Abstract
Chronic kidney disease (CKD) is debilitating, increasing in incidence worldwide, and a financial and social burden on health systems. Kidney failure, the final stage of CKD, is life-threatening if untreated with kidney replacement therapies. Current therapies using commercially-available drugs, such as angiotensin-converting enzyme inhibitors, angiotensin II receptor blockers and calcium channel blockers, generally only delay the progression of CKD. This review article focuses on effective alternative therapies to improve the prevention and treatment of CKD, using plants or plant extracts. Three mechanistic processes that are well-documented in CKD pathogenesis are inflammation, fibrosis, and oxidative stress. Many plants and their extracts are already known to ameliorate kidney dysfunction through antioxidant action, with subsequent benefits on inflammation and fibrosis. In vitro and in vivo experiments using plant-based therapies for pre-clinical research demonstrate some robust therapeutic benefits. In the CKD clinic, combination treatments of plant extracts with conventional therapies that are seen as relatively successful currently may confer additive or synergistic renoprotective effects. Therefore, the aim of recent research is to identify, rigorously test pre-clinically and clinically, and avoid any toxic outcomes to obtain optimal therapeutic benefit from medicinal plants. This review may prove to be a filtering tool to researchers into complementary and alternative medicines to find out the current trends of using plant-based therapies for the treatment of kidney diseases, including CKD.
Keywords: chronic kidney disease, herbal medicine, plant-based therapies, oxidative stress, pathology, inflammation
Introduction
Chronic kidney disease (CKD) is an insidious, multifactorial, and slowly progressive disease, defined using altered kidney structure or dysfunction present for three months or more. 1 CKD is graded into six progressive stages (Stage 1, 2, 3a and 3b, 4, and 5) based on glomerular filtration rate (GFR). 2 While not progressive in everyone, CKD progresses in many people to total and permanent kidney failure, or Stage 5 CKD, previously termed end-stage kidney disease (ESKD).2,3 CKD is one of the major public health burdens in developed and developing countries. Although statistics differ from country to country, the prevalence of this disease has tended to increase from year to year recently. 4 CKD increases rapidly with age, with rates among those aged 75 and over, twice as high as for 65-74 year olds, and around 7 times as high as those aged 18-54 (42%, 21% and 6%, respectively). Prevalence is generally higher in lower socioeconomic groups (14% compared with 8% in higher socioeconomic groups), 5 and Indigenous populations often experience higher risk of CKD than the non-Indigenous populations.6,7
Interest in the health-benefiting properties of plant-based therapies is driven, in part, by the potential to prevent onset, or ameliorate progression, of certain diseases and reduce health care costs. The financial burden to health systems for CKD hospitalizations and treatments, worldwide, is immense and expected to increase.5,6 Complementary and alternative medicines have been used for CKD patients for many years, with reportedly disparate results for improving outcome. 8 In Western countries, these therapies are most likely used to complement conventional medicines, whereas in other countries such as China, other Asian, and African countries, sole use of alternative medicines is practiced for CKD patients. 9 Therefore, an important challenge to researchers is identifying low-cost and effective plant-based therapeutic options for CKD patients in poorer countries and disadvantaged populations. This review will describe the pathogenetic mechanisms of CKD that may be modulated by these therapies and emphasize and summarize information on some plants used for complementary and alternative medicine, using examples from pre-clinical and clinical studies.
Materials and Methods
PubMed, Medline, Google Scholar, Web of Science, Cochrane Library and China Journal Full-text Database for relevant articles published in English were used for this review of pre-clinical and clinical plant-based therapies for CKD. The contents of full articles were checked by Muhammad Ali Khan (MAK) and Glenda Gobe (GG) for quality of investigations. Only English articles were included.
Results
Pre-clinical research using plant-based therapies in in vitro and in vivo experiments demonstrates some robust therapeutic benefits. In the CKD clinic, combination treatments of plant extracts with conventional therapies that are seen as relatively successful currently may confer additive or synergistic renoprotective effects. These results are summarized in the following paragraphs.
Pathogenetic Mechanisms of CKD
The most common causes of CKD are aging, male gender, diabetes, hypertension and lifestyle. 10 Common pathogenetic mechanisms include tubulointerstitial fibrosis and chronic inflammation, tubular atrophy, glomerulosclerosis and proteinuria. 11 Chronic inflammation occurs through a series of biological signaling pathways involving the vasculature and the immune system, leading to the accumulation of inflammatory mediators in the tissue. 12 Fibrosis is a multifaceted cellular response primarily driven by various profibrotic and inflammatory cytokines such as transforming growth factor-β (TGF-β), tumor necrosis factor-α (TNF-α), platelet-derived growth factor, fibroblast growth factor-2, and some of the interleukins (ILs).13,14 Mechanistically, fibrosis and inflammation are exacerbated by oxidative stress. 15 Another key mechanism for kidney fibrosis is epithelial-mesenchymal transition (EMT) which is induced by various factors, including TGF-β, IL-1β and angiotensin II. 16
Hypoxia and inflammation coexist and have interactive roles in CKD. Hypoxia promotes inflammation by increasing vascular permeability, a central process in the movement of inflammatory cells that facilitate the production of inflammatory mediators. In contrast, inflamed tissues are often severely hypoxic because of increased metabolic demands. Some important inflammatory signaling pathways in CKD involve mitogen-activated protein kinase (MAPK), the transcription factor nuclear factor kappa B (NF-κB), p65 and some of the ILs. 17 The MAPK family consists of extracellular signal-regulated kinase, Jun N-terminal kinase and p38 MAPK. NF-κB is a key player in the production of pro-inflammatory cytokines and chemokines, such as TNF-α, IL-1β, IL-6, chemokine motif ligand 2 and macrophage inflammatory protein-2. 17 Hypoxia is closely regulated by hypoxia-inducible factor (HIF). HIF plays a critical role in inflammation and fibrosis during CKD through determining gene transcription, activation of multiple signaling pathways and epigenetic regulation, and contributes to the pathogenesis of comorbidities of CKD such as anemia, and aberrant angiogenesis. 18 Tissue hypoxia also causes mitochondrial dysfunction and oxidative stress, which lead to the generation of reactive oxygen species and reactive nitrogen species. Other leading causes of oxidative stress are proteinuria, uremic toxins, hyperglycemia and increased activity of the intra-renal angiotensin system.15,19
One of the recent research interests for pathogenesis of CKD is continuum of chronic injury after repair of acute kidney injury (AKI).20,21 AKI involves tubular epithelial cell apoptosis and/or necrosis. If the injury is mild, an adaptive repair process can lead to complete recovery. In contrast, if severe or prolonged injury occurs, this may be followed by maladaptive repair that progresses to chronic inflammation, vascular rarefaction, nephron loss, fibrosis and finally progression to CKD. One of the main causes of AKI is ischemia-reperfusion. Ischemia also causes release of damage-associated molecular pattern molecules (DAMPs) by damaged cells.22,23 The DAMPs promote and exacerbate the inflammatory response. 23 Inflammatory mediators such as inducible nitric oxide synthase, and pro- and anti-apoptotic signaling pathways also act in the continuum of disease progression from AKI to CKD. Ultimately, tubulointerstitial fibrosis develops, thereby impairing local oxygenation. Thus, a time-dependent, combined and cumulative interplay of fibrosis, inflammation and tubular cell loss leads finally to progression to CKD. Figure 1 shows relevant selected mechanisms for the pathogenesis of CKD.
Figure 1.
Proposed mechanistic pathways to chronic kidney disease.
Multiple factors and many signaling pathways are responsible for chronic kidney disease (CKD). Ischemia-reperfusion injury (IRI) is one of the key causes of acute kidney injury (AKI) and apoptosis. Hypoxia produces oxidative stress and reactive oxygen species. Tubular inflammation stimulates immune cells to produce and accumulate extracellular matrix (ECM), causing malfunctioning of kidney structure. Chronic tubular inflammation causes vascular rarefaction and nephron loss. The transcription factor nuclear factor-κB (NF-κB) stimulates proinflammatory cytokines. Reactive oxygen species, proinflammatory cytokines and kidney structure malfunction cause kidney fibrosis and, ultimately, CKD.
Current CKD Treatment Strategies
As mentioned previously, there are limited therapies for CKD. Many current therapies rely on modulating the renin angiotensin system (RAS) which consists of angiotensinogen, renin, angiotensin-converting enzyme (ACE), angiotensin II, angiotensin II type 1 receptor and angiotensin II type 2 receptor. The renin-angiotensin-aldosterone system helps to regulate normal blood pressure. 24 Renin and ACE produce angiotensin II, which alone or in combination with other components of the RAS, upregulates TGF-β1 and causes renal fibrosis through extracellular matrix accumulation, via multiple signaling pathways. The RAS also regulates other profibrotic factors that lead to accumulation of extracellular matrix, causing podocyte loss in glomeruli and decrease in GFR. 25 ACE inhibitors (ACEi) and angiotensin receptor blockers (ARB), alone or in combination, are currently the most efficacious treatments for inhibition of the RAS and for maximal therapeutic effect on CKD, 26 although their use is not without controversy for kidney failure. 27 Recently, there has been an increasing interest in diet and the gut microbiome as modulators of CKD progression.28,29 Dietary supplements or natural foods are attractive, beneficial, and feasible complementary strategies to prevent or delay the progression of CKD. Natural products can also prevent kidney fibrosis through suppressing the RAS, inflammation and oxidative stress. 30 These products can also prevent kidney fibrosis by suppressing the Wnt/β-catenin and the TGF-β/small mother against decapentaplegic (Smad) profibrotic signaling pathways. 31 The inclusion of diet regulation in CKD therapies could be cost effective and possibly safer than combined regular drugs in reducing morbidity and mortality of CKD. Figure 2 summarizes the targets for current and alternative CKD therapies.
Figure 2.
Pathways for current and alternative chronic kidney disease treatments.
After conversion of angiotensinogen to angiotensin by renin, angiotensin-converting enzyme (ACE) converts angiotensin I to angiotensin II. Angiotensin II together with angiotensin type 1 and type 2 receptors (AT1R and AT2R) stimulate pro-fibrotic transforming growth factor-β1 (TGF-β1) to produce and accumulate extracellular matrix, causing kidney fibrosis and finally chronic kidney disease (CKD). Inflammation and oxidative stress also stimulate ECM accumulation. The ACE inhibitor (ACEi) inhibits production of angiotensin II. Angiotensin receptor blockers (ARBs) block AT1R and AT2R and so TGF-β1 synthesis is blocked, ultimately inhibiting accumulation of extracellular matrix. Natural products (alternative therapies) may block the final common pathway of kidney fibrosis through multiple mechanisms, including those stimulated by oxidative stress and inflammation.
Plants as Natural Medicines
Plants, algae and fungi have been utilized as natural medicines throughout human history. 32 Medicinal plants are regarded as an acceptable, cheap, easily available and relatively safe source of many active compounds for pharmaceuticals. 9 In China, the use of traditional herbal medicine for kidney disease has shown some advantages over single conventional drug treatment.32,33 The beneficial effect of medicinal plants on kidney disease is often derived from their ability to boost the natural antioxidant defence mechanisms in the body. 34 Different types of phytochemicals such as flavonoids, vitamins, resveratrol, anthocyanin, curcumin and phenolic acid are often found in the plant-based medicines and may act as antioxidant.32,34,35 In this review, plants known to be beneficial in CKD, and to have minimal adverse outcomes, are discussed. Although by no means restricted to these plants, benefit has been demonstrated from Rheum spp. (Rhubarb), Astragalus membranaceus (Astragalus), Cordyceps sinensis (CS), Triptirygium wildfordii Hook F (TwHF), Abelmoschus manihot (L.) medic (AM), Salvia miltiorrhiza (SM), Vitis vinifera (Grape), and Zingiber officinale (Ginger). These plants and their extracts are sometimes used alone, but in many countries they are used in polyherbals for the treatment of kidney disease.36,37 The following section describes these selected plants and their extracts for treating CKD, their mechanisms of action, and their particular beneficial effects for CKD, using pre-clinical and clinical studies. Tables 1 and 2 give a broader list of plants used in pre-clinical and clinical studies for the treatment of CKD.
Table 1.
Medicinal Plants with Known Renoprotective Effects in Pre-clinical Trials.
Common/Botanical Name | Plant part and delivery dose | Animal and renal effect | Ref. |
---|---|---|---|
Abelmoschus manihot | Flower; Huangkui capsule (HKC) (75 mg ‐ 300 mg/kg/d) | Rats: Reduced ER stress and cJun NH2-terminal kinase activation in the liver and kidney; subsequently improved renal injury and also improved lipid metabolic disorders by activating PPARα/γ. | 38 |
Astragalus membranaceus var. mongholicus with Angelica sinensis | Roots (14 g/kg/d); Decoction of water extract | Rats: Decreased urinary protein excretion, up-regulated VEGF and improved kidney microstructure. | 39 |
Cornus officinalis with Radix Astragalus membranaceus | Roots (10 gm – 20 gm/kg/d); Aqueous extract |
Mice: Reduced urinary protein excretion. | 40 |
Cordyceps sinensis with Tripterygium wilfordii Hook. F. | Fruiting body (16 mg/kg/d – 5 g/kg/d); Polyglycosidium powder from both herbs | Rats: Attenuated glomerular damage by preventing the downregulation of podocin and nephrin in diabetic kidney disease. | 41 |
Anthurium schlechtendalii Kunth | Roots (125 mg/kg/d); Aqueous extract |
Rats: Potential nephron-protective action, but not evident in animals under study. | 42 |
Cordyceps cicadae | Fruiting body (46 mg/kg/d – 2 g/kg/d); Extracts | Rats: Inhibited renal fibrosis by inhibiting TGF-β1/CTGF. | 43 |
Cordyceps sobolifera | Fruiting body; Extracts |
Unknown: Prevented progression of glomerulosclerosis. | 44 |
Zingiber officinale | Roots (60 mg/kg); Aqueous extract |
Rats: Prevented glomerular mesangial matrix deposits and protected nephrons. | 45 |
Asparagus racemosus | Roots (125 mg/kg/d); Ethanolic extract |
Rats: Significantly decreased plasma glucose, creatinine, BUN, total cholesterol and triglyceride levels. Attenuated renal hypertrophy, polyuria, hyperfiltration, microalbuminuria, structural abnormalities and oxidative stress. | 42 |
Anrederea cordiforia | Leaves (50 mg/kg/d – 150 mg/kg/d); Ethanolic extract |
Rats: Reduced serum creatinine, urea, influenced renal index and improved kidney function. | 46 |
Urtica dioica | Leaves (100 mg/kg); Ethanolic extract |
Rabbits: Decreased BUN and SCr; increased glutathione and reduced MDA level. | 47 |
Elsholtzia ciliate (Thunb.) Hylander | Whole plant (300 mg/kg – 500 mg/kg); Ethanolic extract |
Rats: Improved tubulointerstitial fibrosis via regulation of TGF-ß and Smad3 expression. | 48 |
Vigna angularis | Seeds (10 mg/kg); Aqueous extract |
Rats: Significantly reduced glucose, triglycerides, VLDL, uric acid, ALT, urea, SCr and increased HDL. | 49 |
Panax ginseng | Whole plant (0.2 g/kg/d – 0.4 g/kg/d); Decoction with water | Mice: Reduced SCr, BUN and histopathology; increased Cr clearance. | 50 |
Rheum palmatum L., Salvia miltiorrhiza, Astragalus membranaceus Fisch. Bunge with Carthamus tinctorius L. | Polyherbal (2 g/kg/d – 8 g/kg/d); Shenkang granules (SKGs) with normal saline |
Rats: Significantly decreased levels of microalbumin, total protein, SCr, BUN and serum lipid. | 51 |
Paeonia lactiflora, Angelica acutiloba, Astragalus membranaceus, Rehmannia glutinosa, Cnidium officinale, Uncaria rhynchophylla with Phellodendron ammurense | Polyherbal (0.15 g/kg – 0.45 g/kg); Shichimotsu-koka-To (SKT) decoction |
Rats: Significantly decreased urinary albumin excretion and prevented loss of ClC-5 in renal cortex. | 52 |
Radix astragali, Salvia miltiorrhiza, Panax notoginseng with Rosewood | Polyherbal (250 mg/kg/d – 500 mg/kg/d); QiShenYiQi (QSYQ) water-ethanol extract |
Rats: Inhibited downstream fibrogenic action and ameliorated renal tubulointerstitial fibrosis. | 53 |
Rheum palmatum, Salvia miltiorrhiza, Cordyceps sinensis, Leonurus sibiricus, Epihedium macranthum, Radix Astragali with Radix Codonopsis Pilosulae | Polyherbal (50 mg/kg); WH30+ |
Rats: Improved Cr clearance and effectively prevented acute renal failure. | 54 |
Curcuma longa | Seeds (1 mg/kg – 5 mg/kg); Powder in food |
Mice: Inhibited MCP-1 mRNA expression and IL-2 mRNA and secretion of MCP-1 and IL-8. | 55 |
Vigna angularis | Flower (300 mg/kg – 500 mg/kg); Aqueous extract |
Rats: Improved renal function of CKD. | 48 |
Mangifera indica | Bark (250 mg/kg); Water and ethanolic extracts |
Rats: Exhibited diuretic effect. | 56 |
Mimosa pudica
|
Leaves (100 mg/kg – 400 mg/kg); Aqueous extract |
Rats: Diuretic; increased electrolyte excretion. | 57 |
Achyranthes aspera | Whole plant (400 mg/kg); Various extracts | Rats: Diuretic; increased electrolyte excretion. | 58 |
Bixa orellana | Leaves (500 mg/kg); Methanolic extract | Rats: Diuretic; increased electrolyte excretion. | 59 |
Taraxacum officinale/Dandelion | Leaves (50 ml/kg); Aqueous extract |
Mice: Potent diuretic comparable to furosemide. | 60 |
Orthosiphon stamineus | Unknown (100 mg/kg – 200 mg/kg); Methanolic extract | Rats: Decreased SCr, blood urea, urinary protein and extent of renal damage. | 61 |
Strychnos potatorum | Seeds (200 mg/kg); Ethanolic extract |
Rats: Normalize BUN, blood protein and SCr. | 62 |
Aerva javanica | Roots (400 mg/kg); Aqueous extract |
Rats: Recover functional biomarkers of kidneys. | 63 |
Ficus religiosa L. | Latex (200 mg/kg); Methanolic extract |
Rats: Increased MDA, decreased SOD, catalase, glutathione level, exhibited nephroprotective and curative activity. | 64 |
Vernonia cinerea | Aerial plant parts (500 mg/kg); Various extracts | Rats: Pronounced curative, prophylactic, and protective action on the Kidney. | 65 |
Acorus calamus | Aerial plant parts (250 mg/kg – 500 mg/kg); Various extracts | Rats: Increased Hb, leukocytes, packed cell volume and mean corpuscular volume, Cr clearance, body weight; decreased neutrophils, granulocytes, uric acid and platelets. | 66 |
Boerhaavia diffusa | Roots (200 mg/kg – 400 mg/kg); Aqueous extract |
Rats: Prevented acetaminophen-induced nephrotoxicity. | 67 |
Aegle marmelos
|
Leaves (250 mg/kg – 750 mg/kg); Aqueous extract |
Rats: Significantly reduced MDA, SCr, urea and BUN; increased GSH and catalase in gentamicin toxicity. | 68 |
Euphorbia neriifolia | Leaves (150 mg/kg – 400 mg/kg); Hydro alcoholic extract | Rats: Significantly restored antioxidant enzymes in the kidney and exhibited protective effect. | 69 |
Pomegranate (Punica granatum) | Peel (286 mg/kg – 667 mg/kg); Ethanolic extract |
Rats: Significantly decreased lipid peroxidation, serum urea and Cr levels; recovered glutathione-S-transferase and antioxidant enzymes. | 70 |
Sesamum indicum | Seeds (500 mg/kg); Ethanolic extract |
Rats: Decreased serum total protein, albumin and globulin; increased blood urea, SCr and uric acid. | 71 |
Launaea procumbens | Aerial plant parts (100 mg/kg – 200 mg/kg); Various extracts |
Rats: Improved Cr clearance; decreased proteinuria and hematuria. | 72 |
Rutin (extracted from, for example, onions, apples, red grapes/wine) | Bioflavonoid (100 mg/kg/d); Powder | Rats: Rutin improved kidney and heart structure and function, decreased HO-1 and PLA-2 expression in a model of CKD. | 73 |
Camellia sinensis | Leaves; Tea polyphenol |
Rats: Reduced tubulointerstitial fibrosis, inflammation, prevented obesity related CKD in offspring programmed by maternal malnutrition. | 74 |
Abbreviations: ALT, alanine amino transferase; BUN, blood urea nitrogen; CIC-5, chloride channel 5; CTGF, connective tissue growth factor; CKD, chronic kidney disease; Cr, creatinine; ER, endoplasmic reticulum; ECM, extracellular matrix; GSH, glutathione; Hb, hemoglobin; HO-1, heme-oxygenase-1; HDL, high density lipoprotein; IL-2, interleukin-2; IL-8, interleukin-8; MDA, malondialdehyde; MCP-1, monocyte chemoattractant protein-1; PPARα/γ, peroxisome proliferator-activated receptor α/γ; PLA-2, phospholipase A2; SCr, serum creatinine; SOD, superoxide dismutase; TGF-β1, transforming growth factor β1; VEGF, vascular endothelial growth factor; VLDL, very low density lipoprotein.
Table 2.
Medicinal Plants with Known Beneficial Effects in Human Clinical Trials.
Common/Botanical Name | Plant part and delivery dose | Renal effect | Ref. |
---|---|---|---|
Rheum officinale; Radix pseudostellariae; Coptis chinensis; Carthamus tinctorius; Salvia miltiorrhiza with Bidentate achyranthes. | Polyherbal; Shen shuaining capsule (1.4 g-2.1 g) |
Reduced SCr, BUN; increased Hb; improved signs and symptoms in patients with CKD. | 36 |
Rheum species | Whole rhubarb plant; Capsule (350 mg) |
Reduced blood glucose, blood urea, SCr, urine protein; increased Hb, urine volume and GFR. | 75 |
Rheum palmatum | Unknown; Baoshen pill |
Improved serum albumin, lipoprotein and apolipoproteins. | 76 |
Rheum officinale | Unknown; Powder plus ACEi |
Decreased SCr, BUN and CKD. | 77 |
Panax ginseng, Astragalus membranaceus, Cassia cinnamomi, Glycyrrhiza uralensis with Rheum palmatum. | Polyherbal; Dahuang decoction |
Reduced BUN, Cr and improved quality of CKD patients. | 78 |
Astragalus membranaceus | Roots; Tablet (15 g) |
Decrease Pr/Cr; increased serum albumin and complete remission of proteinuria. | 79 |
Tripterygium wilfordii Hook F (TwHF) with Irbesartan | Triptolide (1-2 mg); with Irbesartan (150-300 mg) | Significantly reduced excretion of proteins and podocytes; decreased CTGF and TGF-β1. | 80 |
Grape (Vitis viniferae) | Seeds powder (350 mg) | Increase GFR; decreased proteinuria, depressed triglyceride and prevent anemia. | 81 |
Cordyceps sinensis | Fruiting body; Capsule (2 g) |
Ameliorate glomerulosclerosis, renal interstitial fibrosis; decrease triglycerides, lipoproteins, TCH, possess renoprotective and curative effect on CKD. | 82 |
Glycyrrhiza glabra
|
Glycyrrhetinic acid food supplement (500 mg) | Decreased serum potassium concentration and frequency of severe hyperkalaemia. | 83 |
Silybum marianum | Silibinin (350 mg) | Restored imbalance of thiols in patients with end stage diabetic nephropathy. | 84 |
Lespedeza capitate | Unknown; Tincture |
Potential benefit for patients with acute and chronic renal failure. | 85 |
Beta vulgaris | Roots (300 mg); Concentrated juice |
Reduced peripheral systolic, diastolic, mean arterial pressure and also reduced renal resistive index significantly. | 86 |
Coptis species | Berbarine (0.1 g) | Significantly reduced inflammation, OS, renal damage biochemical markers (UACR, urinary osteopontin KIM-1); improved renal hemodynamics. | 87 |
Parietaria judaica | Leaves; Tonic/tea |
Completely safe and effective for the treatment of CKD. | 85 |
Curcuma longa | Curcumin capsule (600 mg – 1500 mg) | Reduced OS of diabetic or nondiabetic proteinuric CKD patients. | 88 |
Curcuma longa with Boswellia serrata | Unknown; Extract (824 and 516 mg) |
Decreased PGE2 and ameliorated inflammation in patients with CKD. | 89 |
Bupleurum root; Pinellia tuber;Alisma rhizome; Scutellaria root; Ginseng; Poria sclerotium; Polypoms sclerotium; Astractylodes lancea rhizome; Jujube; Glycyrrhiza; Cinnamon bark with Ginger. | Roots/bark; Sairei-to polyherbal |
Significantly reduced urinary protein excretion, hematuria and normalized proteinuria. | 90 |
Abbreviations: BUN, blood urea nitrogen; CKD, chronic kidney disease; CTGF, connective tissue growth factor; Cr, creatinine; Hb, hemoglobin; KIM-1, kidney injury molecule 1; OS, oxidative stress; PGE2, prostaglandin E2; SCr, serum creatinine; TGF-β1, transforming growth factor-β1; TCH, total cholesterol; UACR, urine albumin/creatinine ratio.
Rheum spp. (Rhubarb)
Rhubarb, derived from the root of Rheum spp indigenous to Asia, belongs to the Polygonaceae family. 91 Some species are cultivated for their potential to treat CKD.92,93 Rhubarb contains compounds such as saponins, flavonoids, volatile oils, polysaccharides, tannins, stilbene glycosides (resveratrol and piceatannol) and anthraquinone glycosides (physcion, aloe emodin, chrysophanol, emodin and rhein). The anthraquinone glycosides may have some inherent toxicities, but they can be removed from extracts 94 to produce an effective extract that is nephroprotective. 95 Many clinical and pre-clinical trials have consistently shown that extracts of rhubarb can reduce serum creatinine levels and offset other metabolic dysfunction related to kidney failure.
In CKD therapy, rhubarb increases the excretion of nitrogenous and other waste products through the intestine 75 and ameliorates uremic toxin accumulation, as demonstrated in various pre-clinical animal models of kidney failure. Using a model of diabetic nephropathy in mice, the proposed mechanism was thought to target the gut-kidney axis and trigger protective gut microbiota, rather than being directly nephroprotective. 96 The two primary active anthraquinones in rhubarb are rhein and emodin. Rhein prevents kidney damage by modulating various signaling pathways. It down-regulates the Wnt/β-catenin signaling pathway, up-regulates Sirtuin 1, decreases EMT that normally leads to fibrosis and ameliorates dyslipidemia.96–99 Rhein also improves cell metabolism through modulation of the glucose transporter I, decreasing mesangial cell hypertrophy and glomerulosclerosis. 100 Emodin reduces glycation of proteins, inhibits the lipopolysaccharide-induced expression of TLR4 and down-regulates pro-inflammatory TNF-α and IL-6 in the damaged kidney. 101 Emodin also decreases mesangial cell proliferation by inhibiting cellular FLICE-like inhibitor protein, 102 TGF-β1 and fibronectin, 103 p38 MAPK, 104 differentiation and maturation of dendritic cells, and increases the number of regulatory T cells. 105 These results indicate that the active ingredients in rhubarb have multiple mechanisms of action for the treatment of CKD, including regulation of inflammation and the immune response. Most of these reports have used pre-clinical models. 106 Clinically, rhubarb extract alone, or as a polyherbal, was beneficial in CKD patients, 75 delaying CKD progression and decreasing adverse effects of hemodialysis. The most common adverse side effects of rhubarb are nausea, vomiting, diarrhea, electrolyte disorders and liver toxicity.107,108
Astragalus membranaceus (Astragalus)
Astragalus membranaceus (synonymous name of Astragalus propinquus), belongs to the family Fabaceae and the genus Astragalus, which has more than 3000 species worldwide. 109 The plant is indigenous to the north and eastern regions of China but is grown worldwide and used widely in complementary and alternative medicines. Astragalus contains more than 60 bioactive compounds, including polysaccharides, saponins (astragalosides I–VII), flavonoids, amino acids and trace elements.110,111 In vitro and in vivo pre-clinical trials have revealed that Astragalus extract has potent antioxidant and anti-inflammatory effects.112,113 Other mechanisms include downregulating angiotensin receptors, inhibiting nitric oxide synthase and TNF-α production, and stimulating vascular endothelial growth factor and the immune system. 114 In addition, the plant extracts from Astragalus can rebalance profibrotic TGF-β/Smad signaling activity and inhibit endoplasmic reticulum stress-induced pathways.115,116
Several pre-clinical CKD models reported that the plant extract, either alone or as a polyherbal, showed anti-fibrotic effects, ameliorated proteinuria, attenuated kidney injury and promoted Reno protective effects.117,118 Wojcikowski et al. performed a pre-clinical trial in rats using the unilateral ureteral obstruction (UUO) model 119 of kidney fibrosis, with or without Astragalus membranaceus and Angelica sinensis (A and A). The ACEi Enalapril was administered in drinking water in some groups. The combined effect of A and A, with or without Enalapril, had anti-fibrotic benefits. Several clinical trials have reported that Astragalus significantly ameliorated CKD by decreasing proteinuria, with a corresponding increase in creatinine clearance, and hemoglobin levels.79,120 Astragalus is safe for most of the recipients. However, the plant is known to inhibit CYP3A4 hence it could affect the action of some other drugs that are metabolized by this enzyme. 121
Cordyceps sinensis (CS)
CS is a fungus that derives its nutrients from the larvae of Lepidoptera. 122 It is endemic to the Tibetan plateau but is produced commercially now. Various bioactive compounds, including amino acids, polysaccharides, organic acids, trace elements, nucleosides, peptides, steroids and other chemical components are present in CS extracts.123,124 The ability of CS to ameliorate urinary disorders and edema was described over 2000 years ago in traditional Chinese medicine. 9 In vitro studies with CS demonstrate potent antioxidant capacity. 125 In vivo, glomerular protection via inhibition of mesangial cell proliferation was demonstrated. 126 Treatment with CS also decreased apoptosis and kidney tubular atrophy, 127 and enhanced the immune system. 128
Extensive work has been carried out on CS alone, or in combination with other existing drugs, or other plant extracts. Different pre-clinical animal studies have been used to observe the beneficial effect on kidney disease, especially in CKD. In rat models of CKD, CS improved kidney function, reduced cardiovascular and endocrine comorbidities, inhibited oxidative stress, and reduced CKD-triggering metabolites.129–131 Clinically, CS extracts significantly reduced the levels of serum creatinine, proteinuria, hematuria, inflammatory cytokines, monocyte chemo attractant protein-1 and TNF-α. 132 Clinical trials of CS, alone or in combination with other medicinal plant extracts or existing drugs, showed significant improvement of kidney function, with reduced levels of serum creatinine, albuminuria, proteinuria, blood urea nitrogen (BUN) and profibrotic TGF-β1, and increased creatinine clearance.133–137 There are very limited reports of adverse effects of dietary supplements of CS in humans. The most common adverse side effects are nausea, dry mouth and stomach upset. 137
Tripterygium wilfordii Hook F (TwHF)
TwHF is a member of the Celastraceae family, and is native to Eastern and Southern China, Korea, Japan and Taiwan. 138 A stable glycoside named “multi-glycoside of TwHF” (GWT) 139 has been approved by the China State Food and Drug Administration (Z32021007) for the treatment of chronic diseases such as rheumatoid arthritis and glomerulonephritis. In China, GWT has been used extensively for the treatment of proliferative glomerulonephritis for more than three decades.140,141 The major active compound of GWT is triptolide. 142 GWT showed several beneficial therapeutic effects via its immunosuppressive, immunomodulatory, anti-inflammatory, antioxidant and antitumor activities.143,144
Pre-clinical studies of CKD have demonstrated that TwHF reduced tubular apoptosis and kidney fibrosis, as well as improving kidney function by inhibiting the TGF-β1/Smad3 signaling pathways. 144 Clinically, GWT in combination with irbesartan reduced excretion of urinary proteins, including urinary connective tissue growth factor and TGF-β1, in diabetic nephropathy patients. 145 In combination with an ARB, podocyte injury was reduced in a similar patient population. 80 However, the use of TwHF extract is limited due to the lack of high-quality clinical trial data. 146 TwHF causes some common adverse side effects, such as stomach upset, diarrhea, skin rash and changes in skin pigmentation. 147 The other concerning, although reversible, side effects are infertility in men and amenorrhea in women. 148 These adverse side effects limit its usefulness for the treatment of CKD.
Abelmoschus manihot (L.) medic (AM)
AM belongs to the Malvaceae family. It has been used as a traditional medicine for hundreds of years in China, notably for the treatment of CKD. 149 Due to its potential renoprotective properties, the State Food and Drug Administration of China approved the Huangki capsule (HKC, made from AM) as a Class III new drug (GUOYAOZHUNZI Z19990040) for the treatment of chronic glomerulonephritis. The main active components of HKC are total flavonoids extracted from the flowers of AM, sometimes known as Aibika flowers. Recent research considered HKC as an important adjuvant treatment for CKD, especially as a side effect of diabetes, and suggested that HKC is more effective than the ARB losartan in reducing proteinuria.150,151
The beneficial effect of AM or HKC on CKD and other kidney complications relate to potential anti-inflammatory and antioxidant activities, probably via the flavonoids. 152 Several pre-clinical studies in animal models demonstrated that HKC reduced urinary albumin excretion, serum triglycerides, cholesterol levels and podocyte apoptosis. 153 Kidney inflammation and glomerular injury were reduced as evidenced by fewer activated macrophages in glomeruli, reduced TNF-α protein expression in the kidney, and a dampened p38 MAPK signaling pathway.154,155 There are limited data on AM (or HKC) as an anti-fibrotic agent in clinical trials. AM alone, or together with other drugs, reduced levels of proteinuria, BUN, serum creatinine, and also decreased oxidative stress. 156 There were no serious adverse effects from taking AM, except mild to moderate gastrointestinal discomfort.
Salvia miltiorrhiza (SM)
SM, commonly known as Danshen, is a perennial plant of the genus Salvia, within the family Lamiaceae. It is native to most Asian countries, including China. The roots and rhizomes of the plant have been widely used in traditional Chinese medicine for thousands of years. 157 More than 100 bioactive compounds have been isolated from this plant and characterized for their structure. SM has been used for the treatment of numerous diseases, including CKD, because of its potent anti-inflammatory and antioxidant properties.158–161 The bioactivity of SM extract relates to the presence of phenolics, flavonoids, terpenoids, polysaccharides and other related quinone compounds. 160 For instance, Tanshinone IIA, a di-terpenoid isolated from SM, showed antioxidant benefit by activating the Nrf2 signaling pathway, improving the metabolic profile and exhibiting potent renoprotective effects, 160 and the lipophilic fraction attenuated oxidative stress in diabetic nephropathy. 161
Pre-clinical trials of SM alone, or in combination with other drugs, reduced albuminuria, improved kidney function, decreased fibrosis and TGF-β/Smad3 signaling, suppressed pro-inflammatory cytokines and adhesion molecules, and inhibited apoptosis.162,163 SM extracts also ameliorated proteinuria, serum and kidney tissue levels of TGF-β1 and collagen IV, reduced numbers of monocytes/macrophages and exhibited a significant renoprotective effect. Of note, a clinical trial of a polyherbal that contained SM for CKD stabilized the albumin/creatinine ratio, increased hemoglobin content and improved accumulative survival rate of the test group. 164 No serious adverse side effects of SM have been reported.
Vitis vinifera (grape)
Vitis vinifera, or grape, belongs to the Vitaceae family. 165 Grape seeds contain more than 1600 phytonutrients, including flavonoids, catechin, anthocyanins, flavonols, non-flavonoids and pro-anthocyanidins, and their extracts exhibit enormous biological activity. 166 For this review, one of the most important of the biological actions is inhibition of progress in CKD. 167 Grape seed extracts typically act by alleviating oxidative stress and endoplasmic reticulum stress-induced apoptosis.168–170 Pre-clinical studies of grape seed extracts in animal models of CKD have also reported improved kidney function, decreased proteinuria and reduced podocyte cell death. 171 Clinical trials have demonstrated that grape seed extracts increased GFR, decreased proteinuria and triglycerides, prevented anemia, and counteracted plasma low density lipoprotein and thrombocytopenia. 81 There are no known adverse side effects at normal doses of grape seed extracts.
Zingiber officinale (ginger)
Ginger is used widely as a spice but also often in folk medicine. It belongs to the Zingiberaceae family and has been cultivated for thousands of years especially in China and the South Asian countries. 172 Ginger contains many beneficial compounds, the most important of which are 6-, 8-, and 10-gingerol and 6-shogaol. 173 It displays diverse beneficial biological actions due to its potent antioxidant, anti-inflammatory, anti-tumour, anti-diabetic and neuroprotective activities. In pre-clinical animal models of kidney and cardiovascular diseases, ginger extracts lowered blood glucose levels, restored the total carbohydrates, pyruvate, glycogen and total protein in kidney tissue, promoted the regeneration of tubules and restored glomeruli, and reduced fatty infiltration.174–177 One clinical trial of ginger extract in CKD patients on peritoneal dialysis, demonstrated that daily administration of 1000 mg ginger reduced serum fasting glucose, a risk factor for diabetes, diabetic nephropathy and cardiovascular disease. 178 There have been no adverse side effects reported when doses are kept to a moderate level.
Some other nephroprotective medicinal plants
There are many other plants reported to have benefits for kidney failure and/or CKD. Rutin (or quercetin) is a flavonoid present in onions, apples, tea and red wine. This plant extract is readily available “over the counter” and has demonstrated strong antioxidant and anti-inflammatory properties in heart and liver in a high fat-high carbohydrate diet model of metabolic syndrome in rats. 179 Rutin also protected the kidney against ischemia-reperfusion injury, 180 cisplatin-induced nephrotoxicity, 181 and diabetic nephropathy. 182 It normalized BUN, thereby modulating a key factor in the pathogenesis of CKD. 73 However, some other studies did not show benefits. For example, rutin may induce protein-energy malnutrition in CKD. 183 Although there are some promising results, more analyzes are needed to confirm whether, or not, preclinical and clinical benefits exist for this plant extract, particularly in the context of CKD. Clinically, Glycyrrhiza glabra extracts consistently decreased pre-dialysis serum potassium concentrations in chronic hemodialysis patients. 83 Silybum marianum, known as “milk thistle” or silymarin, is a very safe herb that protects against kidney failure and end-stage diabetic nephropathy. 184 Significant benefits have been claimed for Lespedeza tincture for both AKI and CKD patients. 185 Clinically, beetroot juice decreased peripheral systolic and diastolic blood pressure, mean arterial pressure, improved kidney function, histological damage and kidney prognosis, and prevented cardiovascular events.86,186 In a recent study, a beetroot protease inhibitor was isolated and characterized. 187 The results showed the potential of such plant protease inhibitors for peptide-based drug discovery against targets involved in diseases such as cancers and immune system related diseases, such as is seen in CKD in some instances. Coptis rhizome extracts significantly reduced biomarkers of kidney damage, such as urinary albumin-to-creatine ratio, urinary osteopontin and KIM-1, and improved kidney hemodynamics. In addition, clinically, it decreased inflammation and oxidative stress. 87 Extracts of Urtica dioica, or “stinging nettle”, significantly attenuated kidney damage and tubular atrophy, loss of brush border, hydropic epithelial cell degeneration, glomerular shrinkage, and tubulointerstitial fibrosis, as well as demonstrating clinical benefits in patients undergoing partial nephrectomy or kidney transplantation. 188 The polyherbal named “Sairei-To” significantly decreased urinary protein excretion, hematuria and normalized proteinuria in CKD patients. 90 A dietary supplementation with curcumin (turmeric) reduced oxidative stress and proteinuria in CKD patients. 88 A combination of Curcuma longa and Boswellia serrata decreased the levels of inflammatory cytokines and ameliorated inflammatory markers in patients with CKD. 89
Concerns of Toxicities of Plant-Based Extracts to the Kidney
Natural products are often considered safer than traditional drugs, and many of our modern drugs are derived from herbs. Nonetheless, some researchers are concerned over their safe use.189–191 The possible nephrotoxicity and other chronic or insidious health disorders of using popular complementary and alternative medicine in different countries is not properly reported. The most well-known toxicity or side-effect is associated with traditional Chinese herbal medication that uses aristolochic acid and induces aristolochic acid nephropathy (AAN). The clinical features of AAN are characterized by extensive interstitial fibrosis and tubular atrophy in the kidney without obvious glomerular injury. Prolonged use causes uroepithelial malignancies. 189 In addition, traditional herbal remedies have been implicated in 35% of all cases of AKI in Africa. 190 The Therapeutic Goods Administration restricted the use of ephedra in Australia due to its potential toxic effects (quickened heartbeat and elevated blood pressure, heart palpitations, nausea and vomiting). 189 Some herbal drugs that contain specific herbs, nuts and mushrooms may also be associated with inherent nephrotoxicity. Interestingly, the inherent properties of the herbs are not the only source of herb-associated kidney disorders. Herb-drug interactions, mistakes in dosage and identification, contaminants within mixtures, adulteration with heavy metals, and even deliberate adulteration with non-labelled plant extracts are all issues of concern. 191 However, with proper identification, rigorous pre-clinical and clinical trials, strict controls on the presence of adulterants within herbal medicines, labeling of dosages and contraindications, and robust manufacturing techniques, the safety of those consuming herbal medicines should be maintained.
Conclusion
There is no doubt that medicinal plants present a largely untapped source of new CKD therapies. Clinical and pre-clinical trials of plant extracts sometimes demonstrate benefit but some research has also demonstrated that plant extracts may produce chronic organ dysfunction when used for long term due to the presence of harmful chemicals. Therefore, the aim of recent research is to identify, rigorously test pre-clinically and clinically, and avoid such toxic outcomes in order to obtain optimal therapeutic benefit from medicinal plants. This review may prove to be a filtering tool to researchers into complementary and alternative medicines to find out the current trends of using medicinal plants and plant extracts for the treatment of kidney diseases, including CKD.
Acknowledgments
Muhammad Ali Khan is supported by a University of Queensland Research Training Scholarship (RTP) and a Top-up Scholarship from the Kidney Disease Research Collaborative, Translational Research Institute, Brisbane Australia. This group received National Health and Medical Research Council of Australia support from its Centre of Research Excellence grant, 2017-2020.
Abbreviations List
- A and A
Astragalus membranaceus and Angelica sinensis
- AAN
aristolochic acid nephropathy
- AKI
acute kidney disease
- AM
Abelmoschus manihot (L.) medic
- CKD
chronic kidney disease
- CS
Cordyceps sinensis
- DAMPs
damage-associated molecular patterns
- HIF
hypoxia inducible factor
- HKC
Huangki capsule
- IL
interleukin
- MAPK
mitogen activated protein kinase
- NF-κB
nuclear factor kappa B
- SM
Salvia miltiorrhiza
- TGF-β
transforming growth factor-beta
- TNF-α
tumor necrosis factor-alpha
- TwHF
Tripterygium wilfordii Hook
Footnotes
Author Contributions (Roles): Each author has participated sufficiently in the work to take public responsibility for the content. M.A.K. and G.G. conceptualised the review article. All authors contributed in writing, reviewing and editing of the original draft. M.A.K. compiled the manuscript and G.G. supervised the study.
Declaration of Conflicting Interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
Funding: The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This work was supported by the National Health and Medical Research Council of Australia.
ORCID iD: Muhammad Ali Khan https://orcid.org/0000-0002-6980-6112
References
- 1.Levey AS, Coresh J. Chronic kidney disease. The lancet. 2012;379(9811):165‐180. [DOI] [PubMed] [Google Scholar]
- 2.Chen TK, Knicely DH, Grams ME. Chronic kidney disease diagnosis and management: a review . Jama. 2019;322(13):1294‐1304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Levey AS, Eckardt KU, Dorman NM, et al. Nomenclature for kidney function and disease: report of a kidney disease: improving global outcomes (KDIGO) consensus conference. Kidney Int. 2020;97(6):1117‐1129. [DOI] [PubMed] [Google Scholar]
- 4.Xie Y, et al. Analysis of the Global Burden of Disease study highlights the global, regional, and national trends of chronic kidney disease epidemiology from 1990 to 2016 . Kidney Int. 2018;94(3):567‐581. [DOI] [PubMed] [Google Scholar]
- 5.Jha V, et al. Chronic kidney disease: global dimension and perspectives. The Lancet. 2013;382(9888):260‐272. [DOI] [PubMed] [Google Scholar]
- 6.Glassock RJ, Warnock DG, Delanaye P. The global burden of chronic kidney disease: estimates, variability and pitfalls . Nature Reviews Nephrology. 2017;13(2):104. [DOI] [PubMed] [Google Scholar]
- 7.Hoy WE, Wendy EHFRACP, Smith PK, Hughson MD, Fogo AB, Sinniah R, Dowling J, et al. CKD in aboriginal Australians . Am J Kidney Dis. 2010;56(5):983‐993. [DOI] [PubMed] [Google Scholar]
- 8.Zhong Y, Zhong Y, Deng Y, Chen Y, Chuang PY, He. et al. Therapeutic use of traditional Chinese herbal medications for chronic kidney diseases . Kidney Int. 2013;84(6):1108‐1118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Gobe GC, Wojcikowski K. Nontraditional (non-Western pharmaceutical) treatments for chronic kidney disease . Clin Nephrol. 2019; 93(1): 49-54. [DOI] [PubMed] [Google Scholar]
- 10.Webster AC, Nagler EV, Morton RL, Masson P. et al. Chronic kidney disease. The lancet. 2017;389(10075):1238‐1252. [DOI] [PubMed] [Google Scholar]
- 11.Schnaper HW. The tubulointerstitial pathophysiology of progressive kidney disease . Adv Chronic Kidney Dis. 2017;24(2):107‐116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Meng X-M, Nikolic-Paterson DJ, Meng XM, Nicolic PDJ, Lan HY. Inflammatory processes in renal fibrosis . Nature Reviews Nephrology. 2014;10(9):493. [DOI] [PubMed] [Google Scholar]
- 13.Djudjaj S, Boor P. Cellular and molecular mechanisms of kidney fibrosis . Mol Asp Med. 2019;65:16‐36. [DOI] [PubMed] [Google Scholar]
- 14.Meng X-m, Nikolic-Paterson DJ, Meng X, Nicolic PDJ, Lan HY. TGF-β: the master regulator of fibrosis. Nature Reviews Nephrology. 2016;12(6):325. [DOI] [PubMed] [Google Scholar]
- 15.Duni A, et al. Oxidative stress in the pathogenesis and evolution of chronic kidney disease: untangling ariadne’s thread. Int J Mol Sci. 2019;20(15):3711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Lovisa S, Zeisberg M, Kalluri R. Partial epithelial-to-mesenchymal transition and other new mechanisms of kidney fibrosis . Trends in Endocrinology & Metabolism. 2016;27(10):681‐695. [DOI] [PubMed] [Google Scholar]
- 17.Chen L, Deng H, Hengmin Cui. et al. Inflammatory responses and inflammation-associated diseases in organs . Oncotarget. 2018;9(6):7204-7218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Liu J, Wei Q, Guo C, et al. Hypoxia, HIF, and associated signaling networks in chronic kidney disease. Int J Mol Sci. 2017;18(5):950. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Tanaka T. Expanding roles of the hypoxia-response network in chronic kidney disease . Clin Exp Nephrol. 2016;20(6):835‐844. [DOI] [PubMed] [Google Scholar]
- 20.Ferenbach DA, Bonventre JV. Mechanisms of maladaptive repair after AKI leading to accelerated kidney ageing and CKD . Nature Reviews Nephrology. 2015;11(5):264-276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Guzzi F, et al. Molecular mechanisms of the acute kidney injury to chronic kidney disease transition: an updated view . Int J Mol Sci. 2019;20(19):4941. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Mulay SR, Kumar SV, Lech M, Desai J, Anders. et al. How kidney cell death induces renal necroinflammation . In: Seminars in Nephrology. Elsevier; 2016; 36(3): 162-173. [DOI] [PubMed] [Google Scholar]
- 23.Poluzzi C, et al. Biglycan evokes autophagy in macrophages via a novel CD44/Toll-like receptor 4 signaling axis in ischemia/reperfusion injury . Kidney Int. 2019;95(3):540‐562. [DOI] [PubMed] [Google Scholar]
- 24.Xie X, et al. Renin-angiotensin system inhibitors and kidney and cardiovascular outcomes in patients with CKD: a Bayesian network meta-analysis of randomized clinical trials . Am J Kidney Dis. 2016;67(5):728‐741. [DOI] [PubMed] [Google Scholar]
- 25.Wennmann DO, Hsu H-H, Hsu HH, Pavenstädt H. The renin-angiotensin-aldosterone system in podocytes . In: Seminars in Nephrology. Elsevier; 2012; 32(4): 377-384. [DOI] [PubMed] [Google Scholar]
- 26.Bhandari S, Ives N, Brettell EA, et al. Multicentre randomized controlled trial of angiotensin-converting enzyme inhibitor/angiotensin receptor blocker withdrawal in advanced renal disease: the STOP-ACEi trial . Nephrol Dial Transplant. 2016;31(2):255‐261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Qiao Y, et al. Discontinuation of angiotensin converting enzyme inhibitors and angiotensin receptor blockers in chronic kidney disease . In: Mayo Clinic Proceedings. Elsevier; 2019; 94(11): 2220-2229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Aron-Wisnewsky J, Aron WJ, Clément K. The gut microbiome, diet, and links to cardiometabolic and chronic disorders . Nature Reviews Nephrology. 2016;12(3):169. [DOI] [PubMed] [Google Scholar]
- 29.Yang CY, Tarng DC. Diet, gut microbiome and indoxyl sulphate in chronic kidney disease patients . Nephrology. 2018; 23(s4):16‐20. [DOI] [PubMed] [Google Scholar]
- 30.Chen DQ, et al. Natural products for the prevention and treatment of kidney disease . Phytomedicine. 2018;50:50‐60. [DOI] [PubMed] [Google Scholar]
- 31.Zhong Y, et al. Recent advances in traditional Chinese medicine for kidney disease . Am J Kidney Dis. 2015;66(3):513‐522. [DOI] [PubMed] [Google Scholar]
- 32.Wojcikowski K, Johnson DW, Gobe G. Herbs or natural substances as complementary therapies for chronic kidney disease: ideas for future studies . J Lab Clin Med. 2006;147(4):160‐166. [DOI] [PubMed] [Google Scholar]
- 33.Yarnell E. Botanical medicines for the urinary tract . World J Urol. 2002;20(5):285‐293. [DOI] [PubMed] [Google Scholar]
- 34.Shahzad M, et al. Protection against oxidative stress-induced apoptosis in kidney epithelium by Angelica and Astragalus . J Ethnopharmacol. 2016;179:412‐419. [DOI] [PubMed] [Google Scholar]
- 35.Rapa SF, et al. Inflammation and oxidative stress in chronic kidney disease—potential therapeutic role of minerals, vitamins and plant-derived metabolites . Int J Mol Sci. 2020;21(1):263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Wang S, et al. The efficacy of shen shuaining capsule on chronic kidney disease: a systematic review and meta-analysis . Evidence-Based Complementary Altern Med. 2016; 2016: 1-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Bahmani M, et al. Identification of medicinal plants for the treatment of kidney and urinary stones . J Renal Inj Prev. 2016; 5(3):129-133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Ge J, Miao JJ, Sun XY, Yu . et al. Huangkui capsule, an extract from Abelmoschus manihot (L.) medic, improves diabetic nephropathy via activating peroxisome proliferator–activated receptor (PPAR)-α/γ and attenuating endoplasmic reticulum stress in rats . J Ethnopharmacol. 2016;189:238‐249. [DOI] [PubMed] [Google Scholar]
- 39.Meng L, et al. A combination of Chinese herbs, Astragalus membranaceus var. mongholicus and Angelica sinensis, enhanced nitric oxide production in obstructed rat kidney. Vasc Pharmacol. 2007;47(2–3):174‐183. [DOI] [PubMed] [Google Scholar]
- 40.Huang L, Shi X, Liang H. Effects of Radix Astragali and Fructus Corni on urinary protein pattern in nephropathy mice by microfluidic chip . Zhongguo Zhong yao za zhi= Zhongguo zhongyao zazhi= China journal of Chinese materia medica. 2007;32(13):1324‐1328. [PubMed] [Google Scholar]
- 41.Hao L, et al. Effect of Cordyceps sinensis and Tripterygium wilfordii polyglycosidium on podocytes in rats with diabetic nephropathy . Exp Ther Med. 2014;7(6):1465‐1470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Octavio Carvajal-Z DMB-D, Hayward-Jo PM, Aguilar-Us MG, Nolasco-Hi C, Quero-Herr S, Guadalupe G, Zarraval OC, DErmitz DMB, Jones PMH et al. Study of Anthurium schlechtendalii Kunth Extract Effects on Nephroprotective or Renal Damage Remission Capacity . Research Journal of Medicinal Plants. 2017; 11(3): 107-114. [Google Scholar]
- 43.Zhu R, et al. Cordyceps cicadae extracts ameliorate renal malfunction in a remnant kidney model . Journal of Zhejiang University SCIENCE B. 2011;12(12):1024‐1033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Zhu R, Chen Y, Deng Y, et al. The mechanism study of artificial culture Cordyceps sobolifera mycelium preventing the progression of glomerulosclerosis . Chin J Integr Tradit Western Nephrol. 2005;6(70):2005. 74 . [Google Scholar]
- 45.Irshad F, Munawar S, Rasheed A. Effects Of Ginger Extract On Glomerular Mesangial Matrix Of Kidneys In Alloxan Induced Diabetic Nephropathy Of Albino Rats . Journal of Bahria University Medical and Dental College. 2018;8(2):87‐91. [Google Scholar]
- 46.Sukandar E, Fidrianny I, Adiwibowo L. Efficacy of ethanol extract of Anredera cordifolia (Ten) Steenis leaves on improving kidney failure in rats . Int J Pharmacol. 2011;7(8):850‐855. [Google Scholar]
- 47.Salih NA. Effect of nettle (Urtica dioica) extract on gentamicin induced nephrotoxicity in male rabbits . Asian Pac J Trop Biomed. 2015;5(9):756‐760. [Google Scholar]
- 48.Kim TW, et al. Elsholtzia ciliata (thunb.) Hylander attenuates renal inflammation and interstitial fibrosis via regulation of TGF-ß and Smad3 expression on unilateral ureteral obstruction rat model. Phytomedicine. 2016;23(4):331‐339. [DOI] [PubMed] [Google Scholar]
- 49.Baracho NCDV, Monteiro NF, Borges MG, Arguelho RRDM. et al. Effect of aqueous extract of the Vigna angularis in rats subjected to an experimental model of moderate chronic kidney disease1 . Acta Cirúrgica Brasileira. 2016;31(8):527‐532. [DOI] [PubMed] [Google Scholar]
- 50.Doh KC, et al. Ginseng treatment attenuates chronic cyclosporine nephropathy via reducing oxidative stress in an experimental mouse model . Am J Nephrol. 2013;37(5):421‐433. [DOI] [PubMed] [Google Scholar]
- 51.Zhang Y, et al. Effect of Shenkang granules on the progression of chronic renal failure in 5/6 nephrectomized rats . Exp Ther Med. 2015;9(6):2034‐2042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Ma Y, Fujimoto M, Watari H, Kimura M,Shimada. et al. The renoprotective effect of shichimotsukokato on hypertension-induced renal dysfunction in spontaneously hypertensive rats . J Nat Med. 2016;70(2):152‐162. [DOI] [PubMed] [Google Scholar]
- 53.Zhou Z, et al. QiShenYiQi attenuates renal interstitial fibrosis by blocking the activation of β-Catenin . Plos one. 2016;11(9):e0162873. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 54.Ngai HH, Sit W-H, Wan JM. The nephroprotective effects of the herbal medicine preparation, WH30 + , on the chemical-induced acute and chronic renal failure in rats. Am J Chin Med. 2005;33(03):491‐500. [DOI] [PubMed] [Google Scholar]
- 55.Zhong F, et al. Curcumin attenuates lipopolysaccharide-induced renal inflammation . Biol Pharm Bull. 2011;34(2):226‐232. [DOI] [PubMed] [Google Scholar]
- 56.Devi MS. Acute toxicity and diuretic activity of Mangifera indica L. bark extracts . Int J Pharma Bio Sci. 2011;2(3):141‐146. [Google Scholar]
- 57.Sangma TK, et al. Diuretic property of aqueous extract of leaves of Mimosa pudica Linn. on experimental albino rats . J Nat Prod. 2010;3(2010):172‐178. [Google Scholar]
- 58.Srivastav S, et al. Diuretic activity of whole plant extract of Achyranthes aspera Linn . European journal of experimental Biology. 2011;1(2):97‐102. [Google Scholar]
- 59.Radhika B, et al. Diuretic activity of Bixa orellana Linn. leaf extracts . 2010; 3(1): 353-355.
- 60.Râcz–Kotilla E, Racz KE, Racz G, Solomon A. The action of Taraxacum officinale extracts on the body weight and diuresis of laboratory animals . Planta Med. 1974;26(07):212‐217. [DOI] [PubMed] [Google Scholar]
- 61.Mannavalan R. Evaluation of nephroprotective activity of Orthosiphon stamineus Benth extract using rat model . EVALUATION. 2010;2(1):209‐215. [Google Scholar]
- 62.Varghese R, Moideen MM, Suhail MMJ, Dhanapal CK. et al. Nephroprotective effect of ethanolic extract of Strychnos potatorum Seeds in Rat Models. Research journal of pharmaceutical. Biological and Chemical Sciences. 2011;2(3):521‐529. [Google Scholar]
- 63.Movaliya V, Khamar D, Setty M. Nephroprotective activity of aqueous extract of Aerva Javanica roots in cisplatin induced renal toxicity in rats . Pharmacologyonline. 2011;1:68‐74. [Google Scholar]
- 64.Yadav YC, Srivastava DN, Saini V, et al. Nephroprotective and curative Activity of methanolic extract of Ficus religiosa L. latex in albino rats using cisplatin induced nephrotoxicity . Pharmacologyonline. 2011;1:132‐139. [Google Scholar]
- 65.Sreedevi A, Bharathi K, Prasad K. Effect of Vernonia cinerea aerial parts against Cisplatin-induced nephrotoxicity in rats . Pharmacologyonline. 2011;2:548‐555. [Google Scholar]
- 66.Palani S, et al. Therapeutic efficacy of Acorus calamus on Acetaminophen induced nephrotoxicity and oxidative stress in male albino rats . Acta Pharmaceutica Sciencia. 2010;52(1): 89-100. [Google Scholar]
- 67.Pareta SK, et al. Protective effects of Boerhaavia diffusa against Acetaminophen-induced nephrotoxicity in rats . Pharmacologyonline. 2011;2:698‐706. [Google Scholar]
- 68.K KJ, Shete R, Jadhav P. RP-HPLC method of simultaneous nephroprotective role of A. marmelos extract . International Journal of Research in Pharmacy and Chemistry. 2011; 1(3): 617-623.
- 69.Janmeda P, et al. Chemopreventive effect of hydroethanolic extract of Euphorbia neriifolia leaves against DENA-induced renal carcinogenesis in mice . Asian Pac J Cancer Prev. 2011;12(3):677‐683. [PubMed] [Google Scholar]
- 70.Divakar K, et al. Protective effect of the hydro-alcoholic extract of Rubia cordifolia roots against ethylene glycol induced urolithiasis in rats . Food Chem Toxicol. 2010;48(4):1013‐1018. [DOI] [PubMed] [Google Scholar]
- 71.Bhuvaneswari P, Krishnakumari S. Nephroprotective effects of ethanolic extract of Sesamum indicum seeds (Linn.) in streptozotocin induced diabetic male albino rats. International Journal of Green Pharmacy (IJGP. 2012;6(4); 330-335. [Google Scholar]
- 72.Khan RA, Khan MR, Sahreen S. Evaluation of Launaea procumbens use in renal disorders: a rat model . J Ethnopharmacol. 2010;128(2):452‐461. [DOI] [PubMed] [Google Scholar]
- 73.Diwan V, Brown L, Gobe GC. The flavonoid rutin improves kidney and heart structure and function in an adenine-induced rat model of chronic kidney disease . J Funct Foods. 2017;33:85‐93. [Google Scholar]
- 74.Kataoka S, Norikura T, Sato S. Maternal Green tea polyphenol intake during lactation attenuates kidney injury in high-fat-diet-fed male offspring programmed by maternal protein restriction in rats . J Nutr Biochem. 2018;56:99‐108. [DOI] [PubMed] [Google Scholar]
- 75.Khan IA, et al. Evaluation of rhubarb supplementation in stages 3 and 4 of chronic kidney disease: a randomized clinical Trial . Int J Chronic Dis. 2014;2014: 1-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Ji S, Li L, Ji D. Effects of baoshen pill in treating chronic renal failure with long-term hemodialysis . Zhongguo Zhong xi yi jie he za zhi Zhongguo Zhongxiyi jiehe zazhi= Chinese journal of integrated traditional and Western medicine. 1993;13(2):71‐73, 67. [PubMed] [Google Scholar]
- 77.LI LS. Rhubarb in preventing progression of chronic renal disease . Nephrology. 1996;2(1):s146‐s150. [Google Scholar]
- 78.Sheng Z, Li N, Ge X. Clinical study of baoyuan dahuang decoction in the treatment of chronic renal failure . Zhongguo Zhong xi yi jie he za zhi Zhongguo Zhongxiyi jiehe zazhi= Chinese journal of integrated traditional and Western medicine. 1994;14(5):268‐270, 259. [PubMed] [Google Scholar]
- 79.Ahmed MS, et al. Treatment of idiopathic membranous nephropathy with the herb Astragalus membranaceus . Am J Kidney Dis. 2007;50(6):1028‐1032. [DOI] [PubMed] [Google Scholar]
- 80.Ma R, et al. Combination of Tripterygium wilfordii Hook F and angiotensin receptor blocker synergistically reduces excretion of urinary podocytes in patients with type 2 diabetic kidney disease . Biotechnology & Biotechnological Equipment. 2015;29(1):139‐146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Turki K, et al. Grape seed powder improves renal failure of chronic kidney disease patients . EXCLI J. 2016;15:424‐433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Zhang Z, et al. Effect of Cordyceps sinensis on renal function of patients with chronic allograft nephropathy . Urol Int. 2011;86(3):298‐301. [DOI] [PubMed] [Google Scholar]
- 83.Farese S, Kruse A, Pasch A, et al. Glycyrrhetinic acid food supplementation lowers serum potassium concentration in chronic hemodialysis patients . Kidney Int. 2009;76(8):877‐884. [DOI] [PubMed] [Google Scholar]
- 84.Dietzmann J, et al. Thiol-inducing and immunoregulatory effects of flavonoids in peripheral blood mononuclear cells from patients with end-stage diabetic nephropathy . Free Radical Biol Med. 2002;33(10):1347‐1354. [DOI] [PubMed] [Google Scholar]
- 85.Yarnell E, Abascal K. Herbs for relieving chronic renal failure . Altern Complement Ther. 2007;13(1):18‐23. [Google Scholar]
- 86.Kemmner S, et al. Dietary nitrate load lowers blood pressure and renal resistive index in patients with chronic kidney disease: a pilot study . Nitric Oxide. 2017;64:7‐15. [DOI] [PubMed] [Google Scholar]
- 87.Dai P, et al. Renoprotective effects of berberine as adjuvant therapy for hypertensive patients with type 2 diabetes mellitus: evaluation via biochemical markers and color Doppler ultrasonography . Exp Ther Med. 2015;10(3):869‐876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Jiménez-Osorio AS, Jimenez OAS, Niño WRG, Reyes SG, et al. The effect of dietary supplementation with curcumin on redox status and Nrf2 activation in patients with nondiabetic or diabetic proteinuric chronic kidney disease: a pilot study . J Ren Nutr. 2016;26(4):237‐244. [DOI] [PubMed] [Google Scholar]
- 89.Shelmadine BD, et al. A Pilot Study to Examine the Effects of an Anti-inflammatory Supplement on Eicosanoid Derivatives in Patients with Chronic Kidney Disease . J Altern Complement Med. 2017;23(8):632‐638. [DOI] [PubMed] [Google Scholar]
- 90.Yoshikawa N, et al. A prospective controlled study of sairei-to in childhood IgA nephropathy with focal/minimal mesangial proliferation. Japanese Pediatric IgA Nephropathy Treatment Study Group . Nihon Jinzo Gakkai Shi. 1997;39(5):503‐506. [PubMed] [Google Scholar]
- 91.Wang A, Yang M, Liu J. Molecular phylogeny, recent radiation and evolution of gross morphology of the rhubarb genus Rheum (Polygonaceae) inferred from chloroplast DNA trn LF sequences . Ann Bot. 2005;96(3):489‐498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Clementi EM, Misiti F. Potential health benefits of rhubarb . In: Bioactive Foods in Promoting Health. Elsevier; 2010:407–423. [Google Scholar]
- 93.Yokozawa T, et al. Effect of orally administered rhubarb extract in rats with chronic renal failure . Chem Pharm Bull. 1984;32(11):4506‐4513. [DOI] [PubMed] [Google Scholar]
- 94.Zhang L, et al. The pharmacokinetic study on the mechanism of toxicity attenuation of rhubarb total free anthraquinone oral colon-specific drug delivery system . Fitoterapia. 2015;104:86–96. [DOI] [PubMed] [Google Scholar]
- 95.Zhang Z-H Zhang ZH, Vaziri ND, Wei F, Cheng XL, Bai X, Zhao YY. et al. An integrated lipidomics and metabolomics reveal nephroprotective effect and biochemical mechanism of Rheum officinale in chronic renal failure . Sci Rep. 2016;6(1):1‐18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Duan S, et al. The wnt/β-catenin signaling pathway participates in rhein ameliorating kidney injury in DN mice . Mol Cell Biochem. 2016;411(1–2):73‐82. [DOI] [PubMed] [Google Scholar]
- 97.Chen W, et al. Rhein promotes the expression of SIRT1 in kidney tissues of type 2 diabetic rat . Xi bao yu fen zi mian yi xue za zhi= Chinese journal of cellular and molecular immunology. 2015;31(5):615‐619. [PubMed] [Google Scholar]
- 98.Peng L, et al. Rhein inhibits integrin-linked kinase expression and regulates matrix metalloproteinase-9/tissue inhibitor of metalloproteinase-1 ratio in high glucose-induced epithelial-mesenchymal transition of renal tubular cell . Biol Pharm Bull. 2012;35(10):1676‐1685. [DOI] [PubMed] [Google Scholar]
- 99.Gao Q, et al. Rhein improves renal lesion and ameliorates dyslipidemia in db/db mice with diabetic nephropathy . Planta Med. 2010;76(01):27‐33. [DOI] [PubMed] [Google Scholar]
- 100.Zheng JM, et al. Rhein reverses the diabetic phenotype of mesangial cells over-expressing the glucose transporter (GLUT1) by inhibiting the hexosamine pathway . Br J Pharmacol. 2008;153(7):1456‐1464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Zhu XL, et al. Suppression of lipopolysaccharide-induced upregulation of toll-like receptor 4 by emodin in mouse proximal tubular epithelial cells . Mol Med Rep. 2012;6(3):493‐500. [DOI] [PubMed] [Google Scholar]
- 102.Gao J, et al. Emodin suppresses hyperglycemia-induced proliferation and fibronectin expression in mesangial cells via inhibiting cFLIP . PLoS One. 2014;9(4):e93588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Yang J, et al. Emodin attenuates high glucose-induced TGF-β1 and fibronectin expression in mesangial cells through inhibition of NF-κB pathway . Exp Cell Res. 2013;319(20):3182‐3189. [DOI] [PubMed] [Google Scholar]
- 104.Li X, et al. Emodin suppresses cell proliferation and fibronectin expression via p38MAPK pathway in rat mesangial cells cultured under high glucose . Mol Cell Endocrinol. 2009;307(1–2):157‐162. [DOI] [PubMed] [Google Scholar]
- 105.Zhang W, et al. Emodin inhibits the differentiation and maturation of dendritic cells and increases the production of regulatory T cells . Int J Mol Med. 2012;29(2):159‐164. [DOI] [PubMed] [Google Scholar]
- 106.Zhang G, El Nahas A, Nahas EA. The effect of rhubarb extract on experimental renal fibrosis . Nephrol Dial Transplant. 1996;11(1):186‐190. [PubMed] [Google Scholar]
- 107.Khan IA, et al. Comparative evaluation of efficacy and safety profile of rhubarb and α-keto analogs of essential amino acids supplementation in patients with diabetic nephropathy . Saudi J Kidney Dis Transpl. 2016;27(4):710. [DOI] [PubMed] [Google Scholar]
- 108.Dong X, et al. Emodin: a review of its pharmacology, toxicity and pharmacokinetics. Phytother Res. 2016;30(8):1207‐1218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Christenhusz MJ, Byng JW. The number of known plants species in the world and its annual increase . Phytotaxa. 2016;261(3):201‐217. [Google Scholar]
- 110.Sun L, et al. Astragaloside IV prevents damage to human mesangial cells through the inhibition of the NADPH oxidase/ROS/Akt/NF-κB pathway under high glucose conditions . Int J Mol Med. 2014;34(1):167‐176. [DOI] [PubMed] [Google Scholar]
- 111.Nalbantsoy A, et al. Evaluation of the immunomodulatory properties in mice and in vitro anti-inflammatory activity of cycloartane type saponins from Astragalus species . J Ethnopharmacol. 2012;139(2):574‐581. [DOI] [PubMed] [Google Scholar]
- 112.Wojcikowski K, et al. An in vitro investigation of herbs traditionally used for kidney and urinary system disorders: potential therapeutic and toxic effects . Nephrology. 2009;14(1):70‐79. [DOI] [PubMed] [Google Scholar]
- 113.Ji C, et al. Effect of astragaloside IV on indoxyl sulfate-induced kidney injury in mice via attenuation of oxidative stress . BMC Pharmacology and Toxicology. 2018;19(1):1‐9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Zhang J, et al. Astragaloside IV derived from Astragalus membranaceus: a research review on the pharmacological effects . Adv Pharmacol. 2020;87:89‐112. [DOI] [PubMed] [Google Scholar]
- 115.Nie Y, et al. Effects of astragalus injection on the TGFβ/Smad pathway in the kidney in type 2 diabetic mice . BMC Complement Altern Med. 2014;14(1):1‐8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Zuo C, et al. Astragalus mongholicus ameliorates renal fibrosis by modulating HGF and TGF-β in rats with unilateral ureteral obstruction . Journal of Zhejiang University Science B. 2009;10(5):380‐390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Meng L, et al. Altered expression of genes profiles modulated by a combination of Astragali Radix and Angelicae Sinensis Radix in obstructed rat kidney . Planta Med. 2010;76(13):1431‐1438. [DOI] [PubMed] [Google Scholar]
- 118.Lu P-H, Lu PH, et al. An Apriori Algorithm-Based Association Rule Analysis to Identify Herb Combinations for Treating Uremic Pruritus Using Chinese Herbal Bath Therapy . Evidence-Based Complementary Altern Med. 2020;2020: 1-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Wojcikowski K, Wohlmuth H, Johnson DW, Gobe. et al. Effect of Astragalus membranaceus and Angelica sinensis combined with Enalapril in rats with obstructive uropathy . Phytother Res. 2010;24(6):875‐884. [DOI] [PubMed] [Google Scholar]
- 120.Zhang HW, et al. Astragalus (a traditional Chinese medicine) for treating chronic kidney disease . Cochrane Database Syst Rev. 2014;10: 1-63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Pao L-H Pao LH, Hu OYP, Fan HY, Lin CC, Liu LC, Huang PW. et al. Herb-drug interaction of 50 Chinese herbal medicines on CYP3A4 activity in vitro and in vivo . Am J Chin Med. 2012;40(01):57‐73. [DOI] [PubMed] [Google Scholar]
- 122.Zhu J-S, Zhu JS, Halpern GM, Jones K. The scientific rediscovery of a precious ancient Chinese herbal regimen: cordyceps sinensis Part II . The Journal of Alternative and Complementary Medicine. 1998;4(4):429‐457. [DOI] [PubMed] [Google Scholar]
- 123.Tuli HS, Sandhu SS, Sharma A. Pharmacological and therapeutic potential of Cordyceps with special reference to Cordycepin . 3 Biotech. 2014;4(1):1‐12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Zhang X, Shi L, Hu J. Research progress of Cordyceps chemical constituents and pharmacological functions . Zhong Yao Cai. 2000;23:722‐724. [Google Scholar]
- 125.Li SP, et al. A polysaccharide isolated from Cordyceps sinensis, a traditional Chinese medicine, protects PC12 cells against hydrogen peroxide-induced injury . Life Sci. 2003;73(19):2503‐2513. [DOI] [PubMed] [Google Scholar]
- 126.Yang LY, et al. H1-A extracted from Cordyceps sinensis suppresses the proliferation of human mesangial cells and promotes apoptosis, probably by inhibiting the tyrosine phosphorylation of Bcl-2 and Bcl-XL . J Lab Clin Med. 2003;141(1):74‐83. [DOI] [PubMed] [Google Scholar]
- 127.Huang YS, et al. Cordyceps cicadae Prevents Renal Tubular Epithelial Cell Apoptosis by Regulating the SIRT1/p53 Pathway in Hypertensive Renal Injury . Evidence-Based Complementary Altern Med. 2020;2020: 1-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Koh JH, et al. Activation of macrophages and the intestinal immune system by an orally administered decoction from cultured mycelia of Cordyceps sinensis . Biosci, Biotechnol, Biochem. 2002;66(2):407‐411. [DOI] [PubMed] [Google Scholar]
- 129.Liu X, Zhong F, Tang XI, et al. Cordyceps sinensis protects against liver and heart injuries in a rat model of chronic kidney disease: a metabolomic analysis . Acta Pharmacol Sin. 2014;35(5):697‐706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Zhang M, et al. Effect of Cordyceps sinensis powder on renal oxidative stress and mitochondria functions in 5/6 nephrectomized rats . Zhongguo Zhong xi yi jie he za zhi Zhongguo Zhongxiyi jiehe zazhi= Chinese journal of integrated traditional and Western medicine. 2015;35(4):443‐449. [PubMed] [Google Scholar]
- 131.Zhong F, et al. 1H NMR spectroscopy analysis of metabolites in the kidneys provides new insight into pathophysiological mechanisms: applications for treatment with Cordyceps sinensis . Nephrol Dial Transplant. 2012;27(2):556‐565. [DOI] [PubMed] [Google Scholar]
- 132.Zhang HW, et al. Cordyceps sinensis (a traditional Chinese medicine) for treating chronic kidney disease . Cochrane Database Syst Rev. 2014;12: 1-50 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Lan L. Study on effect of Cordyceps sinensis and artemisinin in preventing recurrence of lupus nephritis . Chinese Journal of Integrated Traditional and Western Medicine. 2002;8(2):89‐89. [PubMed] [Google Scholar]
- 134.Sun M, et al. Clinical study on application of bailing capsule after renal transplantation . Zhongguo Zhong xi yi jie he za zhi Zhongguo Zhongxiyi jiehe zazhi= Chinese journal of integrated traditional and Western medicine. 2004;24(9):808‐810. [PubMed] [Google Scholar]
- 135.Zhang Z, Zhang W, Yao K. Treatment of chronic allograft nephropathy with combination of enalapril and bailing capsule . Zhongguo Zhong xi yi jie he za zhi Zhongguo Zhongxiyi jiehe zazhi= Chinese journal of integrated traditional and Western medicine. 2008;28(9):806‐809. [PubMed] [Google Scholar]
- 136.Shao G. Treatment of hyperlipidemia with cultivated Cordyceps--a double-blind, randomized placebo control trial . Zhong xi yi jie he za zhi= Chinese Journal of Modern Developments in Traditional Medicine. 1985;5(11):652‐654. 642. [PubMed] [Google Scholar]
- 137.Hong T, Zhang M, Fan J. Cordyceps sinensis (a traditional Chinese medicine) for kidney transplant recipients . Cochrane Database Syst Rev. 2015;10(8): 1172-1178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Brinker AM, Maa J, Lipsky PE, Raskin I. et al. Medicinal chemistry and pharmacology of genus Tripterygium (Celastraceae) . Phytochemistry. 2007;68(6):732‐766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Ma J, et al. Anti-inflammatory and immunosuppressive compounds from Tripterygium wilfordii . Phytochemistry. 2007;68(8):1172‐1178. [DOI] [PubMed] [Google Scholar]
- 140.Li L. Clinical study of Tripterygium wilfordii Hook in treating glomerulonephritis . Chin J Intern Med. 1981;20:216‐220. [PubMed] [Google Scholar]
- 141.Wan Y. Mechanism of protective effects of effective components in Ttipterygium wilfordii hook. f. on glomerulonephritis . J Clin Exp Med. 2003;207:285‐288. [Google Scholar]
- 142.Zhou ZL, Yang YX, Ding J, Li YC, Miao. et al. Triptolide: structural modifications, structure–activity relationships, bioactivities, clinical development and mechanisms. Nat Prod Rep. 2012;29(4):457‐475. [DOI] [PubMed] [Google Scholar]
- 143.Wan YG, et al. Multi-glycoside of Tripterygium wilfordii Hook. f. reduces proteinuria through improving podocyte slit diaphragm dysfunction in anti-Thy1. 1 glomerulonephritis. J Ethnopharmacol. 2011;136(2):322‐333. [DOI] [PubMed] [Google Scholar]
- 144.Wan YG, et al. Contrasting dose–effects of multi-glycoside of Tripterygium wilfordii HOOK. f. on glomerular inflammation and hepatic damage in two types of anti-Thy1. 1 glomerulonephritis. J Pharmacol Sci. 2012;118(4):433‐446. [DOI] [PubMed] [Google Scholar]
- 145.Ma R, Zhao N, Zhang W. The effects and mechanism of Tripterygium wilfordii Hook F combination with irbesartan on urinary podocyte excretion in diabetic nephropathy patients . Zhonghua Nei Ke Za Zhi. 2013;52(6):469‐473. [PubMed] [Google Scholar]
- 146.Ji S-M, Ji SM, et al. Clinical trial of tripterygium wilfordii hook F. in human kidney transplantation in China. In: Transplantation Proceedings. Elsevier; 2006; 38(5): 1174-1279. [DOI] [PubMed] [Google Scholar]
- 147.Chou WC, et al. Hypovolemic shock and mortality after ingestion of Tripterygium wilfordii hook F.: a case report . Int J Cardiol. 1995;49(2):173‐177. [DOI] [PubMed] [Google Scholar]
- 148.Zhou J, Zhu Q, Yang X. Clinical observation of side effects of Tripterygium preparation . Zhongguo Zhong xi yi jie he za zhi Zhongguo Zhongxiyi jiehe zazhi= Chinese journal of integrated traditional and Western medicine. 1999;19(2):77‐79. [PubMed] [Google Scholar]
- 149.Chen P, Wan Y, Wang C, et al. Mechanisms and effects of Abelmoschus manihot preparations in treating chronic kidney disease . Zhongguo Zhong yao za zhi= Zhongguo zhongyao zazhi= China journal of Chinese materia medica. 2012;37(15):2252‐2256. [PubMed] [Google Scholar]
- 150.Zhang L, Li P, Xing CY, et al. Efficacy and safety of Abelmoschus manihot for primary glomerular disease: a prospective, multicenter randomized controlled clinical trial . Am J Kidney Dis. 2014;64(1):57‐65. [DOI] [PubMed] [Google Scholar]
- 151.Wen Y, et al. Chinese medicine for diabetic kidney disease in China . Nephrology. 2017;22(s4):50‐55. [DOI] [PubMed] [Google Scholar]
- 152.Chen Y, et al. Treatment of chronic kidney disease using a traditional Chinese medicine, Flos Abelmoschus manihot (Linnaeus) Medicus (Malvaceae) . Clin Exp Pharmacol Physiol. 2016;43(2):145‐148. [DOI] [PubMed] [Google Scholar]
- 153.Tu Y, et al. Huangkui capsule, an extract from Abelmoschus manihot (L.) medic, ameliorates Adriamycin-induced renal inflammation and glomerular injury via inhibiting p38MAPK signaling pathway activity in rats. J Ethnopharmacol. 2013;147(2):311‐320. [DOI] [PubMed] [Google Scholar]
- 154.Zhao Q, et al. Effects of huangkui capsule on renal inflammatory injury by intervening p38MAPK signaling pathway in rats with Adriamycin-induced nephropathy . Zhongguo Zhong yao za zhi= Zhongguo zhongyao zazhi= China journal of Chinese materia medica. 2012;37(19):2926‐2934. [PubMed] [Google Scholar]
- 155.Mao ZM, et al. Huangkui capsule attenuates renal fibrosis in diabetic nephropathy rats through regulating oxidative stress and p38MAPK/Akt pathways, compared to α-lipoic acid . J Ethnopharmacol. 2015;173:256‐265. [DOI] [PubMed] [Google Scholar]
- 156.Sun Q, et al. Effect of Huangshukuihua (Flos Abelmoschi Manihot) on diabetic nephropathy: a meta-analysis . J Tradit Chin Med. 2015;35(1):15‐20. [DOI] [PubMed] [Google Scholar]
- 157.Yan CS, et al. Salvia miltiorrhiza: traditional medicinal uses, chemistry, and pharmacology. Chin J Nat Med. 2015;13(3):163‐182. [DOI] [PubMed] [Google Scholar]
- 158.Ji XY, Tan BKH, Huang SH, et al. Effects of Salvia miltiorrhiza after acute myocardial infarction in rats . Novel compounds from natural products in the new millennium. 2004:183‐195. [Google Scholar]
- 159.Pang H, et al. Chemical analysis of the herbal medicine Salviae miltiorrhizae Radix et Rhizoma (Danshen) . Molecules. 2016;21(1):51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Jiang C, et al. Tanshinone IIA protects against folic acid-induced acute kidney injury . Am J Chin Med. 2016;44(04):737‐753. [DOI] [PubMed] [Google Scholar]
- 161.An L, et al. Salvia miltiorrhiza lipophilic fraction attenuates oxidative stress in diabetic nephropathy through activation of nuclear factor erythroid 2-related factor 2 . Am J Chin Med. 2017;45(07):1441‐1457. [DOI] [PubMed] [Google Scholar]
- 162.Lee SH, et al. The protective effect of Salvia miltiorrhiza in an animal model of early experimentally induced diabetic nephropathy . J Ethnopharmacol. 2011;137(3):1409‐1414. [DOI] [PubMed] [Google Scholar]
- 163.You Z, et al. Protective effect of Salvia miltiorrhizae injection on N (G)-nitro-D-arginine induced nitric oxide deficient and oxidative damage in rat kidney . Exp Toxicol Pathol. 2012;64(5):453‐458. [DOI] [PubMed] [Google Scholar]
- 164.Wang YJ, et al. Optimized project of traditional Chinese medicine in treating chronic kidney disease stage 3: a multicenter double-blinded randomized controlled trial . J Ethnopharmacol. 2012;139(3):757‐764. [DOI] [PubMed] [Google Scholar]
- 165.This P, Lacombe T, Thomas MR. Historical origins and genetic diversity of wine grapes . Trends Genet. 2006;22(9):511‐519. [DOI] [PubMed] [Google Scholar]
- 166.Khanal R, Howard L, Prior R. Procyanidin content of grape seed and pomace, and total anthocyanin content of grape pomace as affected by extrusion processing . J Food Sci. 2009;74(6):H174‐H182. [DOI] [PubMed] [Google Scholar]
- 167.Almomen SM, et al. Daily intake of grape powder prevents the progression of kidney disease in obese type 2 diabetic ZSF1 rats . Nutrients. 2017;9(4):345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Li SG, et al. Grape seed proanthocyanidin extract alleviates arsenic-induced oxidative reproductive toxicity in male mice . Biomed Environ Sci. 2015;28(4):272‐280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Chen Q, et al. The protective effect of grape seed procyanidin extract against cadmium-induced renal oxidative damage in mice . Environ Toxicol Pharmacol. 2013;36(3):759‐768. [DOI] [PubMed] [Google Scholar]
- 170.Gao Z, et al. Grape seed proanthocyanidin extract protects from cisplatin-induced nephrotoxicity by inhibiting endoplasmic reticulum stress-induced apoptosis . Mol Med Rep. 2014;9(3):801‐807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Ulusoy S, Ozkan G, Yucesan FB, et al. Anti-apoptotic and anti-oxidant effects of grape seed proanthocyanidin extract in preventing cyclosporine A-induced nephropathy . Nephrology. 2012;17(4):372‐379. [DOI] [PubMed] [Google Scholar]
- 172.Li Y, et al. Preventive and protective properties of Zingiber officinale (ginger) in diabetes mellitus, diabetic complications, and associated lipid and other metabolic disorders: a brief review . Evidence-Based Complementary Altern Med. 2012;2012: 1-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Xu Y, et al. 6-Shogaol ameliorates diabetic nephropathy through anti-inflammatory, hyperlipidemic, anti-oxidative activity in db/db mice . Biomed Pharmacother. 2018;97:633‐641. [DOI] [PubMed] [Google Scholar]
- 174.Atashak S, et al. Effects of ginger (Zingiber officinale Roscoe) supplementation and resistance training on some blood oxidative stress markers in obese men . Journal of Exercise Science & Fitness. 2014;12(1):26‐30. [Google Scholar]
- 175.Ramudu SK, et al. Nephro-protective effects of a ginger extract on cytosolic and mitochondrial enzymes against streptozotocin (STZ)-induced diabetic complications in rats . Chin J Physiol. 2011;54(2):79‐86. [DOI] [PubMed] [Google Scholar]
- 176.Abdulsalam K, Alkalifa A. Effect of Ginger and its Extract on Blood Sugar and on Kidney Function of Type I Diabetic Rats . Middle East Journal of Family Medicine. 2016;7(10):12. [Google Scholar]
- 177.Al Hroob AM, Hroob AMA, et al. Ginger alleviates hyperglycemia-induced oxidative stress, inflammation and apoptosis and protects rats against diabetic nephropathy . Biomed Pharmacother. 2018;106:381‐389. [DOI] [PubMed] [Google Scholar]
- 178.Imani H, et al. Effects of ginger on serum glucose, advanced glycation end products, and inflammation in peritoneal dialysis patients . Nutrition. 2015;31(5):703‐707. [DOI] [PubMed] [Google Scholar]
- 179.Panchal SK, et al. Rutin attenuates metabolic changes, nonalcoholic steatohepatitis, and cardiovascular remodeling in high-carbohydrate, high-fat diet-fed rats . J Nutr. 2011;141(6):1062‐1069. [DOI] [PubMed] [Google Scholar]
- 180.Korkmaz A, Kolankaya D. Inhibiting inducible nitric oxide synthase with rutin reduces renal ischemia/reperfusion injury . Can J Surg. 2013;56(1):6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Kamel KM, et al. Hesperidin and rutin, antioxidant citrus flavonoids, attenuate cisplatin-induced nephrotoxicity in rats . J Biochem Mol Toxicol. 2014;28(7):312‐319. [DOI] [PubMed] [Google Scholar]
- 182.Hao H, Shao Z, Tang D, et al. Preventive effects of rutin on the development of experimental diabetic nephropathy in rats . Life Sci. 2012;91(19–20):959‐967. [DOI] [PubMed] [Google Scholar]
- 183.Hsieh CL, et al. Rutin (quercetin rutinoside) induced protein-energy malnutrition in chronic kidney disease, but quercetin acted beneficially . J Agric Food Chem. 2013;61(30):7258‐7267. [DOI] [PubMed] [Google Scholar]
- 184.Turgut F, et al. Antioxidant and protective effects of silymarin on ischemia and reperfusion injury in the kidney tissues of rats . Int Urol Nephrol. 2008;40(2):453‐460. [DOI] [PubMed] [Google Scholar]
- 185.Desruelles J, Delmon A. Clinical trial of treatment of azotemic conditions with an injectable extract of Lespedeza capitata . Lille medical: journal de la Faculte de medecine et de pharmacie de l’Universite de Lille. 1969;14(2):83‐87. [PubMed] [Google Scholar]
- 186.Kemmner S, et al. P6. 9. Dietary nitrate by beetroot juice can lower renal resistive index in patients with chronic kidney disease. Artery Res. 2014;8(4):147‐148. [Google Scholar]
- 187.Retzl B, et al. Discovery of a Beetroot Protease Inhibitor to Identify and Classify Plant-Derived Cystine Knot Peptides . J Nat Prod. 2020;83(11):3305‐3314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Jan KN, Singh S. Stinging nettle (Urtica dioica L.): a reservoir of nutrition and bioactive components with great functional potential. Journal of Food Measurement and Characterization. 2017;11(2):423‐433. [Google Scholar]
- 189.Wojcikowski K, Johnson DW, Gobe G. Medicinal herbal extracts–renal friend or foe? Part one: The toxicities of medicinal herbs. Nephrology. 2004;9(5):313‐318. [DOI] [PubMed] [Google Scholar]
- 190.Jha V. Herbal medicines and chronic kidney disease . Nephrology. 2010;15:10‐17. [DOI] [PubMed] [Google Scholar]
- 191.Luyckx VA, Naicker S. Acute kidney injury associated with the use of traditional medicines . Nature Clinical Practice Nephrology. 2008;4(12):664‐671. [DOI] [PubMed] [Google Scholar]