Open Access

Dysfunction of the thymus in mice with hypertension

  • Authors:
    • Xianliang Dai
    • Shuaibo Huang
    • Zhiqing He
    • Feng Wu
    • Ru Ding
    • Yihong Chen
    • Chun Liang
    • Zonggui Wu
  • View Affiliations

  • Published online on: February 15, 2017     https://doi.org/10.3892/etm.2017.4125
  • Pages: 1386-1392
  • Copyright: © Dai et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The aim of this study was to evaluate thymus function in mice with hypertension. A total of 60 C57BL/6J mice were randomized into control, sham surgery and two‑kidney, one‑clip groups (n=20 in each). At 4 or 8 weeks after surgery, mice were sacrificed, and blood, spleens, kidneys and thymuses were harvested. The results of reverse transcription-quantitative polymerase chain reaction analysis revealed that the mRNA levels of Forkhead box protein N1 (Foxn1) and autoimmune regulator (AIRE) in the thymus tissue of mice from the HTN group were significantly lower than those from the control group at 4 and 8 weeks (P<0.05). Foxn1 and AIRE expression was also reduced in the sham surgery group at 4 weeks after surgery, but had recovered 4 weeks later. Similar results were observed for the expression of signal‑joint T cell receptor excision circles and the percentages of T cell subsets. The present study indicates that impaired thymus function is associated with hypertension in mice, which suggests that thymus function may be a novel target for the treatment of patients with hypertension.

Introduction

Hypertension is a multifactorial disease associated with various types of cells and affected by multiple factors (1,2). It is characterized by elevated arterial pressure, and may be complicated by damage to the heart, blood vessels, brain, kidneys, retinas and other target organs, and metabolic changes in systemic disease (3). Patients with hypertension exhibit an increased incidence of heart and cerebrovascular disease and associated mortality; therefore, the ultimate goal of antihypertensive therapy is to reduce these risks by controlling blood pressure (4).

In 1970, Ebringer and Doyle reported that 30% of patients with hypertension exhibited increased serum immunoglobulin levels (5) and, with the rapid development of clinical immunology, it has been demonstrated that hypertension is associated with immune dysfunction, which suggests that immune factors are associated with the complications of hypertension (6). Previous studies have revealed that T cells are able to induce high blood pressure, vascular disorders and kidney disease and that possible mechanisms for this may include the release of cytokines, which directly affect vascular and renal function or indirectly stimulate cells to release additional cytokines, and induce the infiltration of inflammatory cells (7,8). The thymus is a primary immune organ, in which T cells are generated and matured (9); therefore, high blood pressure is likely to be associated with thymus function. A recent study has revealed that the thymus atrophies and becomes dysfunctional with age (10). A thymus transcription factor, Forkhead box protein N1 (Foxn1) is an important factor for complete physiological function of the thymus (11). As the thymus atrophies, the expression of the thymus aging-associated gene Foxn1 decreases, leading to a downregulation of Foxn1 with age (12). Increased expression of Foxn1 has been shown to improve thymus function, and potentially promote regeneration of the thymus (13), suggesting that Foxn1 may serve a role in high blood pressure. A previous study reported that atrophy of the thymus was observed in mice with hypertension (14); however, the specific changes in thymus function remain to be elucidated.

The autoimmune regulator (AIRE) is expressed in the thymus, particularly in thymic medullary epithelial cells. The expression profile of peripheral tissue antigens is affected by AIRE (15), and AIRE expression levels have been shown to have functional effects in the thymus (16).

The thymus is a vital immune organ, serving an important role in human health. With increasing age, the thymus gradually shrinks and its functionality declines or is lost completely (17). Thymus function may be used as a proxy to indicate the condition of a patient's immune system to a certain degree. At the CD4CD8-phase, thymocytes begin the rearrangement of their T cell receptor (TCR) α chain, and 95% of T cells are TCRαβ (18). The signal-joint (sj) TCR excision circle (TREC) is an extra-chromosomal DNA fragment ring, which is generated when the TCRδ gene is deleted prior to TCRα gene rearrangement. It is stable but not replicated, so with increased cell generation and dilution, the level of sjTREC is highest in T cell populations most recently generated, and is lower in cell groups after the cell division cycle (1921). The prevalence of sjTREC therefore provides an indication of the number of naive T cells during functional TCR formation. The presence of naive T cells, which enable the immune system to recognize all antigens, serves as an indicator of the generation and output of T cells; therefore, the level of sjTREC may reflect the functional status of the thymus (22,23).

To date, research into hypertension has predominantly focused on the changes in the renin-angiotensin system (24,25), with less investigation of changes in immune mechanisms and organs being performed. Investigating the changes to immune organs in patients with hypertension may identify a novel target for the treatment of hypertension through immune function.

Materials and methods

Animals

A total of 60 8-week-old C57BL/6J male mice with a mean weight of 20 g were purchased from Shanghai SLAC Laboratory Animal Co., Ltd. (Shanghai, China). Animals were housed in a climate-controlled, light-regulated space with 12-hour day and night cycles. They were fed a normal mouse chow and water ad libitum. Animal protocols were approved by the ethics committee of Second Military Medical University (Shanghai, China), and performed according to the Guide for the Care and Use of Laboratory Animals published by the United States National Institutes of Health (NIH Publication no. 85–23, revised 1996).

Induction of two-kidney, one-clip hypertension

Following 1 week of acclimation, mice were randomly divided into the control, sham surgery and two-kidney, one-clip (HTN) groups (n=20 in each). As previously described (26), the mice in the sham surgery and HTN groups were anesthetized with sodium pentobarbital (50 mg/kg; intraperitoneally; cat. no. P3761; Sigma-Aldrich; Merck Millipore, Darmstadt, Germany) the left kidney was exposed via flank incision, renal arteries were separated and a silver clip was placed around the left renal artery. Mice in the sham surgery group underwent an identical surgical procedure with the exception of the arterial clip. At 4 weeks post-surgery, 10 mice from each group were fasted overnight, anesthetized with sodium pentobarbital (50 mg/kg; intraperitoneally) and sacrificed via carotid puncture. The remaining mice were fasted overnight and sacrificed as above at 8 weeks post-surgery. Blood samples were harvested from the carotid artery, and organs of interest (spleen, kidneys and thymus) were immediately harvested, blotted dry, weighed and stored at −80°C until required. Blood samples and spleens were prepared as previously described (27,28), and organ indices were calculated using the following formula: Organ index=organ weight/body weight ×1,000.

Measurement of blood pressure

The systolic blood pressure (SBP) and diastolic blood pressure (DBP) of mice in each group were measured weekly using a non-invasive computerized tail-cuff system (ALC-NIBP; Shanghai Alcott Biotech Co., Ltd., Shanghai, China). The SBP and DBP of each mouse were measured three times and the mean was calculated.

DNA and RNA extraction, and reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was extracted from the thymus of each mouse using TRIzol reagent. The purity of obtained RNA was estimated using a NanoDrop 2000c spectrophotometer (Thermo Fisher Scientific, Inc., Wilmington, DE, USA), and RNA with an A260/A280 ratio >1.8 was used for cDNA synthesis. First strand cDNA synthesis was performed using the First Strand cDNA Synthesis kit (cat. no. 6210A; Takara Bio, Inc., Otsu, Japan) according to the manufacturer's protocol. qPCR was performed using SYBR Green PCR Master Mix iQ (cat. no. 170-8882AP; Bio-Rad Laboratories, Inc., Hercules, CA, USA) and the CFX Connect Real-Time PCR System (Bio-Rad Laboratories, Inc., Hercules, CA, USA), according to the manufacturer's protocols. qPCR conditions were as follows: 3 min at 95°C, followed by two-step PCR at 95°C for 10 sec and 58°C for 30 sec for 50 cycles, with fluorescence monitoring at the end of each elongation step. All primers were obtained from Thermo Fisher Scientific, Inc. (Waltham, MA, USA), and are listed in Table I. Relative mRNA expression of target genes was calculated using the 2−∆∆Cq method (29). Target sequences were normalized to β-actin in multiplexed reactions performed in duplicate.

Table I.

Oligonucleotide primers used for reverse transcription-quantitative polymerase chain reaction.

Table I.

Oligonucleotide primers used for reverse transcription-quantitative polymerase chain reaction.

GeneForwardReverse
m β-actin 5′-GGTCATCACTATTGGCAACG-3′ 5′-ACGGATGTCAACGTCACACT-3′
m Foxn1 5′-TGACGGAGCACTTCCCTTAC-3′ 5′-GACAGGTTATGGCGAACAGAA-3′
m AIRE 5′-GGTTCCTCCCCTTCCATC-3′ 5′-GGCACACTCATCCTCGTTCT-3′
m sjTREC 5′-CATTGCCTTTGAACCAAGCTG-3′ 5′-TTATGCACAGGGTGCAGGTG-3′
m RAG2 5′-TGACGTGGTGTATAGTCGA-3′ 5′-TCCTGAAGTTCTGGGAGA-3′

[i] m, murine; Foxn1, forkhead box protein N1; AIRE, autoimmune regulator; sjTREC, signal joint T cell receptor excision circle; RAG2, recombination activating gene 2.

Quantification of TRECs

Genomic DNA was extracted from thymocytes using the Wizard Genomic DNA Purification kit (cat. no. A1125; Promega Corporation, Madison, WI, USA). qPCR was performed using a CFX Connect real-time PCR System (Bio-Rad Laboratories, Inc.) to detect the number of TRECs. Each amplification was performed using SYBR-Green PCR Master Mix iQ (cat. no. 170-8882AP; Bio-Rad Laboratories, Inc.) according to the manufacturer's protocol. The PCR conditions were as follows: 5 min at 95°C, followed by three-step PCR: 95°C for 15 sec, 60°C for 40 sec and 72°C for 30 sec for 45 cycles, with fluorescence monitoring at the end of each elongation step. PCR data was analyzed using SDS software (version 2.2.3; Applied Biosystems; Thermo Fisher Scientific, Inc.). The PCR product of sjTREC and recombination activating protein 2 (RAG2) sequence was cloned into the PCR3.1 plasmid (Invitrogen; Thermo Fisher Scientific, Inc.) at known concentrations: 106, 105, 104, 103 and 102 copies/µl. Target sequences were normalized to β-actin in multiplexed reactions performed in duplicate. Each sample was performed in triplicate and the data expressed as the number of copies of sjTREC/105 cells based on the mouse genome of 2.6×109 bp (30). All primers were obtained from Thermo Fisher Scientific, Inc., and primer sequences are displayed in Table I.

Flow cytometric analysis

Blood cells and splenocytes were incubated with the appropriate antibodies as previously described (31,32) and flow cytometry was performed using a FACSCalibur cell analyzer (BD Biosciences, Franklin Lakes, NJ, USA). Antibodies (all BioLegend, Inc., San Diego, CA, USA) used are as follows: Alexa Fluor 488 anti-mouse CD3 (cat. no. 100210), PreCP/Cy5.5 anti-mouse CD4 (cat. no. 100540), Alexa Fluor 647 anti-mouse/human CD44 (cat. no. 103018) and PE anti-mouse CD62 L (cat. no. 104408). All antibodies were used at a dilution of 1:12. Data were analyzed using FlowJo software, (version 10.0.7; Tree Star, Inc., Ashland, OR, USA).

Hematoxylin and eosin (H&E) staining

Standard H&E staining was performed according to the manufacturer's protocol (Beijing Xin Hua Luyuan Science and Technology, Ltd., Beijing , China), and viewed under an Olympus IX51 fluorescence microscope (OLYMPUS CORPORATION, Japan).

Statistical analysis

Data are presented as the mean ± standard error of the mean. Data analysis was performed using Student's t-test for comparison between two groups and one-way analysis of variance was performed to test for differences among multiple groups, followed by Fisher's least significant difference test using SPSS 19.0 (IBM SPSS, Armonk, NY, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Blood pressure

At 4 weeks post-surgery, the SBP and DBP of the HTN group were significantly increased (P<0.001) compared with those of the sham and control groups, and reached the hypertension criteria (SBP >140 mmHg; Fig. 1) (27), indicating that the hypertensive models were successfully established.

Changes in thymus function

Organs of interest (spleen, kidneys and thymus) were immediately removed following sacrifice and the organ indices were calculated. Thymus atrophy was observed in the HTN group as illustrated in Fig. 2A-C. The thymic index of the HTN group exhibited no significant differences at 4 or 8 weeks post-surgery compared with the control and sham groups (Table II).

Table II.

Body, thymus and spleen weight, and thymus index of mice.

Table II.

Body, thymus and spleen weight, and thymus index of mice.

Control groupSham groupHTN group



ParametersWk 4 (n=10)Wk 8 (n=10)Wk 4 (n=10)Wk 8 (n=10)Wk 4 (n=10)Wk 8 (n=10)
Body weight, g21.11±0.8226.32±3.6220.32±0.7625.67±1.9920.40±1.2226.33±2.17
Thymus weight, g0.043±0.0510.039±0.0110.038±0.00290.037±0.00610.041±0.00710.038±0.008
Spleen weight, g0.079±0.0130.087±0.020.076±0.00580.079±0.0040.084±0.00970.086±0.0082
Thymus index1.83±0.4751.588±0.3741.818±0.2661.506±0.221.834±0.3541.575±0.225

[i] Values are expressed as the mean ± standard error of the mean. Wk, weeks post-surgery; HTN, 2-kidney, 1-clip hypertension.

H&E staining of the thymus revealed stereotypical deterioration of the thymic epithelial compartment, reduced distinction between the cortical and medullary regions and a reduction in the number of medullary islets per thymic lobe in the HTN group (Fig. 2D-F).

At 4 and 8 weeks post-surgery, the expression of Foxn1 and AIRE mRNA in the thymus was significantly downregulated (4 weeks, Foxn1 and AIRE, P<0.001; 8 weeks, Foxn1 P<0.001 and AIRE P<0.05) in the HTN group compared with the control group (Fig. 2G and H). There was no significant difference in the expression of Foxn1 and AIRE mRNA at 8 weeks post-surgery between the sham and HTN groups; however, there was a significant difference (P<0.001) between the sham and control groups at 4 weeks post-surgery. Patterns of sjTREC expression were similar to those of Foxn1 and AIRE, as shown in Fig. 2I and J. Expression of sjTREC in the thymus was significantly downregulated in the HTN group compared with the control group at 4 and 8 weeks post-surgery (both P<0.001), and a significant difference (P<0.001) was observed between the sham and HTN groups at 8 weeks post-surgery. Furthermore, a significant difference (P<0.001) was observed between the control and sham groups at 4 weeks post-surgery.

Changes in T cell subsets evaluated by flow cytometry

Flow cytometric analysis was performed on blood and splenocytes, as shown in Fig. 3A and B. Significant differences were observed between the T cell subset percentages in the blood and splenocytes of the control group compared with the other groups at 4 weeks post-surgery [blood and splenocytes, effector memory T cells (TEM) P<0.001; splenocytes, naïve T cells P<0.05], whereas there was no significant difference between the HTN and sham groups (Fig. 3C). T cell subsets in the HTN group were significantly different (blood, TEM P<0.05; splenocytes, TEM and naïve T cells P<0.001) compared with those in the sham and control groups, whereas the other two groups exhibited no significant differences from each other 8 weeks post-surgery (Fig. 3D).

Discussion

Various cells and influencing factors are associated with the progression of hypertension. Previous studies have demonstrated that hypertension is associated with immunity. The present study assessed the changes in thymus function in mice with hypertension, and the results demonstrated that thymus functionality is significantly decreased in mice with hypertension, suggesting that the immune system is associated with the process of hypertension. Therefore, treating the thymus may improve functionality, promote immune system function and ameliorate hypertension, potentially providing a novel therapeutic target for the treatment of hypertension.

The present study demonstrated that mice with hypertension exhibited had decreased immune function due to atrophy of the thymus, based on observations of the gross thymus, thymic pathology slices, the thymus index and expression of the anti-thymus aging related gene Foxn1.

As the development and maturation of T cell occurs in the thymus, changes to the T cell subsets are also indicative of abnormal thymus function (3335). sjTREC may be used to evaluate the output of naive T cells and thereby serve as a proxy for thymus output function, indirectly demonstrating thymus dysfunction (3638). Hypertension is acknowledged to be a chronic disease (39,40); however, in the present study hypertension was induced in mice via the two-kidney one-clip method only 4 or 8 weeks prior to sacrifice, which may explain why the thymic index showed no significant differences between the three groups. In future research, it may be better to allow a longer period of time between surgery and sacrifice to investigate whether the thymic index is significantly altered.

Previous studies have demonstrated that Foxn1 is exclusively expressed in the thymus (4143). Increasing Foxn1 expression may induce partial or total recovery of thymus function (13); therefore, increasing the expression of Foxn1 may raise the cure rate and provide new diagnostic methods for hypertension and other immune-related diseases, such as asthenic bulbar paralysis.

A number of studies have suggested that stress may induce a decline in thymus function (4446), by demonstrating that stress induces a transient reduction in thymus functionality; however, when individuals recover to a non-stressed state, thymus function is restored. However, these studies did not explore the mechanisms responsible for the transient effects of stress on thymus function. This may be associated with the increase of angiotensin, renin angiotensin and thyroid hormone, and may also be associated with stimulation of sympathetic nerves. Further research is required to confirm this hypothesis.

In the present study, the two-kidney, one-clip model of hypertension was used, which is secondary renal hypertension; therefore the results demonstrate that the immune system may be associated with the pathological process of renal hypertension. However, it remains to be determined whether the immune system has an association with primary hypertension and other types of secondary hypertension.

In conclusion, in vivo experiments on mice with hypertension suggest that there may be a correlation between hypertension and immune dysfunction. Improving immune system or thymus function may enhance the therapeutic effects of treatments for hypertension, which may provide a novel therapeutic target and treatment concept for treating hypertension, and potentially lead to the development of an anti-hypertension vaccine.

Acknowledgements

The present study was supported by the National Natural Science Foundation of China (grant nos. 81130065, 81072981, 30971101, 31171130 and 30900528), the Shanghai Pujiang Talent Program (D-15), the Shanghai Key Basic Research Program (grant no. 10411956,500), and the Shanghai Project of International Cooperation and Exchange (grant no. 10410701700).

References

1 

Vaziri ND, Wang XQ, Oveisi F and Rad B: Induction of oxidative stress by glutathione depletion causes severe hypertension in normal rats. Hypertension. 36:142–146. 2000. View Article : Google Scholar : PubMed/NCBI

2 

Vinh A, Chen W, Blinder Y, Weiss D, Taylor WR, Goronzy JJ, Weyand CM, Harrison DG and Guzik TJ: Inhibition and genetic ablation of the B7/CD28 T cell costimulation axis prevents experimental hypertension. Ccirculation. 122:2529–2537. 2010. View Article : Google Scholar

3 

Cavasin MA, Liao TD, Yang XP, Yang JJ and Carretero OA: Decreased endogenous levels of Ac-SDKP promote organ fibrosis. Hypertension. 50:130–136. 2007. View Article : Google Scholar : PubMed/NCBI

4 

Neutel JM, Giles T, Punzi H, Weiss RJ, Li H and Finck A: Long-term safety of nebivolol and valsartan combination therapy in patients with hypertension: An open-label, single-arm, multicenter study. J Am Soc Hypertens. 8:915–920. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Ebringer A and Doyle AE: Raised serum IgG levels in hypertension. Br Med J. 2:146–148. 1970. View Article : Google Scholar : PubMed/NCBI

6 

De Ciuceis C, Rossini C, La Boria E, Porteri E, Petroboni B, Gavazzi A, Sarkar A, Rosei EA and Rizzoni D: Immune Mechanisms in Hypertension. High Blood Press Cardiovasc Prev. 21:227–234. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Leibowitz A and Schiffrin EL: Immune mechanisms in hypertension. Curr Hypertens Rep. 13:465–472. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Sonmez A, Kisa U, Uckaya G, Eyileten T, Comert B, Koc B, Kocabalkan F and Ozata M: Effects of losartan treatment on T-cell activities and plasma leptin concentrations in primary hypertension. J Renin Angiotensin Aldosterone Syst. 2:112–116. 2001.PubMed/NCBI

9 

Walters SN, Webster KE, Daley S and Grey ST: A role for intrathymic B cells in the generation of natural regulatory T cells. J Immunol. 193:170–176. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Ruan L, Zhang Z, Mu L, Burnley P, Wang L, Coder B, Zhuge Q and Su DM: Biological significance of FoxN1 gain-of-function mutations during T and B lymphopoiesis in juvenile mice. Cell Death Dis. 5:e14572014. View Article : Google Scholar : PubMed/NCBI

11 

Bredenkamp N, Ulyanchenko S, O'Neill KE, Manley NR, Vaidya HJ and Blackburn CC: An organized and functional thymus generated from FOXN1-reprogrammed fibroblasts. Nat Cell Biol. 16:902–908. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Zook EC, Krishack PA, Zhang S, Zeleznik-Le NJ, Firulli AB, Witte PL and Le PT: Overexpression of Foxn1 attenuates age-associated thymic involution and prevents the expansion of peripheral CD4 memory T cells. Blood. 118:5723–5731. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Bredenkamp N, Nowell CS and Blackburn CC: Regeneration of the aged thymus by a single transcription factor. Development. 141:1627–1637. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Fukuda S, Tsuchikura S and Iida H: Age-related changes in blood pressure, hematological values, concentrations of serum biochemical constituents and weights of organs in the SHR/Izm, SHRSP/Izm and WKY/Izm. Exp Anim. 53:67–72. 2004. View Article : Google Scholar : PubMed/NCBI

15 

Lovewell TR, McDonagh AJ, Messenger AG, Azzouz M and Tazi-Ahnini R: The AIRE-230Y Polymorphism affects aire transcriptional activity: Potential influence on aire function in the thymus. PLoS One. 10:e01274762015. View Article : Google Scholar : PubMed/NCBI

16 

Liston A, Gray DH, Lesage S, Fletcher AL, Wilson J, Webster KE, Scott HS, Boyd RL, Peltonen L and Goodnow CC: Gene dosage-limiting role of Aire in thymic expression, clonal deletion, and organ-specific autoimmunity. J Exp Med. 200:1015–1026. 2004. View Article : Google Scholar : PubMed/NCBI

17 

Berthiaume F, Aparicio CL, Eungdamrong J and Yarmush ML: Age- and disease-related decline in immune function: An opportunity for ‘thymus-boosting’ therapies. Tissue Eng. 5:499–514. 1999. View Article : Google Scholar : PubMed/NCBI

18 

Lang PO, Govind S, Dramé M and Aspinall R: Measuring the TREC ratio in dried blood spot samples: Intra- and inter-filter paper cards reproducibility. J Immunol Methods. 389:1–8. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Lynch HE and Sempowski GD: Molecular measurement of T cell receptor excision circles. Methods Mol Biol. 979:147–159. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Hug A, Korporal M, Schröder I, Haas J, Glatz K, Storch-Hagenlocher B and Wildemann B: Thymic export function and T cell homeostasis in patients with relapsing remitting multiple sclerosis. J Immunol. 171:432–437. 2003. View Article : Google Scholar : PubMed/NCBI

21 

Sodora DL, Douek DC, Silvestri G, Montgomery L, Rosenzweig M, Igarashi T, Bernacky B, Johnson RP, Feinberg MB, Martin MA and Koup RA: Quantification of thymic function by measuring T cell receptor excision circles within peripheral blood and lymphoid tissues in monkeys. Eur J Immunol. 30:1145–1153. 2000. View Article : Google Scholar : PubMed/NCBI

22 

Chiarini M, Sottini A, Bertoli D, Serana F, Caimi L, Rasia S, Capra R and Imberti L: Newly produced T and B lymphocytes and T-cell receptor repertoire diversity are reduced in peripheral blood of fingolimod-treated multiple sclerosis patients. Mult Scler. 21:726–734. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Al-Harthi L, Marchetti G, Steffens CM, Poulin J, Sékaly R and Landay A: Detection of T cell receptor circles (TRECs) as biomarkers for de novo T cell synthesis using a quantitative polymerase chain reaction-enzyme linked immunosorbent assay (PCR-ELISA). J Immunol Methods. 237:187–197. 2000. View Article : Google Scholar : PubMed/NCBI

24 

Jiménez PM, Conde C, Casanegra A, Romero C, Tabares AH and Orías M: Association of ACE genotype and predominantly diastolic hypertension: A preliminary study. J Renin Angiotensin Aldosterone Syst. 8:42–44. 2007. View Article : Google Scholar : PubMed/NCBI

25 

Campagnaro BP, Gava AL, Meyrelles SS and Vasquez EC: Cardiac-autonomic imbalance and baroreflex dysfunction in the renovascular Angiotensin-dependent hypertensive mouse. Int J Hypertens. 2012:9681232012. View Article : Google Scholar : PubMed/NCBI

26 

Zhou YB, Sun HJ, Chen D, Liu TY, Han Y, Wang JJ, Tang CS, Kang YM and Zhu GQ: Intermedin in paraventricular nucleus attenuates sympathetic activity and blood pressure via nitric oxide in hypertensive rats. Hypertension. 63:330–337. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Wei Z, Spizzo I, Diep H, Drummond GR, Widdop RE and Vinh A: Differential phenotypes of tissue-infiltrating T cells during angiotensin II-induced hypertension in Mice. PLoS One. 9:e1148952014. View Article : Google Scholar : PubMed/NCBI

28 

Guzik TJ, Hoch NE, Brown KA, McCann LA, Rahman A, Dikalov S, Goronzy J, Weyand C and Harrison DG: Role of the T cell in the genesis of angiotensin II-induced hypertension and vascular dysfunction. J Exp Med. 204:2449–2460. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-tie quantitative PCR and the 2(−Delta Delta C(T)) Method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

30 

Ortman CL, Dittmar KA, Witte PL and Le PT: Molecular characterization of the mouse involuted thymus: Aberrations in expression of transcription regulators in thymocyte and epithelial compartments. Int Immunol. 14:813–822. 2002. View Article : Google Scholar : PubMed/NCBI

31 

McDonald CA, Payne NL, Sun G, Moussa L, Siatskas C, Lim R, Wallace EM, Jenkin G and Bernard CC: Immuno-suppressive potential of human amnion epithelial cells in the treatment of experimental autoimmune encephalomyelitis. J Neuroinflammation. 12:1122015. View Article : Google Scholar : PubMed/NCBI

32 

Ammirati E, Cianflone D, Vecchio V, Banfi M, Vermi AC, De Metrio M, Grigore L, Pellegatta F, Pirillo A, Garlaschelli K, et al: Effector memory t cells are associated with atherosclerosis in humans and animal models. J Am Heart Assoc. 1:27–41. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Bernardi AI, Andersson A, Stubelius A, Grahnemo L, Carlsten H and Islander U: Selective estrogen receptor modulators in T cell development and T cell dependent inflammation. Immunobiology. 220:1122–1128. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Petrie HT: Cell migration and the control of post-natal T-cell lymphopoiesis in the thymus. Nat Rev Immunol. 3:859–866. 2003. View Article : Google Scholar : PubMed/NCBI

35 

Klausmann S, Sydler T, Summerfield A, Lewis FI, Weilenmann R, Sidler X and Brugnera E: T-cell reprogramming through targeted CD4-coreceptor and T-cell receptor expression on maturing thymocytes by latent Circoviridae family member porcine circovirus type 2 cell infections in the thymus. Emerg Microbes Infect. 4:e152015. View Article : Google Scholar : PubMed/NCBI

36 

Ringhoffer S, Rojewski M, Döhner H, Bunjes D and Ringhoffer M: T-cell reconstitution after allogeneic stem cell transplantation: Assessment by measurement of the sjTREC/βTREC ratio and thymic naïve T cells. Haematologica. 98:1600–1608. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Ou XL, Gao J, Wang H, Wang HS, Lu HL and Sun HY: Predicting human age with bloodstains by sjTREC quantification. PLoS One. 7:e424122012. View Article : Google Scholar : PubMed/NCBI

38 

Douek DC, McFarland RD, Keiser PH, Gage EA, Massey JM, Haynes BF, Polis MA, Haase AT, Feinberg MB, Sullivan JL, et al: Changes in thymic function with age and during the treatment of HIV infection. Nature. 396:690–695. 1998. View Article : Google Scholar : PubMed/NCBI

39 

Feng YJ, Wang HC, Li YC and Zhao WH: Hypertension screening and follow-up management by primary health care system among chinese population aged 35 years and above. Biomed Environ Sci. 28:330–340. 2015.PubMed/NCBI

40 

Yang PR, Shih WT, Chu YH, Chen PC and Wu CY: Frequency and co-prescription pattern of Chinese herbal products for hypertension in Taiwan: A Cohort study. BMC Complement Altern Med. 15:1632015. View Article : Google Scholar : PubMed/NCBI

41 

Romano R, Palamaro L, Fusco A, Giardino G, Gallo V, Del Vecchio L and Pignata C: FOXN1: A master regulator gene of thymic epithelial development program. Front Immunol. 4:1872013. View Article : Google Scholar : PubMed/NCBI

42 

Romano R, Palamaro L, Fusco A, Iannace L, Maio S, Vigliano I, Giardino G and Pignata C: From murine to human nude/SCID: the thymus, T-cell development and the missing link. Clin Dev Immunol. 2012:4671012012. View Article : Google Scholar : PubMed/NCBI

43 

Jin X, Nowell CS, Ulyanchenko S, Stenhouse FH and Blackburn CC: Long-Term Persistence of functional thymic epithelial progenitor cells in vivo under conditions of low FOXN1 expression. PLoS One. 9:e1148422014. View Article : Google Scholar : PubMed/NCBI

44 

Misa-Agustiño MJ, Leiro-Vidal JM, Gomez-Amoza JL, Jorge-Mora MT, Jorge-Barreiro FJ, Salas-Sánchez AA, Ares-Pena FJ and López-Martín E: EMF radiation at 2450 MHz triggers changes in the morphology and expression of heat shock proteins and glucocorticoid receptors in rat thymus. Life Sci. 127:1–11. 2015. View Article : Google Scholar : PubMed/NCBI

45 

Gupta S, Haldar C and Ahmad R: Photoperiodic regulation of nuclear melatonin receptor RORα in lymphoid organs of a tropical rodent Funambulus pennanti: Role in seasonal oxidative stress. J Photochem Photobiol B. 142:141–153. 2015. View Article : Google Scholar : PubMed/NCBI

46 

Novoselova EG, Lunin SM, Khrenov MO, Parfenyuk SB, Novoselova TV, Shenkman BS and Fesenko EE: Changes in immune cell signalling, apoptosis and stress response functions in mice returned from the BION-M1 mission in space. Immunobiology. 220:500–509. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2017
Volume 13 Issue 4

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Dai X, Huang S, He Z, Wu F, Ding R, Chen Y, Liang C and Wu Z: Dysfunction of the thymus in mice with hypertension. Exp Ther Med 13: 1386-1392, 2017
APA
Dai, X., Huang, S., He, Z., Wu, F., Ding, R., Chen, Y. ... Wu, Z. (2017). Dysfunction of the thymus in mice with hypertension. Experimental and Therapeutic Medicine, 13, 1386-1392. https://doi.org/10.3892/etm.2017.4125
MLA
Dai, X., Huang, S., He, Z., Wu, F., Ding, R., Chen, Y., Liang, C., Wu, Z."Dysfunction of the thymus in mice with hypertension". Experimental and Therapeutic Medicine 13.4 (2017): 1386-1392.
Chicago
Dai, X., Huang, S., He, Z., Wu, F., Ding, R., Chen, Y., Liang, C., Wu, Z."Dysfunction of the thymus in mice with hypertension". Experimental and Therapeutic Medicine 13, no. 4 (2017): 1386-1392. https://doi.org/10.3892/etm.2017.4125