Review Open Access
Copyright ©The Author(s) 2001. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Oct 15, 2001; 7(5): 627-629
Published online Oct 15, 2001. doi: 10.3748/wjg.v7.i5.627
Pharmacology of tetrandrine and its therapeutic use in digestive diseases
Ding-Guo Li, Zhi-Rong Wang, Han-Ming Lu, Department of Gastroenterology, Xinhua Hospital, Shanghai Second Medical University, Shanghai 200092, China
Author contributions: All authors contributed equally to the work.
Correspondence to: Dr. Ding Guo Li, Department of Gastroenterology, Xinhua Hospital, Shanghai Second Medical University, Shanghai 200092, China
Received: May 12, 2001
Revised: May 14, 2001
Accepted: May 16, 2001
Published online: October 15, 2001

Abstract
Key Words: Tetrandrine/pharmacology, digestive system diseases/drug therapy



INTRODUCTION

Tetrandrine (Tet) is a dibenzylisoquinoline alkaloid isolated from Stephania tetrandra S. Moore, a Chinese herbal medicine. In the past decade, lots of studies demonstrated that Tet has multiple bioactivities. It is promising to use Tet as an antifibrogenetic in liver or lung fibrosis with or without portal or pulmonary hypertension, as well as an immunomodulating and anticarcinoma drug.

PHARMACOLOGY
Ca2+ channel blocking activity

Abnormal Ca2+ signaling and elevated concentration of intracellular free Ca2+ are the basic pathophysiological events involved in various diseases. As a Ca2+ antagonist, Tet can inhibit extracellular Ca2+ entry, intervene in the distribution of intracellular Ca2+, maintain intracellular Ca2+ homeostasis, and then disrupt the pathological processes. As shown in whole cell patch-clamp recordings, Tet blocked bovine chromaffin cells voltage-operated Ca2+ channel current in a time- and concentration-dependently manner. In rat phaeochromocytoma PC 12 cells, 100 μmol·L¯¹ Tet abolished high K+ (30 mmol•L¯¹) -induced sustained increase in cytoplasmic Ca2+ concentration, inhibit bombesin-induced inositol triphosphate accumulation in NIH/3T3 fibroblast and abolish Ca2+ entry[1]. In rat glioma C6 cells, studied with fluorometric ratio method, Tet did not affect the resting cytoplasmic Ca2+ concentration, but it inhibited IP3 accumulation and the sustained and peak elevation of cytoplasmic Ca2+ concentration induced by bombesin and thapsigargin, a microsomal Ca2+-ATPase inhibitor, in a dose-dependent manner. The dose of Tet needed to abolish the sustained and peak elevation of cytoplasmic Ca2+ concentration induced by bombesin and thapsigargin was 30 μmol•L¯¹[2]. Bickmeyer et al[3] demonstrated that NG108-15 cells treated with 100 μM Tet for seven minutes could block voltage-dependent Ca2+ entry induced by depolarization with 50 mM KCl. Tet could block non-voltage-operated Ca2+ entry activated by intracellular Ca2+ store depletion induced by thapsigargin and could release intracellular Ca2+ in HL-60 cells, and could therefore increase concentration of intracellular free Ca2+, elicit therapeutic effects. We have previ ously demonstrated that Tet could concentration-dependently block extracellular Ca2+ entry into hepatocytes, promote mitochondria Ca2+-uptake, and inhibit Ca2+-mobilizing from mitochondria. However, the blockade of Ca2+ channel played the most important role in maintaining Ca2+ homeostasis, but not intracellular Ca2+ distribution.

In the presence of extracellular Ca2+ (1.3 μmol•L¯¹), glutamate, serotonin and histamine significantly increased the intracellular Ca2+ concentration in a dose-dependent manner. Thirty μmol•L¯¹ Tet significantly inhibited the increase in intracellular Ca2+ concentration induced by glutamate, serotonin and histamine by 28.0%, 46.8% and 29.0%. In Ca2+ free Hanks’ solution, Tet did not produce a significant inhibitory effect on the increase in intracellular Ca2+ concentration caused by serotonin and histamine. These results indicated that Tet conducted blocking of Ca2+ influx from the extracellular site via NMDA, 5-HT2 and histamine type I receptor-operated Ca2+ channels and has no obvious effect on the Ca2+ release from intracellular Ca2+ stores[4]. In addition, Tet also inhibited extracellular Ca2+ entry and intracellular Ca2+ mobilization induced by norepinhpr ine and angiotonin II via corresponding receptor respectively[3,5]. Taking together, in different tissues and different kinds of cells, Tet may block the Ca2+ channel through different mechanisms. It can block the voltage- and/or receptor-operated Ca2+ channel. Nevertheless, in some kind of carcinoma cells, Tet does not affect Ca2+ channel, but promote Ca2+ release from intracellular stores and elevate the cytosolic free Ca2+ concentration.

Immunomodulating activity

Clinically, Stephania tetrandra S. Moore has been thought to be effective in treating autoimmune diseases such as rheumatoid arthritis and systemic lupus erythe matosus. Tet, the active ingredient isolated from Stephania tetrandra S. Moore, has potential immunomodul ating and anti-inflammatory effects. T-lymphocyte splay a critical role as autoactive and pathogenic population in autoimmune and inflammatory diseases. Some experimental data showed that, through down-regulating the protein kinase C (PKC) signaling, interleukin-2 secretion and the expression of the T cell activation antigen (CD71), Tet inhibited phobol 12-myristate 13-acetate (PMA)+ionomycin-induced T cell proliferation dependent on interleukin-2 receptor alpha chain and CD69, such an action was unrelated to Ca2+ channel blockade[6]. Tet (0.1-10 μmol•L¯¹) significantly inhibited neutrophil-monocyte chemotactic factor-1 upregulation and adhesion to fibrinogen induced by N-formyl-methionyl-leucyl-phenylalanim ne and PMA. Tet at 0.1-100 μmol•L¯¹ caused dose- and time-dependent loss of cell viability of mouse peritoneal macrophages, guinea-pig alveolar macrophages and mouse macrophage-like J774 cells, reduced production of oxyg en free radical oxygen, down-regulated synthesis and release of some pro-inflammatory cytokines[7,8].

Nuclear transcription factor kappa B (NF-kappa B) is a multiprotein complex which regulates a variety of genes concerned with immunity and inflammation. For the alveolar macrophages, Tet could inhibit the activation of their NF-kappa B and NF-kappa B-dependent reporter gene expression induced by endotoxin, PMA, and silica in a dose-dependent manner. Western blot analysis suggested that the inhibitory effects of tetrandrine on NF-kappa B activation could be attributed to its ability to suppress signal-induced degradation of I kappa B alpha, a cytoplasmic inhibitor of the NF-kappa B transcription factor[7,9].

Conducting tumor cell apoptosis

To induce tumor cell apoptosis is one of the important chemo-therapeutic strategies for malignent tumors. Tet inhibited both proliferation and clonogenicity of human leukemic U937 cells at an optimal concentration of 2.5 mg•L¯¹. The characteristic morphological changes of apoptosis were observed under light microscopy and DNA fragmentation was noted by gel electrophoresis in these cells. Moreover, flow-cytometric detection of surface phosphatidyl serine expression of cells after treatment with Tet confirmed the induction of apoptosis in these cells[10]. Tet concentration-dependently inhibited the proliferation of human leukemic HL-60 cells. Morphological observation and DNA analysis revealed that Tet caused cell shrinkage with the formation of apoptotic bodies, and showed clear evidence of DNA fragmentation[11]. Tet was found to induce pronounced morphological changes characteristic of apopt osis and extensive DNA fragmentation in the human BM13674 cell line 8 h after treatment[12]. The induction of apoptosis by Tet was much more rapid in CEM-C7 cells (4 h) than in the same cells treated with glucocort icoids (40 h), and did not require de novo protein synthesis[13]. These results indicate that Tet may have value as an anti-neoplastic agent.

Reversing multidrug resistance (MDR)

The occurrence of MDR to chemotherapeutic drugs is a major problem for successful cancer treatment. The overexpression of cell membrane P-glycoprotein (P-gp) is one of the major mechanisms of MDR. P-gp pumps antitumor drugs out of tumor cells, causing drug resistance.

Tet (3 μmol•L¯¹) reduced the paclitaxel concentration required to achieve 50% inhibition of cell growth (EC50) of HCT15 (P-gp-positive) cells about 3100-fold, and also reduced the EC50 value of actinomycin D about 36.0-fold in the cells. Meanwhile, Tet had no effect on the cytotoxic ity of the drugs to SK-OV-3 (P-gp-negative) cells[14]. The non-cytotoxic concentrations of Tet potentiated the growth-inhibitory actions of doxorubicin (Dox) in the Har-resistant HL60 cells. The colony formation efficencies were reduced from 60% by Dox to 0.2% by Tet + Dox. Retardation of the G2M phase cells was increased. But Tet did not potentiate Dox cytotoxity in the sensitive HL60 cells. Dox accumulation in the harringtonine-resistant HL60 cells treated by with was increased. These results indicated that Tet enhanced the cytotoxicity of MDR-related drugs via modulation of P-gp[15]. In addition, Tet could also inhibit platelet- activating factor-induced human platelet aggregation and decrease thromboxane B2 production and thrombus formation[16].

THERAPEUTIC USE IN DIGESTIVE SYSTEM DISEASES
Protective effects on hepatocyte injury

Hepatocyte lesions are common and very important clinically[17-21]. Chen et al[22] observed the effects of Tet on hepatocytic injury induced by CCl4. The result showed that, compared with control group, Tet (1-1000 nmol•L¯¹) increased viability of liver cell (from 71% to 72%-89%), reduced lactate dehydrogenase release, and malondialdehyde (MDA) formation. Tet prevented the increase of the intracellular Ca2+ concentration and the attenuation of the membrane microflow of liver cells. Tet (30 mg•kg¯¹•d¯¹via gavage for two weeks) could markedly re duce the elevation of serum alanine aminotransferase, alkaline phosphatatase and MDA induced by azathioprine. The level of reductive glutathione and SOD were not different from the normal control group. Histological changes in the Tet-treated group were slight[23]. The protective effect on CCl4- or azathioprine-injured hepatocytes may be elicited by inhibiting the lipid peroxidation, improving the membrane microflow, and lessening the Ca2+ concentration. With flow-cytometric technique, we demonstrated that 10-60 mg•L¯¹ Tet could concentration-dependently accelerate the G1 phase cells transforming to S phase cells, and increase the level of DNA in the S phase and protein in the G1, G2 phase cells significantly. Further studies indicated that the effect of Tet in promoting hepatocytes proliferation was not related to blockade of Ca2+ influx[24].

Anti-hepatofibrogenetic activity

Tet could significantly reduce the degree of experimental hepatic fibrogenesis induced by CCl4 in rats; the levels of serum hyaluronic acid and procollagen peptide were decreased, and the liver dysfunction was ameliorated, Tet could also obviously inhibit extracellular matrix formation and collagen deposition. In the liver tissue of rats treated with Tet, hepatic stellate cell (HSC) activation, proliferation, and transformation were down-regulated; the number of desmin-positive cells were reduced significantly. The anti-fibrotic effect of Tet had no significant difference from that of colchicine[25]. HSC activation, proliferation, and transforming into fibroblast are the putative events in hepatic fibrogenesis. Tet could significantly inhibit conventional cultured HSC activation and type I and type III collagen mRNA expressions and protein synthesis were down-regulated. Tet could block HSC proliferation colla gen synthesis induced by platelet-derived growth factor (PDGF), reduce the level of PDGF, PDGF receptor (PDGF-R beta1), transforming growth factor beta1 (TGF beta1) and alpha-smooth muscle actin mRNA, and also down-regulate the au tocrine of PDGF, PDGF-R beta1, TGF beta1. These data suggest that Tet may block hepatic fibrogenesis directly and/or through inhibiting cytokine expressions[26,27]. After taking Tet orally for three months, liver functions of the patients with cirrhosis were obviously improved. Administration Tet for six to eighteen months, serum levels of PIIIP and HA of the patients were markedly reduced. Histological examination showed that, compared with pretreatment or placebo, inflammatory cell infiltration was reduced, and even abolis hed, and that the deposition of ECM, type Iand type III collagen were decrea sed significantly[28].

Anti-portal hypertension

Portal hypertension is one of important manifestations of the patients with cirrhosis. Upper gastrointestinal hemorrhage caused by portal hypertension commonly led to the patient’s death. After injecting Tet intravenously (2.0, 6.0 and 20.0 mg•kg¯¹), portal venous pressure and mean arterial pressure were assessed in cirrhotic rats induced by CCl4. The results demonstrated that Tet induced dose-dependent decreases in portal venous pressure and mean arterial pressure. The maximum percentage reductions of portal venous pressure after Tet in the three different dosages were 5.4% ± 1.0%, 9.2% ± 0.8%, and 23.7% ± 1.2% of baseline, respectively. Total peripheral resistance was also reduced by Tet[29,30]. In portal hypertensive rats induced by partial portal vein ligation, Tet (4, 8, 16 and 24 mg•kg¯¹) induced dose-dependent decreases of portal venous pressure and mean arterial pressure after intravenous infusion. Tet (16 mg•kg¯¹) caused the portal venous pressure decreasing from a baseline of 12.5 mmHg to 10.0 mmHg, and the mean arterial pressure from a baseline of 90 mmHg to 80 mmHg. At 24 mg•kg¯¹, Tet reduced portal venous pressure and mean arterial pressure to 20.3% ± 2.4% and 28.4% ± 1.4% of baseline, resp ectively[31]. The effects of Tet on portal hypotension may be attributed to its actions of blocking voltage- and receptor-operated Ca2+ channels in vascular smooth muscle cells, inhibiting intracellular Ca2+ mobilization and dilating peripheral blood vessels. We had previously observed its clinical therapeutic effects on portal hypertension. Taking Tet orally for 2 consecutive years, the esophageal variceal pressure and the portal blood flow in cirrhotic patients with portal hypertension were significantly reduced. The proportion of patients with no recurrent gastrointestinal bleeding during 2 years’ medication of tetrandrine was 87.9%. It is suggested that Tet would be effective for cirrhotic patients with portal hypertension in preventing recurrent variceal bleeding[32].

Therapeutic effect on portal hypertensive gastropathy

Portal hypertensive gastropathy is caused by dysfunction of submucosal circulation and gastric mucosal barrier damage. Recent studies found that Tet increased prostgalandin E2, GMBE and GAM secretion, reduced the degree of gastric mucosa injury, and lowered the portal pressure. This result indicates that Tet may be useful in portal hypertensive gastropathy.

Preventing pancreatic islet beta cells from toxic injury

Pancreatic islet beta cells could be damaged by alloxan (50 mg•kg¯¹ i.v.) in rats, and diabetic animal models were thus prepared. Pancreatic islet beta cells density in experimental groups pretreated with Tet (100 mg•kg¯¹via gavage) at 1.5 h and 5 h prior to alloxan injection increased from the control value of 13 ± 4 to 62 ± 9 and 65 ± 7 (P < 0.001). When the doses of Tet decreased from 100 mg•kg¯¹ to 50 mg•kg¯¹ and 25 mg•kg¯¹, the pancreatic islet beta cell density were 45 ± 5 and 38 ± 4 (P < 0.01 and P < 0.001)[33].

Footnotes

Edited by Lu HM

References
1.  Takemura H, Imoto K, Ohshika H, Kwan CY. Tetrandrine as a calcium antagonist. Clin Exp Pharmacol Physiol. 1996;23:751-753.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 21]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
2.  Imoto K, Takemura H, Kwan CY, Sakano S, Kaneko M, Ohshika H. Inhibitory effects of tetrandrine and hernandezine on Ca2+ mobilization in rat glioma C6 cells. Res Commun Mol Pathol Pharmacol. 1997;95:129-146.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Bickmeyer U, Hare MF, Atchison WD. Tetrandrine blocks voltage-dependent calcium entry and inhibits the bradykinin-induced elevation of intracellular calcium in NG108-15 cells. Neurotoxicology. 1996;17:335-341.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Xuan B, Liu F, Zhang MY, Xiao JG. Inhibitory effects of tetrandrine on intracellular free Ca2+ increase induced by glutamate, serotonin and histamine in dissociated retina cells. J Ocul Pharmacol Ther. 1996;12:331-336.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
5.  Wong KK. Differential effect of tetrandrine on aortic relaxation and chronotropic activity in rat isolated aorta and atria. Planta Med. 1998;64:663-665.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 4]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
6.  Ho LJ, Chang DM, Lee TC, Chang ML, Lai JH. Plant alkaloid tetrandrine downregulates protein kinase C-dependent signaling pathway in T cells. Eur J Pharmacol. 1999;367:389-398.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 26]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
7.  Shen YC, Chen CF, Wang SY, Sung YJ. Impediment to calcium influx and reactive oxygen production accounts for the inhibition of neutrophil Mac-1 Up-regulation and adhesion by tetrandrine. Mol Pharmacol. 1999;55:186-193.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Pang L, Hoult JR. Cytotoxicity to macrophages of tetrandrine, an antisilicosis alkaloid, accompanied by an overproduction of prostaglandins. Biochem Pharmacol. 1997;53:773-782.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 42]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
9.  Chen F, Sun S, Kuhn DC, Lu Y, Gaydos LJ, Shi X, Demers LM. Tetrandrine inhibits signal-induced NF-kappa B activation in rat alveolar macrophages. Biochem Biophys Res Commun. 1997;231:99-102.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 43]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
10.  Lai YL, Chen YJ, Wu TY, Wang SY, Chang KH, Chung CH, Chen ML. Induction of apoptosis in human leukemic U937 cells by tetrandrine. Anticancer Drugs. 1998;9:77-81.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 40]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
11.  Dong Y, Yang MM, Kwan CY. In vitro inhibition of proliferation of HL-60 cells by tetrandrine and coriolus versicolor peptide derived from Chinese medicinal herbs. Life Sci. 1997;60:PL135-PL140.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 52]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
12.  Song Q, Baxter GD, Kovacs EM, Findik D, Lavin MF. Inhibition of apoptosis in human tumour cells by okadaic acid. J Cell Physiol. 1992;153:550-556.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 66]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
13.  Teh BS, Chen P, Lavin MF, Seow WK, Thong YH. Demonstration of the induction of apoptosis (programmed cell death) by tetrandrine, a novel anti-inflammatory agent. Int J Immunopharmacol. 1991;13:1117-1126.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 30]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
14.  Choi SU, Park SH, Kim KH, Choi EJ, Kim S, Park WK, Zhang YH, Kim HS, Jung NP, Lee CO. The bisbenzylisoquinoline alkaloids, tetrandine and fangchinoline, enhance the cytotoxicity of multidrug resistance-related drugs via modulation of P-glycoprotein. Anticancer Drugs. 1998;9:255-261.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 51]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
15.  He QY, Meng FH, Zhang HQ. Reduction of doxorubicin resistance by tetrandrine and dauricine in harringtonine-resistant human leukemia (HL60) cells. Zhongguo Yao Li Xue Bao. 1996;17:179-181.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Kim HS, Zhang YH, Fang LH, Yun YP, Lee HK. Effects of tetrandrine and fangchinoline on human platelet aggregation and thromboxane B2 formation. J Ethnopharmacol. 1999;66:241-246.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 16]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
17.  Wang MR, Le MZ, Xu JZ, He CL. Establishment and application of experimental model of human fetal hepatocytes: protective effects of silybin and polyporus umbellalus polysaccharides on human fetal hepatocytes. China Natl J New Gastroenterol. 1997;3:228-230.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Huang ZS, Wang ZW, Liu MP, Zhong SQ, Li QM, Rong XL. Protective effects of polydatin against CCl (4)-induced injury to primarily cultured rat hepatocytes. World J Gastroenterol. 1999;5:41-44.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Liu XL, Fan DM. Protective effects of prostaglandin E1 on hepatocytes. World J Gastroenterol. 2000;6:326-329.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Hu YY, Liu CH, Wang RP, Liu C, Liu P, Zhu DY. Protective actions of salvianolic acid A on hepatocyte injured by peroxidation in vitro. World J Gastroenterol. 2000;6:402-404.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Zang GQ, Zhou XQ, Yu H, Xie Q, Zhao GM, Wang B, Guo Q, Xiang YQ, Liao D. Effect of hepatocyte apoptosis induced by TNF-alpha on acute severe hepatitis in mouse models. World J Gastroenterol. 2000;6:688-692.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Chen XH, Hu YM, Liao YQ. Protective effects of tetrandrine on CCl4-injured hepatocytes. Zhongguo Yao Li Xue Bao. 1996;17:348-350.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Hao JW, Sun CC, Zhang L, Zhang X, Wang JX. Protective effects of tetr andrine on Azathioprine-injured hepatocytes. Zhongguo Linchuang Yaolixue Yu Zhiliaoxue Zazhi. 1997;2:180-182.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Liu YL, Li DG, Lu HM, Xu QF. Effects of tetrandrine on hepatocyte prol iferation. Shanghai Dier Yike Daxue Xuebao. 1995;15:212-215.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Sun ZQ, Wang YJ, Quan QZ, Zhang ZJ. Comparative studies of tetrandrine and colchicine anti-fibrogenses. Zhongguo Yaolixue Tongbao. 1996;12:345-347.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Tian Z, Liu S, Li D. [Blocking action of tetrandrine on the cell proliferation induced by PDGF in human lung fibroblasts and liver Ito cell of rats]. Zhonghua Yi Xue Za Zhi. 1997;77:50-53.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Liu HL, Lu HM, Li DG, Jiang ZM, Qi F. Effects of tetrandrine and chlorpromazine on synthesis of collagen and hyaluronic acid in cultured human lung fibroblasts. Zhongguo Yao Li Xue Bao. 1995;16:412-414.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Li DG, Lu HM, Xia WX. Significance of serum collagen type III peptide in antifibrogeneses with Ca2 channal blockers. Zhonghua Yixue Zazhi. 1990;29:453-456.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Huang YT, Cheng YR, Lin HC, Chen CF, Hong CY. Haemodynamic effects of chronic tetrandrine treatment in portal hypertensive rats. J Gastroenterol Hepatol. 1997;12:585-589.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
30.  Huang YT, Cheng YR, Lin HC, Chen SM, Hong CY. Haemodynamic effects of chronic octreotide and tetrandrine administration in portal hypertensive rats. J Gastroenterol Hepatol. 1998;13:266-272.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
31.  Huang YT, Liu TB, Lin HC, Yang MC, Hong CY. Hemodynamic effects of chronic tetrandrine and propranolol administration on portal hypertensive rats. Pharmacology. 1997;54:225-231.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 4]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
32.  Li D, Lu H, Li X, Quan Q, Li X, Lu W. Calcium channel blockers in cirrhotic patients with portal hypertension. Chin Med J (. Engl). 1995;108:803-808.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Sun G, Qi Y, Pan Q. [Quantitative analysis of prevention effect of tetrandrine on pancreatic islet beta cells injury in rats]. Zhonghua Yi Xue Za Zhi. 1997;77:270-273.  [PubMed]  [DOI]  [Cited in This Article: ]