Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2883 2022-08-30 06:19:13 |
2 format correct -27 word(s) 2856 2022-08-31 04:16:40 | |
3 format correct + 3 word(s) 2859 2022-09-01 12:34:46 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Ohara, H.;  Nabika, T. Genetic Modifications to Alter Blood Pressure Level. Encyclopedia. Available online: https://encyclopedia.pub/entry/26651 (accessed on 15 May 2024).
Ohara H,  Nabika T. Genetic Modifications to Alter Blood Pressure Level. Encyclopedia. Available at: https://encyclopedia.pub/entry/26651. Accessed May 15, 2024.
Ohara, Hiroki, Toru Nabika. "Genetic Modifications to Alter Blood Pressure Level" Encyclopedia, https://encyclopedia.pub/entry/26651 (accessed May 15, 2024).
Ohara, H., & Nabika, T. (2022, August 30). Genetic Modifications to Alter Blood Pressure Level. In Encyclopedia. https://encyclopedia.pub/entry/26651
Ohara, Hiroki and Toru Nabika. "Genetic Modifications to Alter Blood Pressure Level." Encyclopedia. Web. 30 August, 2022.
Genetic Modifications to Alter Blood Pressure Level
Edit

Genetic manipulation is one of the indispensable techniques to examine gene functions both in vitro and in vivo. In particular, cardiovascular phenotypes such as blood pressure cannot be evaluated in vitro system, necessitating the creation of transgenic or gene-targeted knock-out and knock-in experimental animals to understand the pathophysiological roles of specific genes on the disease conditions. 

knock-out genome-editing SHR

1. Introduction

Hypertension is the leading preventable risk factor for cerebro-cardiovascular complications, including heart failure and stroke. Effective anti-hypertensive drugs with different pharmacological actions have been developed; nevertheless, it is deemed that there are 1.28 billion hypertensive patients globally and 0.7 billion or more patients are untreated [1]. Given the resulting mortality and disability as well as the high prevalence, hypertension is still a major public health burden in the world.
It is needless to say that gene-targeted knock-out (KO) and knock-in (KI) or transgenic rodent models have greatly contributed to understanding the pathophysiological basis of hypertension and its vascular complications. In particular, mice have been widely used as the best experimental animal since the gene engineering technique to create KO models was established for over 30 years ago. By contrast, it had been technically difficult to create KO rats for a long time because of the difficulty of rat ES cell culture. Recent advances in genome-editing technologies, however, have made it possible to easily create KO rats similar to mice [2][3]. Given that spontaneous cerebro-cardiovascular disease models, such as Dahl salt-sensitive (SS) and stroke-prone spontaneously hypertensive rats (SHRSP), have been commercially available, a genome-editing strategy using the rat disease models has much potential to clarify the novel pathogenesis of hypertension.

2. Mouse Models

Essential hypertension is a highly complex pathological condition that is formed by synergistic influences of multiple lifestyles, social, environmental, and genetic factors. Since blood pressure (BP) is collaboratively controlled by various organs and tissues, there are many studies that have investigated tissue (or cell)-specific roles of genes on BP regulation using conventional and conditional KO or transgenic mice. In contrast to rats, no spontaneous hypertensive mouse models have been established; accordingly, angiotensin II (Ang II)-infused models have been widely used to investigate the pathogenesis of Ang II-related hypertension. Deoxycorticosterone acetate (DOCA)-salt or high-salt diet (usually containing 4% or 8% NaCl) models have been also used to investigate the pathogenesis of salt-sensitive hypertension. 

2.1. Kidney

The kidney plays pivotal roles in arterial BP regulation by controlling blood volume and plasma electrolyte balance. Activities of the renin–angiotensin–aldosterone system (RAAS) and mineral transporters (Na+/H+ exchanger; NHE, Na+–K+–Cl co-transporter; NKCC, Na+–Cl co-transporter; NCC, epithelial sodium channel; ENaC, etc.) distributed along with nephron are important for physiological BP regulation; thus, genes that may regulate those activities have been widely investigated (Table 1).
Ang II regulates BP via Ang II type 1 receptor (Agtr1a, AT1R). As BP lowering effects were observed in proximal tubules (PT) or collecting duct (CD)-specific KO mice [4][5], blockade of AT1R signaling in renal epithelial cells would be a pharmacological target for hypertension therapy. Of note, AT1R-associated protein (Agtrap), which is widely distributed along renal tubules, has been found to suppress AT1R signaling by facilitating internalization of AT1R resulting in decreased cell surface expression of AT1R [6][7], suggesting that activation of endogenous AGTRAP has potential to reduce BP. In fact, it has been reported that the renal-specific overexpression and conventional KO mice show lower and higher BP phenotype compared with the wild-type (WT) control, respectively [8][9][10][11]. In contrast to the results in mice, however, the deletion in Dahl SS rats exacerbated renal damage under a 4% NaCl diet condition with no change in BP [12]. Although AGTRAP may play double-edged roles in reno-cardiovascular functions in a context-specific manner, it is a potential candidate gene located in a genome-wide association studies (GWAS) loci for BP in humans [12].
Although Ang II is the most well-known bioactive peptide hormone in the RAAS, (pro)renin and Ang-(1-7) produced by angiotensin-converting enzyme 2 (ACE2) are also known to regulate BP via its specific receptors. The (Pro)renin receptor (PRR) that specifically recognizes both prorenin and renin was cloned by Nguyen et al. in 2002 [13]. In the kidney, PRR is mainly expressed in renal vasculature, PT and distal tubules (DT), and CD and enhances the catalytic activity of (pro)renin that converts Ang I to Ang II, resulting in an increase in Ang II production [14]. Consistent with the physiological function of PRR, decreases in BP elevation induced by Ang II infusion have been observed in both tubular- and CD-specific KO mice through inhibition of ENaC activation [15][16][17]. Ang-(1-7) generated by mainly ACE2 is a vasoactive peptide that induces a vasodilation response by binding to Mas receptor [18]. Therefore, ACE2-Ang-(1-7)-Mas axis exerts a counteracting effect on Ang II that causes BP elevation. Ni et al. reported that conventional double KO of both ACE2 and Mas receptor in mice caused greater Ang II-induced BP elevation when compared with the WT littermates [19]. In addition, they also showed that the dual deletion of ACE2 and Mas receptor worsened hypertensive nephropathy, suggesting that ACE2-Ang-(1-7)-Mas receptor axis has protective roles in both the development of hypertension and the resulting hypertensive kidney injury.
Tubuloglomerular feedback (TGF) is an important physiological system to regulate long-term BP by sensing blood volume and electrolyte balance at the level of juxtaglomerular apparatus in each nephron [20]. Accumulating evidence has shown that local activities of renal oxide synthases (NOS), which produce a major chemical vasodilator NO, play an important role in the regulation of the TGF system. NOS families are composed of three isoforms, i.e., neuronal NOS (nNOS, encoded by Nos1), inducible NOS (iNOS, Nos2), and endothelial NOS (eNOS, Nos3). Although all the three isoforms are expressed in the kidney, Nos1 and Nos3 are thought to be major isoforms that physiologically participate in the TGF because of low baseline expression of Nos2. Interestingly, Lu et al. showed that macula densa-specific deletion of Nos1 exacerbated a high-salt diet-induced BP elevation under a condition of Ang II infusion accompanied by reduced glomerular filtration rate (GFR) and Na+ excretion [21]. It was also reported that local NOS1 activity at the macula densa contributed to a sex difference in BP response to Ang II [22]. Moreover, Hyndman et al. and Gao et al. have investigated renal-specific roles of NOS1 and NOS3 on BP regulation using CD-specific and nephron-specific KO mice, respectively [23][24]. They suggested that deletion of the two isoforms caused greater high-salt-induced BP elevation by enhancing ENaC [25] and NCC activities in the tubular cells, respectively.
Pathophysiological roles of NEDD4-2 (encoded by Nedd4l) and with-no-lysine kinases 1 and 4 (Wnk1 and Wnk4) in (salt-sensitive) hypertension have been well-investigated in humans as well as in rodent models. NEDD4-2 is an E3 ubiquitin ligase that ubiquitylates ENaC to down-regulate its cell surface expression and activity [26]. Although NEDD4-2 was initially found as a ENaC-specific regulator in the kidney [25], Ronzaud et al. reported that NEED4-2 also regulated NCC activity and its renal tubule-specific deletion caused salt-dependent hypertension [27]. Consequently, NEED4-2 is involved in the pathogenesis of salt-sensitive hypertension through the two-independent pathways that controls renal Na+ homeostasis. WNK1 and WNK4 are known to be responsible genes of pseudohypoaldosteronism type 2 (PHA2) that is caused by large deletions in intron 1 of WNK1 or gain-of function mutations in WNK4 [28]. Mechanistically, WNKs phosphorylate SPAK/OSR1, thereby activating NCC in the DT and resulting in increased Na+ reabsorption and salt-sensitive hypertension [28][29]; however, the molecular network may be a little complicated as a paradoxical role of kidney-specific WNK1 lacking a kinase domain on the development of salt-sensitive hypertension was reported [30]. Moreover, Mu et al. suggested a unique pathway involving salt-sensitive hypertension caused by epigenetic down-regulation of WNK4 [31]. In this context, kelch-like protein 3 (KLHL3) and cullin 3 (CUL3), which are the E3 ubiquitin ligase complex to degrade WNK, have also received much attention as target molecules to prevent salt-sensitive hypertension [28][29].
Unlike the local mechanisms in the kidney described above, Pan et al. uniquely identified the liver–kidney and liver–adipocytes axis to control BP via a hepatocytes-producing hormone, fibroblast growth factor 21 (FGF21), which has pleiotropic effects on glucose and lipid metabolism [32]. They found that FGF21 augmented peroxisome proliferator-activated receptor γ (PPARγ)-mediated activation of ACE2 in both the kidney and adipocytes; thereby, an increase in Ang-(1-7) production reduced both BP and vascular injury. Because FGF21 production was stimulated by Ang II, the FGF21–ACE2 axis may counteract Ang II-induced hypertension and the vascular injury. This might be a key mechanism in obesity-related hypertension.
Besides the above, multiple mechanisms have been proposed such as by circadian clock- [33][34], osmotic stress- [35], and genome-wide association study (GWAS)-related genes [36][37] as well.
Table 1. Target molecules in kidney.

References

  1. Zhou, B.; Carrillo-Larco, R.M.; Danaei, G.; Riley, L.M.; Paciorek, C.J.; Stevens, G.A.; Gregg, E.W.; Bennett, J.E.; Solomon, B.; Singleton, R.K.; et al. Worldwide trends in hypertension prevalence and progress in treatment and control from 1990 to 2019: A pooled analysis of 1201 population-representative studies with 104 million participants. Lancet 2021, 398, 957–980.
  2. Mashimo, T. Gene targeting technologies in rats: Zinc finger nucleases, transcription activator-like effector nucleases, and clustered regularly interspaced short palindromic repeats. Dev. Growth Differ. 2014, 56, 46–52.
  3. Kaneko, T.; Mashimo, T. Simple Genome Editing of Rodent Intact Embryos by Electroporation. PLoS ONE 2015, 10, e0142755.
  4. Stegbauer, J.; Chen, D.; Herrera, M.; Sparks, M.A.; Yang, T.; Konigshausen, E.; Gurley, S.B.; Coffman, T.M. Resistance to hypertension mediated by intercalated cells of the collecting duct. JCI Insight 2017, 2, e92720.
  5. Li, X.C.; Leite, A.P.O.; Zheng, X.; Zhao, C.; Chen, X.; Zhang, L.; Zhou, X.; Rubera, I.; Tauc, M.; Zhuo, J.L. Proximal Tubule-Specific Deletion of Angiotensin II Type 1a Receptors in the Kidney Attenuates Circulating and Intratubular Angiotensin II-Induced Hypertension in PT-Agtr1a−/− Mice. Hypertension 2021, 77, 1285–1298.
  6. Cui, T.; Nakagami, H.; Iwai, M.; Takeda, Y.; Shiuchi, T.; Tamura, K.; Daviet, L.; Horiuchi, M. ATRAP, novel AT1 receptor associated protein, enhances internalization of AT1 receptor and inhibits vascular smooth muscle cell growth. Biochem. Biophys. Res. Commun. 2000, 279, 938–941.
  7. Tsurumi, Y.; Tamura, K.; Tanaka, Y.; Koide, Y.; Sakai, M.; Yabana, M.; Noda, Y.; Hashimoto, T.; Kihara, M.; Hirawa, N.; et al. Interacting molecule of AT1 receptor, ATRAP, is colocalized with AT1 receptor in the mouse renal tubules. Kidney Int. 2006, 69, 488–494.
  8. Wakui, H.; Tamura, K.; Masuda, S.-I.; Tsurumi-Ikeya, Y.; Fujita, M.; Maeda, A.; Ohsawa, M.; Azushima, K.; Uneda, K.; Matsuda, M.; et al. Enhanced Angiotensin Receptor-Associated Protein in Renal Tubule Suppresses Angiotensin-Dependent Hypertension. Hypertension 2013, 61, 1203–1210.
  9. Ohsawa, M.; Tamura, K.; Wakui, H.; Maeda, A.; Dejima, T.; Kanaoka, T.; Azushima, K.; Uneda, K.; Tsurumi-Ikeya, Y.; Kobayashi, R.; et al. Deletion of the angiotensin II type 1 receptor–associated protein enhances renal sodium reabsorption and exacerbates angiotensin II–mediated hypertension. Kidney Int. 2014, 86, 570–581.
  10. Kobayashi, R.; Wakui, H.; Azushima, K.; Uneda, K.; Haku, S.; Ohki, K.; Haruhara, K.; Kinguchi, S.; Matsuda, M.; Ohsawa, M.; et al. An angiotensin II type 1 receptor binding molecule has a critical role in hypertension in a chronic kidney disease model. Kidney Int. 2017, 91, 1115–1125.
  11. Kinguchi, S.; Wakui, H.; Azushima, K.; Haruhara, K.; Koguchi, T.; Ohki, K.; Uneda, K.; Matsuda, M.; Haku, S.; Yamaji, T.; et al. Effects of ATRAP in Renal Proximal Tubules on Angiotensin-Dependent Hypertension. J. Am. Heart Assoc. 2019, 8, e012395.
  12. Flister, M.J.; Tsaih, S.W.; O’Meara, C.C.; Endres, B.; Hoffman, M.J.; Geurts, A.M.; Dwinell, M.R.; Lazar, J.; Jacob, H.J.; Moreno, C. Identifying multiple causative genes at a single GWAS locus. Genome Res. 2013, 23, 1996–2002.
  13. Nguyen, G.; Delarue, F.; Burcklé, C.; Bouzhir, L.; Giller, T.; Sraer, J.D. Pivotal role of the renin/prorenin receptor in angiotensin II production and cellular responses to renin. J. Clin. Investig. 2002, 109, 1417–1427.
  14. Ramkumar, N.; Kohan, D.E. The nephron (pro)renin receptor: Function and significance. Am. J. Physiol. Renal Physiol. 2016, 311, F1145–F1148.
  15. Ramkumar, N.; Stuart, D.; Mironova, E.; Bugay, V.; Wang, S.; Abraham, N.; Ichihara, A.; Stockand, J.D.; Kohan, D.E. Renal tubular epithelial cell prorenin receptor regulates blood pressure and sodium transport. Am. J. Physiol. Renal Physiol. 2016, 311, F186–F194.
  16. Peng, K.; Lu, X.; Wang, F.; Nau, A.; Chen, R.; Zhou, S.F.; Yang, T. Collecting duct (pro)renin receptor targets ENaC to mediate angiotensin II-induced hypertension. Am. J. Physiol. Renal Physiol. 2017, 312, F245–F253.
  17. Prieto, M.C.; Reverte, V.; Mamenko, M.; Kuczeriszka, M.; Veiras, L.C.; Rosales, C.B.; McLellan, M.; Gentile, O.; Jensen, V.B.; Ichihara, A.; et al. Collecting duct prorenin receptor knockout reduces renal function, increases sodium excretion, and mitigates renal responses in ANG II-induced hypertensive mice. Am. J. Physiol. Renal Physiol. 2017, 313, F1243–F1253.
  18. Mendoza-Torres, E.; Oyarzún, A.; Mondaca-Ruff, D.; Azocar, A.; Castro, P.F.; Jalil, J.E.; Chiong, M.; Lavandero, S.; Ocaranza, M.P. ACE2 and vasoactive peptides: Novel players in cardiovascular/renal remodeling and hypertension. Ther. Adv. Cardiovasc. Dis. 2015, 9, 217–237.
  19. Ni, J.; Yang, F.; Huang, X.R.; Meng, J.; Chen, J.; Bader, M.; Penninger, J.M.; Fung, E.; Yu, X.Q.; Lan, H.Y. Dual deficiency of angiotensin-converting enzyme-2 and Mas receptor enhances angiotensin II-induced hypertension and hypertensive nephropathy. J. Cell. Mol. Med. 2020, 24, 13093–13103.
  20. Ushiogi, Y.; Takabatake, T.; Häberle, D.A. Blood pressure and tubuloglomerular feedback mechanism in chronically salt-loaded spontaneously hypertensive rats. Kidney Int. 1991, 39, 1184–1192.
  21. Lu, Y.; Wei, J.; Stec, D.E.; Roman, R.J.; Ge, Y.; Cheng, L.; Liu, E.Y.; Zhang, J.; Hansen, P.B.L.; Fan, F.; et al. Macula Densa Nitric Oxide Synthase 1β Protects against Salt-Sensitive Hypertension. J. Am. Soc. Nephrol. 2016, 27, 2346–2356.
  22. Zhang, J.; Qu, L.; Wei, J.; Jiang, S.; Xu, L.; Wang, L.; Cheng, F.; Jiang, K.; Buggs, J.; Liu, R. A new mechanism for the sex differences in angiotensin II-induced hypertension: The role of macula densa NOS1beta-mediated tubuloglomerular feedback. Am. J. Physiol. Renal Physiol. 2020, 319, F908–F919.
  23. Hyndman, K.A.; Boesen, E.I.; Elmarakby, A.A.A.; Brands, M.W.; Huang, P.; Kohan, D.E.; Pollock, D.M.; Pollock, J.S. Renal collecting duct NOS1 maintains fluid-electrolyte homeostasis and blood pressure. Hypertension 2013, 62, 91–98.
  24. Gao, Y.; Stuart, D.; Takahishi, T.; Kohan, D.E. Nephron-Specific Disruption of Nitric Oxide Synthase 3 Causes Hypertension and Impaired Salt Excretion. J. Am. Heart Assoc. 2018, 7, e009236.
  25. Hyndman, K.A.; Bugaj, V.; Mironova, E.; Stockand, J.D.; Pollock, J.S. NOS1-dependent negative feedback regulation of the epithelial sodium channel in the collecting duct. Am. J. Physiol. Renal Physiol. 2015, 308, F244–F251.
  26. Raikwar, N.S.; Thomas, C.P. Nedd4-2 isoforms ubiquitinate individual epithelial sodium channel subunits and reduce surface expression and function of the epithelial sodium channel. Am. J. Physiol. Renal Physiol. 2008, 294, F1157–F1165.
  27. Ronzaud, C.; Loffing-Cueni, D.; Hausel, P.; Debonneville, A.; Malsure, S.R.; Fowler-Jaeger, N.; Boase, N.A.; Perrier, R.; Maillard, M.; Yang, B.; et al. Renal tubular NEDD4-2 deficiency causes NCC-mediated salt-dependent hypertension. J. Clin. Investig. 2013, 123, 657–665.
  28. Furusho, T.; Uchida, S.; Sohara, E. The WNK signaling pathway and salt-sensitive hypertension. Hypertens. Res. 2020, 43, 733–743.
  29. Sohara, E.; Uchida, S. Kelch-like 3/Cullin 3 ubiquitin ligase complex and WNK signaling in salt-sensitive hypertension and electrolyte disorder. Nephrol. Dial. Transplant. 2016, 31, 1417–1424.
  30. Liu, Z.; Xie, J.; Wu, T.; Truong, T.; Auchus, R.J.; Huang, C.-L. Downregulation of NCC and NKCC2 cotransporters by kidney-specific WNK1 revealed by gene disruption and transgenic mouse models. Hum. Mol. Genet. 2011, 20, 855–866.
  31. Mu, S.; Shimosawa, T.; Ogura, S.; Wang, H.; Uetake, Y.; Kawakami-Mori, F.; Marumo, T.; Yatomi, Y.; Geller, D.S.; Tanaka, H.; et al. Epigenetic modulation of the renal β-adrenergic–WNK4 pathway in salt-sensitive hypertension. Nat. Med. 2011, 17, 573–580.
  32. Pan, X.; Shao, Y.; Wu, F.; Wang, Y.; Xiong, R.; Zheng, J.; Tian, H.; Wang, B.; Wang, Y.; Zhang, Y.; et al. FGF21 Prevents Angiotensin II-Induced Hypertension and Vascular Dysfunction by Activation of ACE2/Angiotensin-(1-7) Axis in Mice. Cell Metab. 2018, 27, 1323–1337.e1325.
  33. Crislip, G.R.; Douma, L.G.; Masten, S.H.; Cheng, K.Y.; Lynch, I.J.; Johnston, J.G.; Barral, D.; Glasford, K.B.; Holzworth, M.R.; Verlander, J.W.; et al. Differences in renal BMAL1 contribution to Na+ homeostasis and blood pressure control in male and female mice. Am. J. Physiol. Renal Physiol. 2020, 318, F1463–F1477.
  34. Douma, L.G.; Costello, H.M.; Crislip, G.R.; Cheng, K.Y.; Lynch, I.J.; Juffre, A.; Barral, D.; Masten, S.; Roig, E.; Beguiristain, K.; et al. Kidney-specific KO of the circadian clock protein PER1 alters renal Na+ handling, aldosterone levels, and kidney/adrenal gene expression. Am. J. Physiol. Renal Physiol. 2022, 322, F449–F459.
  35. Hiramatsu, A.; Izumi, Y.; Eguchi, K.; Matsuo, N.; Deng, Q.; Inoue, H.; Nakayama, Y.; Nonoguchi, H.; Aramburu, J.; Lopez-Rodriguez, C.; et al. Salt-Sensitive Hypertension of the Renal Tubular Cell-Specific NFAT5 (Nuclear Factor of Activated T-Cells 5) Knockout Mice. Hypertension 2021, 78, 1335–1346.
  36. Ueda, K.; Nishimoto, M.; Hirohama, D.; Ayuzawa, N.; Kawarazaki, W.; Watanabe, A.; Shimosawa, T.; Loffing, J.; Zhang, M.Z.; Marumo, T.; et al. Renal Dysfunction Induced by Kidney-Specific Gene Deletion of Hsd11b2 as a Primary Cause of Salt-Dependent Hypertension. Hypertension 2017, 70, 111–118.
  37. Shao, S.; Li, X.D.; Lu, Y.Y.; Li, S.J.; Chen, X.H.; Zhou, H.D.; He, S.; Guo, Y.T.; Lu, X.; Gao, P.J.; et al. Renal Natriuretic Peptide Receptor-C Deficiency Attenuates NaCl Cotransporter Activity in Angiotensin II-Induced Hypertension. Hypertension 2021, 77, 868–881.
  38. Geurts, A.M.; Cost, G.J.; Freyvert, Y.; Zeitler, B.; Miller, J.C.; Choi, V.M.; Jenkins, S.S.; Wood, A.; Cui, X.; Meng, X.; et al. Knockout Rats via Embryo Microinjection of Zinc-Finger Nucleases. Science 2009, 325, 433.
  39. Szpirer, C. Rat models of human diseases and related phenotypes: A systematic inventory of the causative genes. J. Biomed. Sci. 2020, 27, 84.
  40. Sato, M.; Nakamura, S.; Inada, E.; Takabayashi, S. Recent Advances in the Production of Genome-Edited Rats. Int. J. Mol. Sci. 2022, 23, 2548.
  41. Nabika, T.; Ohara, H.; Kato, N.; Isomura, M. The stroke-prone spontaneously hypertensive rat: Still a useful model for post-GWAS genetic studies? Hypertens. Res. 2012, 35, 477–484.
  42. Schulz, A.; Kreutz, R. Mapping genetic determinants of kidney damage in rat models. Hypertens. Res. 2012, 35, 675–694.
  43. Yang, Y.; Kimura-Ohba, S.; Thompson, J.; Rosenberg, G.A. Rodent Models of Vascular Cognitive Impairment. Transl. Stroke Res. 2016, 7, 407–414.
  44. Doggrell, S.A.; Brown, L. Rat models of hypertension, cardiac hypertrophy and failure. Cardiovasc. Res. 1998, 39, 89–105.
  45. Liška, F.; Mancini, M.; Krupková, M.; Chylíková, B.; Křenová, D.; Šeda, O.; Šilhavý, J.; Mlejnek, P.; Landa, V.; Zídek, V.; et al. Plzf as a candidate gene predisposing the spontaneously hypertensive rat to hypertension, left ventricular hypertrophy, and interstitial fibrosis. Am. J. Hypertens. 2014, 27, 99–106.
  46. Liška, F.; Landa, V.; Zídek, V.; Mlejnek, P.; Šilhavý, J.; Šimáková, M.; Strnad, H.; Trnovská, J.; Škop, V.; Kazdová, L.; et al. Downregulation of Plzf Gene Ameliorates Metabolic and Cardiac Traits in the Spontaneously Hypertensive Rat. Hypertension 2017, 69, 1084–1091.
  47. Lin, Z.-H.; Fukuda, N.; Jin, X.-Q.; Yao, E.-H.; Ueno, T.; Endo, M.; Saito, S.; Matsumoto, K.; Mugishima, H. Complement 3 Is Involved in the Synthetic Phenotype and Exaggerated Growth of Vascular Smooth Muscle Cells From Spontaneously Hypertensive Rats. Hypertension 2004, 44, 42–47.
  48. Han, Y.; Fukuda, N.; Ueno, T.; Endo, M.; Ikeda, K.; Xueli, Z.; Matsumoto, T.; Soma, M.; Matsumoto, K. Role of Complement 3a in the Synthetic Phenotype and Angiotensin II-Production in Vascular Smooth Muscle Cells from Spontaneously Hypertensive Rats. Am. J. Hypertens. 2012, 25, 284–289.
  49. Ikeda, K.; Fukuda, N.; Ueno, T.; Endo, M.; Kobayashi, N.; Soma, M.; Matsumoto, K. Role of complement 3a in the growth of mesangial cells from stroke-prone spontaneously hypertensive rats. Clin. Exp. Hypertens. 2014, 36, 58–63.
  50. Negishi, E.; Fukuda, N.; Otsuki, T.; Katakawa, M.; Komatsu, K.; Chen, L.; Tanaka, S.; Kobayashi, H.; Hatanaka, Y.; Ueno, T.; et al. Involvement of complement 3 in the salt-sensitive hypertension by activation of renal renin-angiotensin system in spontaneously hypertensive rats. Am. J. Physiol.-Ren. Physiol. 2018, 315, F1747–F1758.
  51. Rubattu, S.; Volpe, M.; Kreutz, R.; Ganten, U.; Ganten, D.; Lindpaintner, K. Chromosomal mapping of quantitative trait loci contributing to stroke in a rat model of complex human disease. Nat. Genet. 1996, 13, 429–434.
  52. Rubattu, S.; Di Castro, S.; Schulz, H.; Geurts, A.M.; Cotugno, M.; Bianchi, F.; Maatz, H.; Hummel, O.; Falak, S.; Stanzione, R.; et al. Ndufc2 Gene Inhibition Is Associated with Mitochondrial Dysfunction and Increased Stroke Susceptibility in an Animal Model of Complex Human Disease. J. Am. Heart Assoc. 2016, 5, e002701.
  53. Mahal, Z.; Fujikawa, K.; Matsuo, H.; Zahid, H.M.; Koike, M.; Misumi, M.; Kaneko, T.; Mashimo, T.; Ohara, H.; Nabika, T. Effects of the Prdx2 depletion on blood pressure and life span in spontaneously hypertensive rats. Hypertens. Res. 2019, 42, 610–617.
  54. Kattoor, A.J.; Goel, A.; Mehta, J.L. LOX-1: Regulation, Signaling and Its Role in Atherosclerosis. Antioxidants 2019, 8, 218.
  55. Liang, Y.-Q.; Kakino, A.; Matsuzaka, Y.; Mashimo, T.; Isono, M.; Akamatsu, T.; Shimizu, H.; Tajima, M.; Kaneko, T.; Li, L.; et al. LOX-1 (Lectin-Like Oxidized Low-Density Lipoprotein Receptor-1) Deletion Has Protective Effects on Stroke in the Genetic Background of Stroke-Prone Spontaneously Hypertensive Rat. Stroke 2020, 51, 1835–1843.
  56. Xiao, B.; Harada, Y.; Kawakami, K.; Nabika, T. A 1.8-Mbp fragment on chromosome 1 affects sympathetic response to stress: Evaluation in reciprocal congenic strains between stroke-prone spontaneously hypertensive rat and Wistar-Kyoto rat. J. Hypertens. 2011, 29, 257–265.
  57. Ferdaus, M.Z.; Xiao, B.; Ohara, H.; Nemoto, K.; Harada, Y.; Saar, K.; Hübner, N.; Isomura, M.; Nabika, T. Identification of Stim1 as a candidate gene for exaggerated sympathetic response to stress in the stroke-prone spontaneously hypertensive rat. PLoS ONE 2014, 9, e95091.
  58. Odongoo, B.; Ohara, H.; Ngarashi, D.; Kaneko, T.; Kunihiro, Y.; Mashimo, T.; Nabika, T. Pathophysiological significance of Stim1 mutation in sympathetic response to stress and cardiovascular phenotypes in SHRSP/Izm: In vivo evaluation by creation of a novel gene knock-in rat using CRISPR/Cas9. Clin. Exp. Hypertens. 2021, 43, 34–41.
  59. Rapp, J.P.; Garrett, M.R. Will the real Dahl S rat please stand up? Am. J. Physiol. Renal Physiol. 2019, 317, F1231–F1240.
  60. Moreno, C.; Hoffman, M.; Stodola, T.J.; Didier, D.N.; Lazar, J.; Geurts, A.M.; North, P.E.; Jacob, H.J.; Greene, A.S. Creation and characterization of a renin knockout rat. Hypertension 2011, 57, 614–619.
  61. Mattson, D.L.; Lund, H.; Guo, C.; Rudemiller, N.; Geurts, A.M.; Jacob, H. Genetic mutation of recombination activating gene 1 in Dahl salt-sensitive rats attenuates hypertension and renal damage. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2013, 304, R407–R414.
  62. Zhou, X.; Zhang, Z.; Shin, M.K.; Horwitz, S.B.; Levorse, J.M.; Zhu, L.; Sharif-Rodriguez, W.; Streltsov, D.Y.; Dajee, M.; Hernandez, M.; et al. Heterozygous disruption of renal outer medullary potassium channel in rats is associated with reduced blood pressure. Hypertension 2013, 62, 288–294.
  63. Endres, B.T.; Priestley, J.R.; Palygin, O.; Flister, M.J.; Hoffman, M.J.; Weinberg, B.D.; Grzybowski, M.; Lombard, J.H.; Staruschenko, A.; Moreno, C.; et al. Mutation of Plekha7 attenuates salt-sensitive hypertension in the rat. Proc. Natl. Acad. Sci. USA 2014, 111, 12817–12822.
  64. Jin, C.; Sun, J.; Stilphen, C.A.; Smith, S.M.; Ocasio, H.; Bermingham, B.; Darji, S.; Guha, A.; Patel, R.; Geurts, A.M.; et al. HV1 acts as a sodium sensor and promotes superoxide production in medullary thick ascending limb of Dahl salt-sensitive rats. Hypertension 2014, 64, 541–550.
  65. Rudemiller, N.; Lund, H.; Jacob, H.J.; Geurts, A.A.M.; Mattson, D.L. CD247 modulates blood pressure by altering T-lymphocyte infiltration in the kidney. Hypertension 2014, 63, 559–564.
  66. Holditch, S.J.; Schreiber, C.A.; Nini, R.; Tonne, J.M.; Peng, K.-W.; Geurts, A.; Jacob, H.J.; Burnett, J.C.; Cataliotti, A.; Ikeda, Y. B-Type Natriuretic Peptide Deletion Leads to Progressive Hypertension, Associated Organ Damage, and Reduced Survival. Hypertension 2015, 66, 199–210.
  67. Kumarasamy, S.; Waghulde, H.; Gopalakrishnan, K.; Mell, B.; Morgan, E.; Joe, B. Mutation within the hinge region of the transcription factor Nr2f2 attenuates salt-sensitive hypertension. Nat. Commun. 2015, 6, 6252.
  68. Nayak, S.; Khan, M.A.; Wan, T.C.; Pei, H.; Linden, J.; Dwinell, M.R.; Geurts, A.M.; Imig, J.D.; Auchampach, J.A. Characterization of Dahl salt-sensitive rats with genetic disruption of the A2B adenosine receptor gene: Implications for A2B adenosine receptor signaling during hypertension. Purinergic Signal. 2015, 11, 519–531.
  69. Cowley, A.W., Jr.; Yang, C.; Zheleznova, N.N.; Staruschenko, A.; Kurth, T.; Rein, L.; Kumar, V.; Sadovnikov, K.; Dayton, A.; Hoffman, M.; et al. Evidence of the Importance of Nox4 in Production of Hypertension in Dahl Salt-Sensitive Rats. Hypertension 2016, 67, 440–450.
  70. Cheng, X.; Waghulde, H.; Mell, B.; Morgan, E.E.; Pruett-Miller, S.M.; Joe, B. Positional cloning of quantitative trait nucleotides for blood pressure and cardiac QT-interval by targeted CRISPR/Cas9 editing of a novel long non-coding RNA. PLoS Genet. 2017, 13, e1006961.
  71. Kumarasamy, S.; Waghulde, H.; Cheng, X.; Haller, S.T.; Mell, B.; Abhijith, B.; Ashraf, U.M.; Atari, E.; Joe, B. Targeted disruption of regulated endocrine-specific protein (Resp18) in Dahl SS/Mcw rats aggravates salt-induced hypertension and renal injury. Physiol. Genom. 2018, 50, 369–375.
  72. Waghulde, H.; Cheng, X.; Galla, S.; Mell, B.; Cai, J.; Pruett-Miller, S.M.; Vazquez, G.; Patterson, A.; Vijay Kumar, M.; Joe, B. Attenuation of Microbiotal Dysbiosis and Hypertension in a CRISPR/Cas9 Gene Ablation Rat Model of GPER1. Hypertension 2018, 72, 1125–1132.
  73. Abais-Battad, J.M.; Lund, H.; Dasinger, J.H.; Fehrenbach, D.J.; Cowley, A.W., Jr.; Mattson, D.L. NOX2-derived reactive oxygen species in immune cells exacerbates salt-sensitive hypertension. Free Radic. Biol. Med. 2020, 146, 333–339.
  74. Pulimeno, P.; Bauer, C.; Stutz, J.; Citi, S. PLEKHA7 is an adherens junction protein with a tissue distribution and subcellular localization distinct from ZO-1 and E-cadherin. PLoS ONE 2010, 5, e12207.
  75. Levy, D.; Ehret, G.B.; Rice, K.; Verwoert, G.C.; Launer, L.J.; Dehghan, A.; Glazer, N.L.; Morrison, A.C.; Johnson, A.D.; Aspelund, T.; et al. Genome-wide association study of blood pressure and hypertension. Nat. Genet. 2009, 41, 677–687.
  76. Hong, K.W.; Jin, H.S.; Lim, J.E.; Kim, S.; Go, M.J.; Oh, B. Recapitulation of two genomewide association studies on blood pressure and essential hypertension in the Korean population. J. Hum. Genet. 2010, 55, 336–341.
  77. Fox, E.R.; Young, J.H.; Li, Y.; Dreisbach, A.W.; Keating, B.J.; Musani, S.K.; Liu, K.; Morrison, A.C.; Ganesh, S.; Kutlar, A.; et al. Association of genetic variation with systolic and diastolic blood pressure among African Americans: The Candidate Gene Association Resource study. Hum. Mol. Genet. 2011, 20, 2273–2284.
  78. Ho, J.E.; Levy, D.; Rose, L.; Johnson, A.D.; Ridker, P.M.; Chasman, D.I. Discovery and replication of novel blood pressure genetic loci in the Women’s Genome Health Study. J. Hypertens. 2011, 29, 62–69.
  79. Lin, Y.; Lai, X.; Chen, B.; Xu, Y.; Huang, B.; Chen, Z.; Zhu, S.; Yao, J.; Jiang, Q.; Huang, H.; et al. Genetic variations in CYP17A1, CACNB2 and PLEKHA7 are associated with blood pressure and/or hypertension in She ethnic minority of China. Atherosclerosis 2011, 219, 709–714.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : ,
View Times: 298
Revisions: 3 times (View History)
Update Date: 01 Sep 2022
1000/1000