05.017Search in Google Scholar PubMed
© 2022 Yukai Cao et al., published by Sciendo
This work is licensed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.
Articles in the same Issue
- Perspective
- “Nothing burns like the cold”: Cardiovascular disease in frigid zones
- Promoting factors behind hypertension in cold areas
- Cold stress-regulated immune responses: Insights, challenges, and perspectives
- Increased risk of cardiovascular disease in cold temperatures
- Review
- Pathogenesis and preventive measures of environment-related cardiovascular disease in northern China
- Effect of aging on cardiovascular responses to cold stress in humans
- RNA modification by M6A methylation in cardiovascular diseases: Current trends and future directions
- Original Article
- Extreme temperature increases the severity of intracerebral hemorrhage: An analysis based on the cold region of China
- Meta Analysis
- The influence of extreme cold ambient temperature on out of hospital cardiac arrest: A systemic review and meta-analysis
Articles in the same Issue
- Perspective
- “Nothing burns like the cold”: Cardiovascular disease in frigid zones
- Promoting factors behind hypertension in cold areas
- Cold stress-regulated immune responses: Insights, challenges, and perspectives
- Increased risk of cardiovascular disease in cold temperatures
- Review
- Pathogenesis and preventive measures of environment-related cardiovascular disease in northern China
- Effect of aging on cardiovascular responses to cold stress in humans
- RNA modification by M6A methylation in cardiovascular diseases: Current trends and future directions
- Original Article
- Extreme temperature increases the severity of intracerebral hemorrhage: An analysis based on the cold region of China
- Meta Analysis
- The influence of extreme cold ambient temperature on out of hospital cardiac arrest: A systemic review and meta-analysis