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 -- 2105 2023-06-19 02:56:07 |
2 format Meta information modification 2105 2023-06-19 04:04:16 |

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.
Park, C.H.; Tanaka, T.; Akimoto, Y.; Jeon, J.P.; Yokozawa, T. Rokumijiogan, Hachimijiogan and Bakumijiogan in Nephrectomized Rats. Encyclopedia. Available online: https://encyclopedia.pub/entry/45764 (accessed on 07 May 2024).
Park CH, Tanaka T, Akimoto Y, Jeon JP, Yokozawa T. Rokumijiogan, Hachimijiogan and Bakumijiogan in Nephrectomized Rats. Encyclopedia. Available at: https://encyclopedia.pub/entry/45764. Accessed May 07, 2024.
Park, Chan Hum, Takashi Tanaka, Yoshie Akimoto, Jin Pyeong Jeon, Takako Yokozawa. "Rokumijiogan, Hachimijiogan and Bakumijiogan in Nephrectomized Rats" Encyclopedia, https://encyclopedia.pub/entry/45764 (accessed May 07, 2024).
Park, C.H., Tanaka, T., Akimoto, Y., Jeon, J.P., & Yokozawa, T. (2023, June 19). Rokumijiogan, Hachimijiogan and Bakumijiogan in Nephrectomized Rats. In Encyclopedia. https://encyclopedia.pub/entry/45764
Park, Chan Hum, et al. "Rokumijiogan, Hachimijiogan and Bakumijiogan in Nephrectomized Rats." Encyclopedia. Web. 19 June, 2023.
Rokumijiogan, Hachimijiogan and Bakumijiogan in Nephrectomized Rats
Edit

According to the underlying concepts of TCM(traditional Chinese medicine) theory, chronic kidney disease (CKD) is commonly regarded as caused by Qi deficiency and Yang deficiency due to Xieqi repletion. The basic treatment principle of TCM is Yang/Qi-replenishing and Xieqi-draining. 

nephrectomy Rokumijiogan Hachimijiogan Bakumijiogan

1. Introduction

Chronic kidney disease (CKD) is one of the major public health problems worldwide. Over the past 20 years, the number of deaths from CKD has increased by 82.3%, which is approximately the third largest increase among the 25 leading causes of death [1]. Treatment of stages 1 to 4 of patients with CKD focus on slowing kidney damage and preventing or treating complications and comorbidities through medication or diet. In the final stage (stage 5 or end-stage renal disease) of CKD, renal replacement therapy such as dialysis or kidney transplantation is needed to sustain life [2]. The prognosis of CKD patients has improved with the development of kidney dialysis and medical management systems. However, maintaining continuous dialysis is a clear burden on patients in terms of both physical and mental parts, and social issues including financial burdens have arisen due to the overall number of patients receiving dialysis. Several conservative treatments for CKD may be applied in this situation.
Herbal medicines, including traditional medicines using natural products, have attracted a lot of attention due to their significant and varied pharmacologic actions and bioactive phytoconstituents without side effects or toxicity [3][4]. Medicinal plants, also called medicinal herbs, have been used against various diseases for thousands of years around the world. For example, medicinal plant species with antioxidant properties including phenolics, carotenoids, and vitamins are widely engaged as traditional medicine and functional food and help prevent and/or treat diseases caused from an overproduction of reactive oxygen species (ROS) such as cancer, neurodegenerative diseases, and cardiovascular disease [5]. According to the viewpoint of traditional Chinese medicine (TCM), CKD is commonly considered a Qi (vital energy) deficiency and Yang (warms and activates the body) shortage, that is, exhausted vigor caused by Xieqi (evil influence) repleteness. The basis of clinical management is to replenish Yangqi and drain Xieqi [6]. Herbal drug compositions containing Rokumijiogan (RJG) are used for renal insufficiency in traditional East Asian medicine. It is one of the representative improving body function medications and is widely used to treat frequent urination, micturition disorder, and edema, resulting from nephrosis and chronic nephritis [4][7][8].

2. HJG and BJG Extracts

HJG is comprised of the following (the values after the name indicate the proportion of each ingredient expressed in parts per whole): Radix Rehmanniae 6.0, Fructus Corni 3.0, Rhizoma Dioscoreae 3.0, Rhizoma Alismatis 3.0, Poria 3.0, Cortex Moutan 2.5, Cortex Cinnamomi 1.0, and Radix Aconiti 0.5. These eight crude drugs were boiled gently in 10 times their volume of water for 60 min, filtered, and the powder was spray-dried to obtain an extract at a yield of about 10% by weight of the original preparation. The composition of BJG was: Radix Rehmanniae 0.750, Fructus Corni 0.370, Rhizoma Dioscoreae 0.370, Rhizoma Alismatis 0.280, Poria 0.280, Cortex Moutan 0.280, Fructus Schisandrae 0.199, and Radix Ophiopogonis 0.280. The BJG powder was prepared with a yield of about 12% by weight of the crude drug. To analyze the components, HJG and BJG were separately pulverized and a portion (0.5 g) was extracted by vortexing at room temperature for 8 h with 5 mL of MeOH and then sonicated for 20 min. After filtration through a 0.45 mm membrane filter, an aliquot (10 μL) of the sample solution was injected into the high-performance liquid chromatography (HPLC) column. Chromatographic separations were performed on a Cosmosil 5C18-AR II (Nacalai Tesque, Kyoto, Japan) reverse-phase column (250 × 4.6 mm i.d.), using 50 mM H3PO4 (A) and CH3CN (B) as the mobile phase at 4 to 30% (39 min) and 30 to 75% (15 min). The column temperature was 40 °C and flow rate was 0.8 mL/min. The separated compounds were detected by JASCO MD-2010 HPLC diode array detectors, identified, and quantified on the basis of chromatographic retention times of coeluted pure standards using Class LC-10 Version 1.62 software (Shimadzu, Kyoto, Japan). Eleven compounds (GS, gallic acid, morroniside, loganin, paeoniflorin, PG, cornuside, cinamic acid, 6′-O-benzoyl paeoniflorin (BP), cinamdalehyde, and paeonol) of HJG extract were detected in the HPLC chromatogram. In the elution profile of BJG extract, GS, gallic acid, morroniside, loganin, paeoniflorin, PG, cornuside, BP, paeonol, schizandrin, gomisin A, and schizandrin were identified [9].

3. Renal Histological Findings and Renal Functional Parameters

Prolonged proteinuria due to subtotal nephrectomy causes irreversible structural changes in nephrons and renal dysfunction, including glomerular-capillary hypertension, proximal tubule damage, stimulation of nuclear signals such as nuclear factor-kappa B (NF-κB), and NF-κB-mediated inflammatory events in the interstitium, interstitial inflammatory responses, and consequently, fibrosis [10][11]. Moreover, an enlarged glomerular function resulting from nephrectomy is elicited by high glomerular filtration, and glomerular enlargement and destruction, eventually bringing about glomerulosclerosis [12]. These observations indicate that renal ablation induced the early stage of CKD with renal morphologic changes, such as accelerated mesangial matrix production, mesangial cell proliferation, and glomerular endothelial cell injury, as well as tubulointerstitial damage. In the histopathological study, clear histological types indicating glomerular sclerosis, tubulointerstitial damage, and arteriolar sclerotic lesion were discovered after nephrectomy, whereas glomerulo and tubulointerstitial damage was ameliorated in 5/6 nephrectomized (5/6Nx) rats that were orally administered HJG and BJG. Arteriolar sclerotic lesions were also improved [9]. Although improvement in activity on the progression of kidney lesions was more marked in the HJG-treated group than in the BJG-administered group, the two types of Chinese medicine prescriptions showed improvement in renal pathological lesions by nephrectomy. Uremic toxins maintained due to renal dysfunction by direct or secondary effects on renal tissue aggravate renal function and tissue, generating a vicious cycle that results in end-stage renal disease [13]. Consistent with this, rats given HJG and BJG showed improvements in biochemical parameters including renal profiles (urinary protein, blood urea nitrogen, serum creatinine, and creatinine clearance) [9].

4. Biomarkers Associated with Oxidative Stress in the Kidney

In experiments using unilaterally 5/6Nx rats, decreased antioxidative activity and increased oxidative stress in the remaining kidney were observed by Harris et al. [14], Ozbek [15], Tamay-Cach et al. [16], and Ling and Kuo [17]. Enhanced ROS production leads to various forms of tissue damage and functional disabilities by attacking, denaturing, and changing structural and functional molecules and by activating redox-responsive transcriptional factors and the signal transduction pathway. These phenomena sequentially increase necrotic and apoptotic cell death, inflammatory-driven fibrosis, and other injuries. In the kidney, oxidative stress is a persistent hallmark of progressive renal disease, and it plays an important role in progressive worsening of the renal function and various structures, as well as numerous other complications [18].
Renal ROS levels in vehicle-treated 5/6Nx rats were significantly higher than those of sham control rats, but in the Nx-BJG group, the ROS levels showed that a clear sign of recovery almost sham control levels after administration of BJG for 10 weeks. However, there were no differences between the vehicle-treated 5/6Nx and HJG-treated 5/6Nx groups. Moreover, levels of renal thiobarbituric acid-reactive substances (TBARS) were 0.52 nmol/mg protein in the sham control group, whereas it was significantly increased in the 5/6Nx-Veh group (1.19 nmol/mg protein). These levels of renal TBARS were markedly decreased in BJG-treated 5/6Nx rats. Conversely, the activities of the renal antioxidant enzymes (superoxide dismutase, reduced glutathione/oxidized glutathione ratio) in the renal tissue in 5/6Nx-Veh rats were significantly decreased compared with sham control rats (p < 0.001), whereas both HJG- and BJG-treated groups showed the effect of increasing the activity of antioxidant enzymes compared to those of the 5/6Nx-Veh group. Unlike the consequence of oxidative metabolism, the activities of antioxidant enzymes showed greater in the 5/6Nx-HJG groups than in the 5/6Nx-BJG groups [19]. These results suggest that HJG or BJG administration can be considered as having different mechanisms.

5. Renal NADPH Oxidase-4 (Nox-4), p22phox, Nrf2, and Heme Oxygenase-1 (HO-1) Protein Expressions

Although the causes of increased ROS generation in kidney disease are very diverse, the studies have focused on the fact that NADPH oxidase mainly contributes to the process of ROS generation [20]. Structurally, NADPH oxidase includes a membrane-associated cytochrome b558 consisting of one p22phox (phox is an acronym for phagocyte oxidase), one gp91phox subunit, and at least four cytosolic components (p47phox, p67phox, p40phox, and the small GTPase rac1 or rac2) [21]. Notably, among them, Nox-4 and p22phox have been identified as major sources of ROS production in tissues and cells and could play roles in some pathological conditions [22][23][24].
In addition, oxidative stress provokes variations in the Nrf2 complex and the upregulated transcription of Nrf2-dependent genes such as HO-1 [25]. Nrf2 is a redox-sensitive transcription factor involved in protecting cells from oxidative stress-related damage [26][27]. NADPH oxidase-derived ROS and ROS-mediated mitogen-activated protein kinases (MAPKs) may arbitrate the induction of antioxidant HO-1 and its transcription factor, Nrf2 [28][29]. Therefore, regulating the Nrf2/HO-1 antioxidant signaling pathway may be a promising target of the pathology of oxidative stress. In results, the 5/6Nx vehicle-treated group showed reduced expressions of the Nrf2/HO-1 antioxidant signaling pathway in the remnant renal tissue compared to those of sham control rats. Otherwise, administration of BJG effectively relieved oxidative stress and upregulated Nrf2 and HO-1 [9]. Activation of intracellular signaling cascades, including NADPH oxidase-derived ROS and, consequently, induced MAPKs may arbitrate the stimulation of Nrf2/HO-1 expression.

6. Renal c-Jun N-Terminal Kinase (JNK), Phosphor (p)-JNK, c-Jun, and Transforming Growth Factor-β1 (TGF-β1) Protein Expressions

JNK activation is triggered by proinflammatory cytokines and ROS. JNK has been suggested to play a critical role in signaling events via the activation of activator protein-1 transcription factor, phosphorylation of c-Jun, and induction of proinflammatory molecules expression such as intercellular adhesion molecule-1 (ICAM-1). ROS generation is also induced by JNK activation [30]. JNK activation can be modulated via stabilization/gene expression through its phosphorylation, p-JNK [31]. Only nuclear-translocated p-JNK induces c-Jun upregulation [32]. TGF-β-induced fibronectin expression requires JNK activation, which, in turn, regulates c-Jun activation [33].

7. Renal NF-κB, Cyclooxygenase-2 (COX-2), Inducible Nitric Oxide Synthase (iNOS), Monocyte Chemotactic Protein-1 (MCP-1), and ICAM-1 Protein Expressions

NF-κB signaling is a central pathway of inflammatory stimuli involved in the modulation of transcription and multiple inflammation-related mediator expressions such as COX-2 and iNOS [34]. The severe and progressive renal damage in the 5/6Nx model was associated with increased NF-κB activity, whereas the amelioration of proteinuria and renal structural damage was associated with the inhibition of NF-κB system activity. Increased oxidative stress induces the expression of MCP-1 and ICAM-1 genes. Macrophage infiltration regulated by chemokines such as MCP-1, and adhesion molecules such as ICAM-1, has been reported to be related with deposition of renal immune complex and elevated production of renal chemokine [35][36]. As shown in the study published by Park et al. [9][19], it showed that renal NF-κBp65 and its associated inflammation-related mediator, chemokine, and adhesion molecule expressions were markedly increased in vehicle-treated 5/6Nx rats compared with sham-operated rats. In contrast, the BJG-treated group showed significant suppression of the expression of NF-κBp65 and NF-κB-mediated target proteins. ICAM-1 protein expression was also reduced by BJG administration. The administration of HJG led to significant down-regulation of NF-κBp65 and COX-2 expression, whereas it slightly altered iNOS and MCP-1 levels.

8. Protective Role of RJG-Containing Components against Free Radical-Induced Oxidative Stress in Renal Tubular Epithelial Cells

Reactive oxygen and free radicals have various effects on the progression and development of renal disease. The results of several studies have provided evidence that renal tubular obstruction is involved in renal damage, and it is well known that renal tubular cells, including LLC-PK1 renal proximal tubule cells, are susceptible to free radical damage [37][38]. Therefore, an in vitro model of oxidative damage, in which LLC-PK1 cells are exposed to free radicals, would be useful in the search for compounds that can provide effective protection.
As mentioned above [9], GS, gallic acid, morroniside, loganin, paeoniflorin, PG, cornuside, BS, and paeonol were identified as major components of HJG and BJG extracts. In addition, schizandrin was detected in the BJG extract.
Although various kinds of free radical initiators are known, 3-morpholinosydnonimine (SIN-1) has been used successfully as a peroxynitrite initiator, since it generates free radicals at a measurable and constant rate via thermal decomposition without biotransformation. To elucidate the protective effect against SIN-1 in cells, researchers measured cell viability and demonstrated that the exposure of renal tubular LLC-PK1 cells to SIN-1 resulted in decreased cell viability. Among the 10 components tested, the most potent activity was exhibited by GS, which showed the highest viability (89.4% at 50 μg/mL) and protected against cell damage induced by SIN-1 in time- and dose-dependent manners. PG, paeonol, and cornuside also had inhibitory effects, suggesting that the compounds originating from Corni Fructus and Moutan Cortex protected LLC-PK1 cells from SIN-1-induced cytotoxicity. Schizandrin, originating from Schisandrae Fructus and contained in BJG, presented similarly strong viability of 67.4% at 50 μg/mL. These findings suggest that Corni Fructus- and Moutan Cortex-originating compositions might act as free radical scavengers and afford important protection against oxidative stress induced by SIN-1.

References

  1. Radhakrishnan, J.; Remuzzim, G.; Saran, R.; Williams, D.E.; Rios-Burrows, N.; Powe, N.; Brück, K.; Wanner, C.; Stel, V.; Venuthurupalli, S.; et al. Taming the chronic kidney disease epidemic: A global view of surveillance efforts. Kidney Int. 2014, 86, 246–250.
  2. Goh, Z.S.; Griva, K. Anxiety and depression in patients with end-stage renal disease: Impact and management challenges—A narrative review. Int. J. Nephrol. Renovasc. Dis. 2018, 11, 93–102.
  3. Astin, J.A.; Marie, A.; Pelletier, K.R.; Hansen, E.; Haskell, W.L. A review of the incorporation of complementary and alternative medicine by mainstream physicians. Arch. Intern. Med. 1998, 158, 2303–2310.
  4. Ha, H.; Lee, J.K.; Lee, H.Y.; Koh, W.S.; Seo, C.S.; Lee, M.Y.; Huang, D.S.; Shin, H. Safety evaluation of Yukmijihwang-tang: Assessment of acute and subchronic toxicity in rats. Evid. Based Complement. Alternat. Med. 2011, 2011, 672136.
  5. Singla, R.K.; De, R.; Efferth, T.; Mezzetti, B.; Sahab Uddin, M.; Sanusi; Ntie-Kang, F.; Wang, D.; Schultz, F.; Kharat, K.R.; et al. The International Natural Product Sciences Taskforce (INPST) and the power of Twitter networking exemplified through # INPST hashtag analysis. Phytomed. Int. J. Phytother. Phytopharm. 2023, 108, 154520.
  6. Lu, Z.; Zhong, Y.; Liu, W.; Xiang, L.; Deng, Y. The efficacy and mechanism of Chinese herbal medicine on diabetic kidney disease. J. Diabetes Res. 2019, 2019, 2697672.
  7. Kang, D.G.; Sohn, E.J.; Moon, M.K.; Mun, Y.J.; Woo, W.H.; Kim, M.K.; Lee, H.S. Yukmijihwang-tang ameliorates ischemia/reperfusion-induced renal injury in rats. J. Ethnopharmacol. 2006, 104, 47–53.
  8. Lee, J.K.; Lee, N.H.; Ha, H.K.; Lee, H.Y.; Jung, D.Y.; Choi, J.Y.; Seo, C.S.; Shin, H.K. Analysis of studies on Yukmijihwang-tang for establishment of evidence based medicine. Korean J. Orient. Physiol. Pathol. 2009, 23, 15–26.
  9. Park, C.H.; Lee, S.L.; Noh, J.S.; Yokozawa, T. Rokumi-jio-gan-containing prescriptions attenuate oxidative stress, inflammation, and apoptosis in the remnant kidney. Evid. Based Complement. Alternat. Med. 2012, 2012, 587902.
  10. Remuzzi, G.; Ruggenenti, P.; Benigni, A. Understanding the nature of renal disease progression. Kidney Int. 1997, 51, 2–15.
  11. Remuzzi, G.; Bertani, T. Pathophysiology of progressive nephropathies. N. Engl. J. Med. 1998, 339, 1448–1456.
  12. Brenner, B.M.; Meyer, T.W.; Hostetter, T.H. Dietary protein intake and the progressive nature of kidney disease: The role of hemodynamically mediated glomerular injury in the pathogenesis of progressive glomerular sclerosis in aging, renal ablation, and intrinsic renal disease. N. Engl. J. Med. 1982, 307, 652–659.
  13. Sakai, T.; Kumano, K.; Takara, S.; Kuwao, S.; Ise, M.; Sugano, M.; Uehara, Y. Effect of oral-adsorbent (AST-120) to chronic renal failure (CRF) in rats. Jpn. J. Nephrol. 1989, 31, 359–365.
  14. Harris, D.C.; Chan, L.; Schrier, R.W. Remnant kidney hypermetabolism and progression of chronic renal failure. Am. J. Physiol. Ren. Physiol. 1988, 254, F267–F276.
  15. Ozbek, E. Induction of oxidative stress in kidney. Int. J. Nephrol. 2012, 2012, 465897.
  16. Tamay-Cach, F.; Quintana-Pérez, J.C.; Trujillo-Ferrara, J.G.; Cuevas-Hernández, R.I.; Del Valle-Mondragón, L.; García-Trejo, E.M.; Arellano-Mendoza, M.G. A review of the impact of oxidative stress and some antioxidant therapies on renal damage. Ren. Fail. 2016, 38, 171–175.
  17. Ling, X.C.; Kuo, K.L. Oxidative stress in chronic kidney disease. Ren. Replace Ther. 2018, 4, 53.
  18. Kim, H.J.; Vaziri, N.D. Contribution of impaired Nrf2-Keap1 pathway to oxidative stress and inflammation in chronic renal failure. Am. J. Physiol. Ren. Physiol. 2010, 298, F662–F671.
  19. Park, C.H.; Lee, S.; Okamoto, T.; Tanaka, T.; Yokozawa, T. Rokumi-jio-gan-containing prescriptions regulate oxidative stress through improving dyslipidemia in a subtotal nephrectomized rat model. J. Ethnopharmacol. 2013, 148, 449–458.
  20. Bedard, K.; Krause, K.H. The NOX family of ROS-generating NADPH oxidases: Physiology and pathophysiology. Physiol. Rev. 2007, 87, 245–313.
  21. Babior, B.M.; Lambeth, J.D.; Nauseef, W. The neutrophil NADPH oxidase. Arch. Biochem. Biophys. 2002, 397, 342–344.
  22. Etoh, T.; Inoguchi, T.; Kakimoto, M.; Sonoda, N.; Kobayashi, K.; Kuroda, J.; Sumimoto, H.; Nawata, H. Increased expression of NAD(P)H oxidase subunits, NOX4 and p22phox, in the kidney of streptozotocin-induced diabetic rats and its reversibity by interventive insulin treatment. Diabetologia 2003, 46, 1428–1437.
  23. Kawahara, T.; Ritsick, D.; Cheng, G.; Lambeth, J.D. Point mutation in the proline-rich region of p22phox are dominant inhibitors of Nox1- and Nox2-dependent reactive oxygen generation. J. Biol. Chem. 2005, 280, 31859–31869.
  24. McKallip, R.J.; Jia, W.; Schlomer, J.; Warren, J.W.; Nagarkatti, P.S.; Nagarkatti, M. Cannabidiol-induced apoptosis in human leukemia cells: A novel role of cannabidiol in the regulation of p22phox and Nox4 expression. Mol. Pharmacol. 2006, 70, 897–908.
  25. Tong, K.I.; Katoh, Y.; Kusunoki, H.; Itoh, K.; Tanaka, T.; Yamamoto, M. Keap1 recruits Neh2 through binding to ETGE and DLG motifs: Characterization of the two-site molecular recognition model. Mol. Cell. Biol. 2006, 26, 2887–2900.
  26. Harvey, C.J.; Thimmulappa, R.K.; Singh, A.; Blake, D.J.; Ling, G.; Wakabayashi, N.; Fujii, J.; Myers, A.; Biswal, S. Nrf2-regulated glutathione recycling independent of biosynthesis is critical for cell survival during oxidative stress. Free Radic. Biol. Med. 2009, 46, 443–453.
  27. Li, W.; Kong, A.N. Molecular mechanisms of Nrf2-mediated antioxidant response. Mol. Carcinog. 2009, 48, 91–104.
  28. Anwar, A.A.; Li, F.Y.L.; Leake, D.S.; Ishii, T.; Mann, G.E.; Siow, R.C.M. Induction of heme oxygenase 1 by moderately oxidized low-density lipoproteins in human vascular smooth muscle cells: Role of mitogen-activated protein kinases and Nrf2. Free Radic. Biol. Med. 2005, 39, 227–236.
  29. He, M.; Siow, R.C.M.; Sugden, D.; Gao, L.; Cheng, X.; Mann, G.E. Induction of HO-1 and redox signaling in endothelial cells by advanced glycation end products: A role for Nrf2 in vascular protection in diabetes. Nutr. Metab. Cardiovasc. Dis. 2011, 21, 277–285.
  30. Hojo, Y.; Saito, Y.; Tanimoto, T.; Hoefen, R.J.; Baines, C.P.; Yamamoto, K.; Haendeler, J.; Asmis, R.; Berk, B.C. Fluid shear stress attenuates hydrogen peroxide-induced c-Jun NH2-terminal kinase activation via a glutathione reductase-mediated mechanism. Circ. Res. 2002, 91, 712–718.
  31. Kallunki, T.; Deng, T.; Hibi, M.; Karin, M. c-Jun can recruit JNK to phosphorylate dimerization partners via specific docking interactions. Cell 1996, 87, 929–939.
  32. Gupta, S.; Campbell, D.; Dériijard, B.; Davis, R.J. Transcription factor ATF2 regulation by the JNK signal transduction pathway. Science 1995, 267, 389–393.
  33. Hocevar, B.A.; Brown, T.L.; Howe, P.H. TGF-β induces fibronectin synthesis through a c-Jun N-terminal kinase-dependent, Smad4-independent pathway. EMBO J. 1999, 18, 1345–1356.
  34. Surh, Y.J.; Chun, K.S.; Cha, H.H.; Han, S.S.; Keum, Y.S.; Park, K.K.; Lee, S.S. Molecular mechanisms underlying chemopreventive activities of anti-inflammatory phytochemicals: Down-regulation of COX-2 and iNOS through suppression of NF-kappa B activation. Mutat. Res. 2001, 480–481, 243–268.
  35. Chow, F.; Ozols, E.; Nikolic-Paterson, D.J.; Atkins, R.C.; Tesch, G.H. Macrophages in mouse type 2 diabetic nephropathy: Correlation with diabetic state and progressive renal injury. Kidney Int. 2004, 65, 116–128.
  36. Kosugi, T.; Nakayama, T.; Heinig, M.; Zhang, L.; Yuzawa, Y.; Sanchez-Lozada, L.G.; Roncal, C.; Johnson, R.J.; Nakagawa, T. Effect of lowering uric acid on renal disease in the type 2 diabetic db/db mice. Am. J. Physiol. Ren. Physiol. 2009, 297, F481–F488.
  37. Gobé, G.; Willgoss, D.; Hogg, N.; Schoch, E.; Endre, Z. Cell survival or death in renal tubular epithelium after ischemia-reperfusion injury. Kidney Int. 1999, 56, 1299–1304.
  38. Schena, F.P.; Grandaliano, G.; Gesualdo, L. The role of tubular cells in the progression of renal damage: Guilty or innocent? Ren. Fail. 2001, 23, 589–596.
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: 296
Revisions: 2 times (View History)
Update Date: 19 Jun 2023
1000/1000