You are on page 1of 23

RPS Pharmacy and Pharmacology Reports, 2023, 2, 1–23

https://doi.org/10.1093/rpsppr/rqad002
Advance access publication 18 January 2023
Review

Advance drug delivery and combinational drug approaches


for hepatoprotective action of berberine: a progressive
overview with underlying mechanism
Satish Sardana1, Rupa Gupta1, Kumud Madan2, , Dheeraj Bisht3, , Vijay Singh Rana4, ,
Samir Bhargava4, and Neeraj Kumar Sethiya4,*,

Downloaded from https://academic.oup.com/rpsppr/article/2/1/rqad002/6991356 by guest on 30 March 2023


1
Amity Institute of Pharmacy, Amity University Haryana, Gurugram, India
2
School of Pharmacy, Sharda University, Greater Noida, India
3
Department of Pharmaceutical Sciences, Sir J. C. Bose Technical Campus Bhimtal, Kumaun University, Nainital, India
4
Faculty of Pharmacy, DIT University, Dehradun, India
*
Correspondence: Neeraj Kumar Sethiya, Faculty of Pharmacy, DIT University, Mussoorie Diversion Road, Dehradun, Uttarakhand-248009, India.
Email: neeraj.sethiya@dituniversity.edu.in

Abstract
Objectives Berberine has attracted prominent interest recently due to its wide pharmacological actions in the management and treatment
of several diseases including the liver. However, restricted bioavailability and permeability make this drug as a better choice to develop sev-
eral value-added products for the improvement of both safety and efficacy. Much of researches has already been conducted in this direction
using several approaches to fix this issue. Therefore, the current article was designed to summarize all approaches taking together to enhance
hepatoprotection by berberine including molecular mechanism.
Methods Online scientific databases from PubMed were assessed for collecting information on berberine. All the collected information were
classified and incorporated into different sections such as recent progress of research on advance drug delivery systems and combinational ap-
proaches addressing the above issue for improvement of hepatoprotective action of berberine.
Key findings The electronically PubMed database search yielded 7454 articles from different countries in several languages. Out of them 270
articles published between 1932 and 2022 were included, corresponding to all detailed overviews on berberine including research pertaining to
toxicity and safety, biodistribution, pharmacokinetics, biopharmaceutics classification system, hepatoprotection against various hepatotoxicant
agent, advance drug delivery system, combinational drug approaches, clinical trial for hepatoprotection and patents. The review of the literature
reveals that berberine exhibits a potent hepatoprotective action with several molecular action mechanisms. Additionally, current trends of formu-
lation technology for enhancement of hepatoprotective action of berberine in terms of safety and efficacy are well co-related in present work.
Conclusion It was well established and concluded from the present work that both advance drug delivery system and combinational drug ap-
proaches may serve for enhancement of restricted hepatoprotective action of berberine due to poor bioavailability, solubility and permeability.
Additionally, berberine-based advanced delivery system including in combination with bioavailability and permeability enhancers may provide an
added advantage in the near future to meet the objectives.

Received: November 19, 2022. Editorial Acceptance: January 17, 2023


© The Author(s) 2023. Published by Oxford University Press on behalf of the Royal Pharmaceutical Society.
This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial License (https://creativecommons.org/
licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For
commercial re-use, please contact journals.permissions@oup.com
2 Satish Sardana et al.

Graphical Abstract

Downloaded from https://academic.oup.com/rpsppr/article/2/1/rqad002/6991356 by guest on 30 March 2023


Keywords: Berberine; liver; hepatoprotection; advanced drug delivery; drug combinations
Abbreviations: ACC, 1-aminocyclopropane-1-carboxylic acid; ACCα, acetyl-CoA carboxylase alpha; ACLY, ATP-citrate lyase; ACOX1, Acyl-CoA oxidase; Akt/
FoxO3a/Skp2, Akt kinase/Forkhead box O transcription factor/S-phase kinase-associated protein; ALT/AST, alanine and aspartate aminotransferase; AMPK,
activated protein kinase; AMPK-α, activated protein kinase catalytic α-subunit; AST, aspartate transaminase; ATF6/SREBP, activating transcription factor
6/ sterol regulatory element-binding proteins; Bcl-2, B-cell leukemia/lymphoma 2 protein; BCL2, B-cell lymphomagenesis; Ber, berberine; CCl4, carbon tetra
chloride; CCND1, cyclin D1 protein; CD147, cluster of differentiation 147; CD36, cluster of differentiation 36; CDKIs p21Cip1, cyclin-dependent kinase inhibitors;
CPT1α, carnitine palmitoyltransferase 1α; CXCR4, chemokine receptor type 4; ERK1/2, extracellular signal-regulated kinase ½; FASN, fatty acid synthase;
FBG, fasting blood glucose; FXR/SREBP-1c/FAS, farnesoid X-receptor/sterol regulatory element-binding protein-1c/ fatty acid synthetase; G4 PAMAM, poly-
amidoamine; G6P, glucose-6-phosphate dehydrogenase; GGT, γ-glutamyl transferase; HCC, hepatocellular carcinoma; HeLa cells, henrietta lacks cell line;
HepG2, human hepatoma cell line; HHL-5, human hepatocyte line 5; HMGB1/TLR4/NF-κB, high mobility group box 1/ toll-like receptor 4/ nuclear factor kappa
B; HNF4α, hepatocyte nuclear factor 4α; Huh7, human hepatoma-derived; HUVECs, human umbilical vein endothelial cells; IFG, impaired fasting glucose; IL,
interleukin; LDR, low-density lipoprotein; L-PK, L-type pyruvate kinase; LPS, lipopolysaccharide; LSI, lifestyle intervention; MCD, methionine–choline-deficient;
MMP-2, matrix metallopeptidase-2; mTOR, mammalian target of rapamycin; MTTP, microsomal triglyceride transfer protein; NAFLD, non-alcoholic fatty liver
disease; NASH, non-alcoholic steatohepatitis; NF-κB, nuclear factor kappa B; NLRP3, NLR family pyrin domains-containing protein-3; Nrf2/HO-1, Nuclear factor
erythroid 2-related factor/ heme oxygenase-1; Nrf2-Keap1, nuclear factor erythroid 2–related factor 2- Kelch-like ECH-associated protein 1; NUR77, nuclear
receptor 77; P2X7, purinergic type 2 receptor family (the second signal to inflammasome activation); P38MAPK, p38 mitogen-activated protein kinases; PANSS,
positive and negative syndrome scale; PCSK9, proprotein convertase subtilisin/kexin type 9 serine protease; PEPCK, phosphoenolpyruvate carboxykinase;
Pio, pioglitazone; PPAR, peroxisome proliferator-activated receptors; PPARα, peroxisome proliferator-activated receptor α; PPARγ, peroxisome proliferator-
activated receptor gamma; P-TEFb, positive transcription elongation factor b; RCT, randomised clinical trial; ROS, reactive oxygen species; SirT3, Sirtuin T3
gene; SREBP-1c, sterol regulatory element-binding protein-1c; T2DM, Type 2 diabetes mellitus; TBK1 and IKKɛ, TANK-binding kinase 1 & IκB kinases; THRSP,
thyroid hormone responsive protein; TLR4, toll-like receptor 4; TNF-α, tumour necrosis factor alpha; UCP2, uncoupling protein 2; VEGF, vascular endothelial
growth factor

Introduction Physically berberine (berberis species) is a yellow-coloured


Berberine is a plant-based compound characterized pigment having strong fluorescent characteristics under ul-
chemically as quaternary ammonium salt of cationic traviolet light. Due to its stronger yellow colour, berbe-
isoquinoline alkaloids group (2,3-methylenedioxy-9,10- rine and berberine-containing plants were widely utilized
dimethoxyprotoberberine chloride; C20H18NO4+) with a in the past for the purpose of dye in textile, wool, leather
molecular mass of 336.36122  g/mol. The compound was and wood industries.[5] The multifunctional nature of berbe-
isolated first time from Hydrastis canadensis (goldenseal) rine as a health-promoting agent was established therapeu-
in 1917.[1, 2] Subsequently, many of rhizomes, roots and tically due to diversified effects on several enzymes, many
barks of various plants are explored including Arcangelisia receptors and pathways related to cell signalling that have
flava, Argemone mexicana, Berberis aquifolium, B. aristata, been widely explored and reviewed in subsequent time inter-
B. vulgaris, B. darwinii, B. petiolaris, Coptis chinensis, vals.[6–22] Further, hepatoprotective activity of berberine have
C. japonica, C. teeta, Cortex rhellodendri, Eschscholzia been explored experimentally using various models including
californica, Hydrastis canadensis, Mahonia aquifolium, in vitro, in vivo, ex vivo and clinical trial with underlying
Phellodendron amurense, Rhizoma coptidis, Tinospora molecular mechanism reviewed in various time interval.[23–25]
cordifolia and Xanthorhiza simplicissima as a possible In brief, many of the review articles are focussed spe-
source for identification and separation of berberine majorly cially NAFLD,[26–32] liver fibrosis,[33] obesity and liver func-
from Berberideae, Panaveraceae, Ranunculaceae, Rutaceae tions.[34] Despite many therapeutic advances including
and Menispermaceae families.[3, 4] hepatoprotection berberine is also categorized under BCS III
Advance drug delivery and combinational drug approaches 3

drugs due to its poor oral bioavailability and intestinal ab- for the most part of studies. However, the potential risk of
sorption.[4] This poor bioavailability and intestinal absorp- clinically relevant pharmacological interaction is mainly lim-
tion issues limit the effective clinical application of berberine ited to cyclosporine and warfarin.[52, 53] Additionally, in a very
to translate therapeutic and health-promoting outcome.[14] In recent article biodistribution and pharmacokinetic profile of
this connection, several attempts have been made to the im- both berberine and its metabolites were discussed on liver
provement of oral bioavailability and intestinal absorption hepatocytes.[54]
via advance drug delivery system have been discussed and
reviewed by several authors.[20, 35–40] However, still there is no
any work pertaining to summarizes approaches such as com- Biodistribution, Pharmacokinetics and BCS
bination therapy, complexation, structure modification and Class of Berberine
recent advance drug delivery system for the improvement of As per the biopharmaceutical classification system (BCS), ber-
hepatoprotective action of berberine is present in the litera- berine has been placed as a class III drug.[55–57] As per reports,
ture. Therefore, the current study was designed to deliver the it was found to be sparingly soluble in water with poor intes-
recent progress of work established towards making berbe- tinal absorption and oral bioavailability. In fact, during the

Downloaded from https://academic.oup.com/rpsppr/article/2/1/rqad002/6991356 by guest on 30 March 2023


rine as suitable drug candidates for prevention and treatment study, the absolute bioavailability of berberine was found to
of liver-related disorder by incorporating several approaches. be <1%, and even absorbed berberine excreted back to the in-
Additionally, data related to toxicity profile, pharmacoki- testinal lumen via P-glycoprotein involvement from the gut.[10,
netics, molecular mechanism of hepatoprotection on various 58]
In brief, a study shown on a rat using noncompartmental
liver diseases, drug delivery system, clinical trial and recent model, where unbound berberine was transported to bile via
status of intellectual property right protection are summar- active transportation and which was further metabolized in
ized for better understanding. liver by action of P450 enzyme system through demethylation
in phase I and glucuronidation in the phase II, respectively.[59]
Additionally, four main glucuronide conjugates based berbe-
Methods rine metabolites were identified in rats, that is berberrubine,
The exhaustive search of the literature was accomplished demethyleneberberine, jatrorrhizine and thalifendine.[60]
with the information retrieved by using online electronic Subsequently, intestinal bacterial flora was found to play role
search on PubMed database, google and Indian Patent search in enterohepatic circulation of both berberine and its con-
using Berberine as the searching keyword. Publications does jugated metabolites.[58] On the contrary, only a very small
not fall under toxicity and safety, biodistribution, pharma- amount of unchanged form of berberine was found to be
cokinetics, biopharmaceutics classification system (BCS), eliminated by urines.[61] Further, berberine was found to be
hepatoprotection against various hepatotoxicant agent distributed into all major organs with the highest amount
focussing mechanism of action, advance drug delivery obtained in the liver and readily excreted via the urine after
system, combinational drug approaches and clinical trial for absorption.[62]
hepatoprotection were excluded from the study. The elec-
tronic PubMed database search yielded 7454 articles from
different countries in several languages. Out of the 270 ar- Berberine as Hepatoprotective Agent
ticles published between 1932 and 2022 were included. The Evidence on berberine exhibited hepatoprotective actions
major limitation of the present work is several studies are on various liver toxicity models by both in vitro and in vivo
published in other than the English language. Further, the along with postulated mechanism of action were summarized
present study was designed in such a way to accomplish our in Table 1. In brief liver protection by berberine on various
objective and should be not similar to the earlier published models including chemical/metal/drug-induced hepatotox-
review icity, parasite/microorganism infected liver damage, obesity/
fatty liver damage, hepatocellular carcinoma, hepatitis c virus
and ischemia/reperfusion injuries were incorporated. Among
Toxicity and Safety Profile these chemical/metal/drug-induced hepatotoxicity and
LD50 of pure berberine was reported to be 25 mg/kg in mice NAFLD were found to be the most studied models. Several
and considered to be safe with rare to mild adverse effects.[41] dose by several routes of administration has been incorpo-
However, some studies also claim the development of gastric rated to get the effect as depicted in Table 1. In this context,
lesions as associated side effects of berberine and its deriva- 5 mg/kg/b.w. was the minimum oral dose and 324 mg/kg/b.w.
tives.[42, 43] Such, gastrointestinal side effects including diar- was found to be the maximum dose of berberine to translate
rhoea and constipation may be associated with the delays of hepatoprotective effects. However, for intraperitoneal admin-
small intestinal transit time from experimental evidence.[44] istration minimum dosage was 0.4 mg/kg/b.w and the max-
Some of the notable interaction warrants the safety of the imum dose was 10 mg/kg/b.w, respectively. The brief details
berberine including berberine displaces bilirubin from al- of the studies are summarized herewith:
bumin,[45] thiopental, tolbutamide and warfarin, from their
protein binding sites to increase plasma levels[46] and increase Chemical/metal/drug-induced hepatotoxicity
the level of cyclosporine A.[47, 48] Some of the clinical studies Hepato-toxicants such as acetaminophen,[63, 64] arsenic,[65]
also indicated adverse effects such as transient elevation of carbon tetra chloride,[66–72] cisplatin,[73] cyclophosphamide,[74]
bilirubin level in serum,[49] distribution of sex-hormone syn- diethylnitrosamine,[75] doxorubicin,[76–78] ethanol,[79–81] ferrous
thesis pathway,[50] and suppression of both immune func- sulfate,[82] lead acetate,[83] methotrexate,[84–86] paraquat,[87] ra-
tions, that is cellular and humoral.[51] Overall, the safe use pamycin,[88] tert-butyl hydroperoxide,[89] streptozotocin,[90,
of berberine including a dosage ranges of 500–1000 mg/day 91]
thioacetamide,[92, 93] tunicamycin[94] and more than two
4 Satish Sardana et al.

Table 1 Hepatoprotective action of berberine and postulated molecular mechanism

Dose (kg/body Model Mechanism


weight/day)

CHEMICAL/METALS/DRUG INDUCED
Acetaminophen
5 mg i.p. Steatohepatitis and acute acetaminophen tox- Interferes with activation of the NLRP3 inflammasome pathway
icity via interference with P2X7 (a purinergic receptor responsible for
inflammasome activation). [63]
5 mg i.p. Acetaminophen-induced toxicity in mice Inhibition of hepatocyte necrosis, inflammatory response and oxidative
stress.[64]
Arsenic
10, 25 and 50 Arsenic-induced mitochondria toxicity on rat Reduced ROS generation, without restoring mitochondrial membrane

Downloaded from https://academic.oup.com/rpsppr/article/2/1/rqad002/6991356 by guest on 30 March 2023


μM liver. integrity.[65]
Carbon tetra chloride (CCl4)
120 mg oral CCl4-induced toxicity in rats Antioxidant action.[66]
80–120 mg oral CCl4-induced acute toxicity in rats Effective for both prevention and treatment of liver toxicity.[67]
5 and 10 mg i.p. CCl4-intoxicated mice Attenuate oxidative or nitrosative stress along with inflammatory re-
sponse inhibition.[68]
3 and 9 mg i.p. CCl4-intoxicated mice Liver fibrosis amelioration via suppression of hepatic oxidative stress
and fibrogenic potential followed by degradation stimulation of colla-
gen deposits by MMP-2.[69]
25 and 50 mg CCl4-induced liver fibrosis in mice AMPK activation and blocking of expression of Nox4 and Akt.[70]
oral
50 mg oral CCl4-induced NASH rats Ameliorated hyperlipidemia, oxidative stress, observed neurotoxicity,
inflammation, hyperglycemia and hyperinsulinemia.[71]
5, 10 and 15 mg CCl4-induced liver injury in rats Effectively regulating Nrf2-Keap1-antioxidant-responsive element-
oral related proteins and genes expression and p53 pathway-mediated hepa-
tocyte apoptosis inhibition.[72]
Cisplatin
0.4 mg i.p. Cisplatin-induced nephro- and hepato-toxicity Intensified enzymatic oxidant status and downregulate lipid
in rats peroxidation by decrease in TLR4 gene expression.[73]
Cyclophosphamide
50 mg oral Cyclophosphamide-induced hepato-toxicity in Exhibit anti-inflammatory and antioxidant action by alleviating ele-
rats vated serum marker enzymes.[74]
Diethylnitrosamine
200 mg oral Diethylnitrosamine-induced cirrhosis in rats Improve intestinal dysbacteriosis, responsible to reduce liver toxicity
induced by pharmacological or pathological intervention.[75]
Doxorubicin
60 mg/kg oral Doxorubicin-induced hepatotoxicity in mice Attenuated inflammatory cell infiltration, vascular congestion,
hepatocellular degeneration, necrosis and fibrosis.[76]
5–20 mg oral Doxorubicin-induced acute hepatorenal toxicity Liver protective effects.[77]
in rats
Various dose Doxorubicin-induced hepatotoxicity Exhibit antioxidant and immune response with low to moderate cyto-
chrome modulation.[78]
Ethanol
200 mg oral Ethyl alcohol-induced inflammation and lipid Attenuated vascular congestion, lipid synthesis (FASN, THRSP, ACC,
deposition in rats AMPK-α, ACLY), regulating uptake of fatty acids (CD36) and lipid
oxidation (CPT1α, PPARα, ACOX1).[79]
10, 50 and Ethanol induce liver disease in male mice Exhibit immunosuppressive response.[80]
100 mg oral (C57BL/6J)
200 and 300 mg Ethanol-induced oxidative stress and steatosis Restoring hepatocyte nuclear factor 4α/microsomal triglyceride trans-
oral in mice fer protein pathways, peroxisome proliferator-activated receptor, that
is α/peroxisome proliferator-activated receptor-gamma Co-activator-
1α.[81]
Ferrous sulfate (FeSO4)
10 mg oral FeSO4-induced hepatic and renal damages in Reduction in lipid peroxidation and ability to chelate iron.[82]
rats.
Lead acetate
50 mg oral Lead acetate induce toxicity in rats Inhibiting lipid peroxidation and enhancing antioxidant defenses.[83]
Methotrexate
Advance drug delivery and combinational drug approaches 5

Table 1 Continued

Dose (kg/body Model Mechanism


weight/day)

25 and 50 mg Methotrexate-induced liver injury Attenuated both oxidative stress and apoptosis, possibly via Nrf2/
oral HO-1 pathway and PPARγ upregulation.[84]
100 mg oral Methotrexate-induced liver toxicity in rats Ameliorative both oxidative stress and any of biochemical changes.[85]
50 mg oral Methotrexate induced liver toxicity in rats P38MAPK, Keap-1 and NF-κB inhibition. Further, reduced expression
of pro-apoptotic protein Bax and apoptotic protein caspase-3 with
increase in the expression of anti-apoptotic protein Bcl-2.[86]
Paraquat
5 mg oral Paraquat-induced toxicity in rat Significant decrease in ROS formation, cell death and LDH release.
Also inhibits cellular glutathione depletion and over all improve in liver
function enzyme level.[87]

Downloaded from https://academic.oup.com/rpsppr/article/2/1/rqad002/6991356 by guest on 30 March 2023


Rapamycin
62.5 µM Rapamycin-mediated human hepatoma cell Synergistically inhibiting the mTOR signalling pathway mediated
(HCC cells SMMC7721 and HepG2) death through CD147 with at least in part.[88]
Tert-butyl hydroperoxide
0.5 and 5 mg Tert-butyl hydroperoxide-induced oxidative Exhibit chemopreventive role via reducing oxidative stress in living
i.p. damage in liver of rat systems.[89]
Streptozotocin
75, 150 and Streptozotocin-type 2 diabetic rats liver Up-regulation of P-TEFb expression, antilipid peroxidation and antiox-
300 mg oral idant status.[90]
75, 150 and Streptozotocin-type 2 diabetic rats liver Metabolic-related PPARalpha/delta/gamma protein expression modu-
300 mg oral lation in liver.[91]
Thioacetamide
50 mg oral Thioacetamide injection in rats Treat liver fibrosis via antioxidant and anti-inflammatory action.[92]
10 mg i.p. Rat liver xenobiotic-metabolizing enzymes after Normalization of both cytochromes P450-dependent and flavin-
partial hepatectomy containing monooxygenases.[93]
Tunicamycin
75, 150 and Tunicamycin induced ER-stress liver injury mice Improves ER stress in hepatocytes and regulate gut microbiota in
300 mg oral (C57BL/6) mice.[94]
More than one
4 mg oral Acetaminophen or CCl4-induced toxicity in Selective therapeutic effect against acetaminophen.[95]
rodents
200 mg oral Thioacetamide (TAA) and carbon tetrachloride Inducing ferrous redox reaction to activate ROS-mediated HSC
(CCl4)-induced liver fibrogenesis in mouse ferroptosis.[96]
50, 100 or Rat liver fibrosis-induced by multiple hepato- Regulation of the both anti-oxidant system and lipid peroxidation.[97]
200 mg oral toxic factors.
PARASITE/MICROORGANISM
12 mg/kg oral Schistosoma mansoni-induced hepatic injury Ameliorate liver damage and oxidative stress conditions caused by
in mice schistosomiasis.[98]
HFD/OBESITY/FATTY LIVER
200 mg oral HFD-feds rats Partially counteract dysregulation of MTTP by reversing the methyla-
tion state, leading to reduce in hepatic fat content.[118]
187.5 mg oral HFD-fed NAFLD rats Improve insulin resistance by up-regulating both mRNA and protein
levels of IRS-2 (key molecule identified for insulin signaling path-
way).[99]
162 and 324 mg HFF-fed NAFLD rats Downregulate the expression of UCP2 proteins and UCP2 mRNA
oral levels in hepatic tissue.[100]
100 mg oral HFD-fed rats Improve mitochondrial SirT3 activity, normalizing mitochondrial func-
tion and energetic deficit state caused by impaired OXPHOS.[119]
200 mg oral HFD-fed steatotic animal Completely reversed the MRAK052686 and Nrf2 reduced expres-
sion.[122]
200 mg oral HFD-fed NAFLD rats Restore the expression of L-PK via demethylation and the increase in
histone H3 and H4 acetylation levels.[101]
200 mg oral HFD-feds mice Alleviates NASH and its predisposing factors. Normalization of gut
microbiota might underlie its effect.[120]
100 mg oral Obesity-associated NAFLD on mice Suppression of inflammation without involving AMPK.[102]
50 mg oral HFD-fed mice and oleate-palmitate-induced Inhibition of the ERK/mTOR pathway.[258]
lipotoxicity hepatocytes
6 Satish Sardana et al.

Table 1 Continued

Dose (kg/body Model Mechanism


weight/day)

200 mg oral MCD-fed NAFLD male mice (C57BL/6J) Involvement of ATF6/SREBP-1c pathway for reversing ER stress-
activated lipogenesis.[103]
150 mg oral HFD-fed NAFLD rats Restore the liver function and provide significant protection via ameli-
orating barrier function of intestine.[104]
50 or 100 mg HFD-fed blunt snout bream Megalobrama Attenuated liver damage via the protection for mitochondria.[259]
oral amblycephala
150 mg oral Wild type (WT) and intestine-specific FXR Inhibit BSH, elevate TCA and activate FXR, majorly involve in uptake
knockout (FXRint–/–) mice for Hepatic Lipid of long-chain fatty acids inhibition in the liver.[105]
Metabolism
10 µM and HFD-induced NAFLD rats and Huh7 cells Improve damage caused by oxidative stress.[260]

Downloaded from https://academic.oup.com/rpsppr/article/2/1/rqad002/6991356 by guest on 30 March 2023


200 mg oral
0.2 g oral HFDC-fed NASH mice CXCR4 signalling pathways inhibition via restoration of the balance of
α1-AT and NE levels.[123]
150 mg oral HFD-induced NAFLD rats Inhibiting glucogenesis via regulating lipid metabolism.[106]
100 mg oral C57BL/6 mice with AD (containing 1.25% cho- Liver protection under cholesterol overloading via autophagic flux
and 20 µg/ml lesterol and 0.5% cholic acid) and HCD (con- regulation including cholesterol metabolism and COX2-prostaglandin
taining 1.25% cholesterol) and HepG2 cells synthesis inhibition.[261]
5 mg i.p. HFHS-fed liver-specific SIRT1 knockout mice Mediating autophagy and FGF21 activation.[262]
and their wild-type littermates for hepatic ste-
atosis
200 mg oral HFD-fed NAFLD mice Ameliorate lipid accumulation and inflammation via reduction in LPS
production and release of inflammatory cytokines by hepatic macro-
phages.[107]
100 mg oral HFD-fed NAFLD rats Activation of the Nrf2/ARE signalling pathway.[108]
250 mg/kg oral STZ injection + HFHC for NASH-HCC mice Involvement of p38MAPK/ERK-COX2 pathway for inflammation and
model angiogenesis genes regulation.[124]
30 mg/ml HFD-fed NAFLD mice and patients Caused phosphorylation of SREBP-1c and AMPK. Further, attenuates
hepatic steatosis and reduces liver TG synthesis via AMPK-SREBP-1c-
SCD1 pathway activation.[109]
300 mg oral HFD-fed NAFLD mice Fatty acid β-OX partly inhibition via SIRT3-mediated LCAD
deacetylation caused by HFD.[110]
100 mg oral HFD-fed NAFLD rodents SIRT3/AMPK/ACC pathway activation to ameliorates, HFD-induced
hepatic steatosis.[111]
300 mg oral HFD-fed NAFLD rats Angptl2 pathway regulation.[112]
200 mg oral HFD-fed NAFLD rats Inhibit nuclear translocation of NF-κB via involvement of TLR4/
MyD88/NF-κB pathway.[113]
100 mg oral HFD-fed NAFLD rats Reverse the abnormal expression of LDLR and MTTP followed by
lipid synthesis inhibition.[114]
300 mg oral A rat model of NASH was established by a Restoration of Treg/Th17 ratio and regulation of chemerin/CMKLR1
high-fat diet in rats signalling pathway responsible for overall inflammation and lipid dep-
osition reduction in liver.[125]
0, 5, 25 and HFD-fed ApoE-/- for therosclerotic lesions and Improves lipid disorder, reduced aortic plaque formation and alleviated
50 µg/ml and hepatic steatosis hepatic lipid accumulation, mainly associated with PCSK9 down-
50–100 mg oral regulation through ERK1/2 pathway.[263]
50 mg oral HFD-fed NAFLD and NASH mouse Multiple genes expression modulation responsible for hepatic stellate
cell activation and cholangiocyte proliferation.[31]
50 mg/kg oral HFD-fed black sea bream (Acanthopagrus Reduced hepatic lipid accumulation by lipolysis gene expression
schlegelii) upregulation and lipogenesis gene expression downregulation followed
by improvement in muscle lipid contents.[115]
1.4 and 0.075 g HFD-fed NALFD male mice Repressed complex I in liver and gut, mainly responsible for lipid
oral metabolism inhibition (alleviates from both obesity and fatty
liver).[116]
5–10 mg i.p. HFHS-fed NAFLD mouse and a palmitate- Transcriptional regulation of SETD2 activity for hepatoprotection via
treated hepatocyte steatosis model were gener- steatosis.[117]
ated
300 mg oral HFD-fed mice Elevated HIF-2 expression, insulin resistance and disorder of lipid me-
tabolism.[121]
Advance drug delivery and combinational drug approaches 7

Table 1 Continued

Dose (kg/body Model Mechanism


weight/day)

HEPATOCELLULAR CARCINOMA (HCC)/OTHER CELL LINES


1–300 μM Tight-seal whole-cell patch-clamp techniques in Calcium and potassium channels inhibitory actions in isolated rat hep-
enzymatically isolated rat hepatocytes. atocytes.[264]
0–20 μg/ml Activated rat hepatic stellate cells (CFSCs) Hepatic stellate cell proliferation inhibition for liver fibrosis preven-
tion.[126]
0–96 mM and HCC- HepG2 cells and HepG2 human HCC Induction of p53 and Fas apoptotic system activity.[127]
40 or 80 mg i.p. xenograft mice
0.1, 5, 10, 15, HepG2 cells treated with high concentrations Effective against high-concentration leptin-induced NAFLD via up-
20, 25 and 30 of leptin. regulation of the mRNA expression of leptin receptor.[128]
μM/l

Downloaded from https://academic.oup.com/rpsppr/article/2/1/rqad002/6991356 by guest on 30 March 2023


0, 5, 10 and 15 Tumour-induced angiogenesis using HCC Prevents secretion of VEGF and down-regulates VEGF mRNA expres-
mM HUVECs sion.[139]
1–100 μM Primary hepatocytes were isolated from non- Transcription regulation on hepatic genes responsible for fatty acid and
fasted male SD glucose metabolism.[265]
0.8–50 μM HCC and SMMC-7721 cells. Both ROS-triggered caspase-dependent induction and caspase-
independent apoptosis pathways.[140]
10, 50 and 100 Human HCC-HepG2 AMPK activation in HepG2 cells followed by both apoptotic and
μM autophagic death induction.[129]
15 μM Human HCC-HepG2 Lipid-lowering effects by improvement in low-density lipoprotein re-
ceptor expression.[130]
200 µM and H22, HepG2 and Bel-7404 cells and H22 trans- Arachidonic acid metabolic pathway suppression.[131]
12.5, 25 and planted tumour model in mice HCC
50 mg oral
100 μM HCC HepG2 cell line Retarded cancer cell growth by regulating SP1 and miR-22-3p and its
downstream targets, that is BCL2 and CCND1, in HCC.[132]
25, 50, 100 and HCC cell lines Bel-7402 and SMMC-7721 HCC cell invasion and migration via uPA receptor inactivation by up-
200 μM regulation of PAI-1 and down-regulation of uPA.[141]
0, 10, 50 and HepG2 Involvement of NF-κB p65 pathway for apoptosis promotion.[133]
100 μM
0.01, 0.1 and NAFLD based on HepG2 cells induced by oleic Improvement on lipid metabolism disorder via FXR/SREBP-1c/FAS
1 μM acid pathway regulation.[134]
1–10 μM Steatosis using HepG2 hepatocytes Suppression of TNF-α expression and mitochondrial biogenesis induc-
tion to decreased cellular ROS.[135]
10 mM Immortalized MIHA hepatocyte cell line for Early Growth Response 1 (EGR1) level upregulation responsible for
hepatic steatosis miR-373 expression transactivation.[266]
0, 30, 60 and Cell cycle arrest in HCC Promotes CDKIs p21Cip1 and p27Kip1 expression via Akt/FoxO3a/
120 μM Skp2 axis regulation. Further induces G0/G1 phase cell cycle ar-
rest.[142]
200 mg oral ABCA1 in QSG-7701 hepatocytes and in Upregulation of ABCA1 protein levels via involvement of PKCδ to re-
NASH-induced mice duce the phosphorylation of serine residues in ABCA1.[267]
50 and 100 μM HepG2 cell lines Antiproliferative effect of berberine was due Akt, PHLPP2 and Mst1
involvement (an autoinhibitory triangle).[268]
5, 10 and 20μM L02 hepatocytes injury induced by D-GalN Inhibits inflammation and mitochondria-dependent apoptosis.[269]
(5mM)/TNF-α (100 ng/ml)
2.5 mmol/l FGF21 expression in primary mouse hepato- Induced AMPK activation responsible for hepatic FGF21 expression
cytes. via NUR77.[270]
0, 10, 20 and Radiation-induced oxidative stress and apopto- Strengthens radiosensitivity through Nrf2 signalling pathway suppres-
40 μM sis in Huh7, HHL-5 and HepG2 cells sion.[143]
2.68 mM HCC-HepG2 cells. Radiosensitizer towards treating liver cancer by blocking autophagy
and cell cycle arrest resulting in senescence.[136]
1, 5 and 25 μg/ FFA-induced steatosis on HepG2 cells Activate SIRT1-FoxO1-SREBP2 signal pathway.[137]
ml
50 µM HCC cells (Hep3B, HepG2), HEK293 and Antagonizes β-catenin pathway via β-catenin translation and mTOR
Huh7 cells activity inhibition.[138]
HEPATITIS C VIRUS
0, 2, 5, 10, 20, Cell culture-derived HCV viral pseudoparticles Targets viral E2 glycoprotein.[144]
50 and 100 μM bearing HCV glycoproteins for hepatitis C virus
entry-related assays
8 Satish Sardana et al.

Table 1 Continued

Dose (kg/body Model Mechanism


weight/day)

0, 1, 5, 10, 20, Human hepatoma cells harboring hepatitis C Autophagy inhibition irrespective of the HCV genome presence.[145]
40, 80, 100, 200 virus RNA
and 400 μM
ISCHEMIA/REPERFUSION (I/R)
100 mg oral IR hepatic injury after orthotopic liver trans- Promotes liver transplantation during I/R injury partly via Sirt1/
plantation (OLT) on rats FoxO3α-mediated autophagy activation.[146]
18.6 mM Preservation solution for rat model of ex vivo Preserves mitochondrial function and bioenergetics by protecting liver
liver transplant from toxic effects caused by I/R.[147]
100 mg oral Hepatic cold ischemia rat model Reducing apoptosis, possibly via PI3K/Akt/mTOR signalling pathway
modulation.[148]

Downloaded from https://academic.oup.com/rpsppr/article/2/1/rqad002/6991356 by guest on 30 March 2023


or multiple[95–97] were studied for hepatoprotective action of • ER stress improvement in hepatocytes and gut microbi-
berberine. Among these carbon, tetra chlorides induce liver ota regulation.
toxicity is one of the most studied model. Based on the data
the major postulated mechanism for hepatoprotection of ber-
berine are: Parasite/microorganism
In this model very few studies were conducted. One of the
• NLRP3 inflammasome pathway activation interference study in this context was conducted by inducing hepatic in-
(associated with the P2X7 activation), inflammatory cell jury in mice by using Schistosoma mansoni (a water-borne
infiltration and inflammation suppression via IL-10, IL- parasite). During this study berberine (12  mg/kg oral) was
6, LPS, TNF-α and ET involvement. found to ameliorate both liver damage and oxidative stress
• Inhibition of oxidative stress through ROS generation conditions produced due to schistosomiasis.[98]
and also participate by regulating antioxidant-responsive
element-related proteins and genes, that is Nrf2-Keap1- Obesity/fatty liver
expression. High-fat diet causing NAFLD,[99–117] obesity[118–121] and
• Inhibition of hepatocyte necrosis via p53 pathway- NASH[115, 122–125] were major protocol adopted for testing the
mediated hepatocyte apoptosis. effect of berberine through various mechanism. The minimum
• Improvement in fibrogenic potential via involvement of and maximum selected dose was found to be 30 to 300 mg/kg
MMP-2 responsible for stimulation of collagen deposits for oral administration and 5 to 10 mg/kg for intraperitoneal
degradation. administration, respectively. Additionally, the dose upto 50
• AMPK activation and blocking of expression of Nox4 µg/ml was tested for in vitro model studied. Animal such as
and Akt. rats, mice, black sea bream and blunt snout bream are ma-
• Reducing lipid peroxidation via TLR4 gene expression jorly used. Based on the data the major postulated mechanism
regulation. for hepatoprotection of berberine are:
• Attenuated vascular congestion, lipid synthesis (FASN,
THRSP, ACC, AMPK-α, ACLY), regulating uptake of • Reduced hepatic fat content by reversing HFD-elicited
fatty acids (CD36) and lipid oxidation (CPT1α, PPARα, dysregulation of MTTP.
ACOX1). • Protein and mRNA levels of IRS-2 upregulation leading
• Low to moderate cytochrome modulatory po- to improvement in insulin resistance.
tentials (Normalization of both flavin-containing • Down-regulation of both UCP2 proteins and UCP2
monooxygenases and cytochromes P450). mRNA expression levels in hepatic tissue.
• Restoring hepatocyte nuclear factor 4α/microsomal • Improvement in mitochondrial SirT3 activity followed
triglyceride transfer protein pathways, peroxisome by mitochondrial function normalization and energetic
proliferator-activated receptor, that is α/peroxisome deficit caused by impaired OXPHOS.
proliferator-activated receptor-gamma Co-activator- • Completely reverse the reduced Nrf2 and MRAK052686
1α. expression.
• Ability to chelate iron and Nrf2/HO-1 pathway • Restore the expression of L-PK via demethylation and
upregulation. the increase in histone H3 and H4 acetylation levels.
• Keap-1, P38MAPK and NF-κB inhibition. • Suppression of inflammation without involving AMPK.
• Reduced expression of apoptotic protein caspase-3 and • Inhibition of the ERK/mTOR pathway.
pro-apoptotic protein Bax followed by increased anti- • Involvement of ATF6/SREBP-1c pathway for reversing
apoptotic protein Bcl-2 expression. ER stress-activated lipogenesis.
• Significant decrease in LDH release. Inhibits cellular glu- • Ameliorating intestinal barrier function.
tathione depletion inhibiting the mTOR signalling path- • Restore liver function by mitochondria protection and
way mediated through CD147. normalization of gut microbiota.
• Up-regulation of P-TEFb expression. • Inhibit BSH, elevate TCA and activate FXR majorly in-
• PPARalpha/delta/gamma protein expression modulation volved in the uptake of long-chain fatty acids inhibition
in liver responsible for metabolism. in the liver.
Advance drug delivery and combinational drug approaches 9

• Progression of hepatic steatosis to steatohepatitis was • Hepatic stellate cell proliferation inhibition for liver fi-
downregulated via reduction of oxidative stress. brosis prevention.
• CXCR4 signalling pathways inhibition via restoration of • Induction of p53 and Fas apoptotic system activity.
the balance of α1-AT and NE levels. • Effective for high-concentration leptin-induced NAFLD
• Inhibiting glucogenesis via regulating lipid metabolism. via up-regulation of the mRNA expression of leptin re-
• Liver protection under cholesterol overloading via ceptor.
autophagic flux regulation including cholesterol metab- • Prevents secretion of VEGF and down-regulates VEGF
olism and COX2-prostaglandin synthesis inhibition. mRNA expression.
• Mediating autophagy and FGF21 activation. • Transcription regulation on hepatic genes responsible for
• Ameliorate lipid accumulation and inflammation via re- fatty acid and glucose metabolism.
duction in LPS production and release of inflammatory • Both ROS-triggered caspase-dependent induction and
cytokines by hepatic macrophages. caspase-independent apoptosis pathways.
• Activation of the Nrf2/ARE signalling pathway. • AMPK activation in HepG2 cells followed by both apop-
• Involvement of p38MAPK/ERK-COX2 pathway for in- totic and autophagic death induction.

Downloaded from https://academic.oup.com/rpsppr/article/2/1/rqad002/6991356 by guest on 30 March 2023


flammation and angiogenesis genes regulation. • Lipid-lowering effects by improvement in low-density
• Caused phosphorylation of SREBP-1c and AMPK. Further, lipoprotein receptor expression.
attenuates hepatic steatosis and reduces liver TG synthesis • Arachidonic acid metabolic pathway suppression.
via AMPK-SREBP-1c-SCD1 pathway activation. • Retarded cancer cell growth by regulating SP1 and miR-
• Fatty acid β-OX partly inhibition via SIRT3-mediated 22-3p and its downstream targets, that is BCL2 and
LCAD deacetylation caused by HFD. CCND1, in HCC.
• SIRT3/AMPK/ACC pathway activation to ameliorates, • HCC cell invasion and migration via uPA receptor inac-
HFD-induced hepatic steatosis. tivation by up-regulation of PAI-1 and down-regulation
• Regulate Angptl2 pathway. of uPA.
• Inhibit nuclear translocation of NF-κB via involvement • Involvement of NF-κB p65 pathway for apoptosis pro-
of TLR4/MyD88/NF-κB pathway. motion.
• Reverse the abnormal expression of LDLR and MTTP • Improvement on lipid metabolism disorder via FXR/
followed by lipid synthesis inhibition. SREBP-1c/FAS pathway regulation.
• Restoration of Treg/Th17 ratio and regulation of chemerin/ • Suppression of TNF-α expression and mitochondrial bi-
CMKLR1 signalling pathway responsible for overall in- ogenesis induction to decreased cellular ROS.
flammation and lipid deposition reduction in liver. • Early Growth Response 1 level upregulation responsible
• Improves lipid disorder, reduced aortic plaque formation for miR-373 expression transactivation.
and alleviated hepatic lipid accumulation, mainly asso- • Promotes CDKIs p21Cip1 and p27Kip1 expression via
ciated with PCSK9 down-regulation through ERK1/2 Akt/FoxO3a/Skp2 axis regulation. Further induces G0/
pathway. G1 phase cell cycle arrest.
• Multiple genes expression modulation responsible for • Upregulation of ABCA1 protein levels via involvement of
hepatic stellate cell activation and cholangiocyte prolif- PKCδ to reduce the phosphorylation of serine residues in
eration. ABCA1.
• Reduced hepatic lipid accumulation by lipolysis gene ex- • Antiproliferative effect of berberine was due Akt,
pression upregulation and lipogenesis gene expression PHLPP2 and Mst1 involvement (an autoinhibitory trian-
downregulation followed by improvement in muscle gle).
lipid contents. • Inhibits inflammation and mitochondria-dependent ap-
• Repressed complex I in the liver and gut, mainly respon- optosis.
sible for lipid metabolism inhibition (Alleviates from • Induced AMPK activation responsible for hepatic FGF21
both obesity and fatty liver). expression via NUR77.
• Transcriptional regulation of SETD2 activity for • Strengthens radiosensitivity through Nrf2 signalling
hepatoprotection via steatosis. pathway suppression.
• Elevated HIF-2 expression, insulin resistance and disor- • Radiosensitizer towards treating liver cancer by blocking
der of lipid metabolism. autophagy and cell cycle arrest resulting in senescence.
• Activate SIRT1-FoxO1-SREBP2 signal pathway.
• Antagonizes β-catenin pathway via β-catenin translation
Hepatocellular carcinoma and mTOR activity inhibition.
Hepatic stellate,[126] HCC (hepatocellular carcinoma)
HepG2,[127–138] HUVECs,[139] SMMC-7721,[140, 141] H22,[131] Bel
7404,[131, 141] MIHA,[133] QSG-7701,[142] Huh7,[138] HHL-5,[143] Hepatitis c virus
Hep3B and HEK293[138] cells line was used to investigate the A study included cell culture-derived HCV viral
hepatoprotective action of berberine through various mech- pseudoparticles bearing HCV glycoproteins for hepatitis C
anism. Upto 96 mM dose of berberine was found to be tested. virus entry-related assays of berberine (0, 2, 5, 10, 20, 50
The various postulated mechanism for hepatoprotective ac- and 100 μM) via targeting the viral E2 glycoprotein was es-
tion of berberine are: tablished.[144] In another studies, effect of berberine (0, 1, 5,
10, 20, 40, 80, 100, 200 and 400 μM) was investigated on
• Calcium and potassium channels inhibitory actions in i- human hepatoma cells nurturing hepatitis C virus RNA via
solated rat hepatocytes. autophagy inhibition.[145]
10 Satish Sardana et al.

Table 2 Drug delivery system associated with improvement of bioavailability of berberine

Drug delivery approaches/methods Outcome

Dendrimer
Dendrimer of G4 PAMAM via conjugation and encapsulation ap- Conjugated dendrimers was found to be more prominent.[149]
proaches was developed
Erythrocyte-hemoglobin self-assembly system
Interaction with erythrocyte and the combination with Hb Low plasma and high tissue concentration was achieved.[150]
Hydrogel/beads
Self-assembled beads developed by shaking alpha-cyclodextrin with Bioavailability improvement of poorly water soluble or lipophilic
soybean oil drug.[151]
A novel composite pH-responsive hydrogel beads using carboxy- Improvement in efficacy and stability.[152]
methyl starch-g-poly (acrylic acid)/palygorskite/starch/sodium algi-

Downloaded from https://academic.oup.com/rpsppr/article/2/1/rqad002/6991356 by guest on 30 March 2023


nate (CMS-g-PAA/PGS/ST/SA)
Silk sericin-derived hydrogel by thiol-ene click chemistry Eco-friendly way for biomedical industries.[153]
Liposomes
Liposomes was developed through co-precipitation method from Improve the absorption by 4-fold compared with free drug.[154]
berberine bisulfate and polyvinyl pyrrolidone (PVP) with the ratio of
1: 5.
Liposomes was developed by thin-film hydration/extrusion method. Exhibits tumour growth inhibition in HepG2 tumour-bearing mice.[155]
Memory fibers
Programme release through shape memory fibres by core-sheath wet- Smart drug delivery-based vehicles of targeted drug with highly adjust-
spinning technology able doses.[156]
Microparticles
Microparticles in hollow capsular devices (in house 3D printed) via Improve sustained and prolonged absorption with oral bioavailability
emulsion crosslinking method enhancement.[157]
Microemulsion
Oleic acid, Tween 80 and PEG400 was incorporate to get the Promising for oral drug delivery system.[158]
microemulsion
Microemulsion technique was developed via pseudo tertiary phase Improvement of absorption in the intestinal tract.[159]
diagrams
Zuojin Wan microemulsion-based gel and hydrogel delivery system Improvement in relative bioavailability for transdermal route.[160]
Nanoparticles/nano-crystals/nanocarriers
Biogenic gold nanoparticles using Trapa bispinosa was developed Active against folic acid expressing HeLa cells.[161]
Chitosan-coating based nano-liposomal carrier Promising for the oral delivery and improve in bioavailability.[162]
Chitosan/fucoidan-taurine based conjugated nanoparticles Effective for treatment of defective intestinal epithelial tight junction’s
barrier.[163]
Polymeric nanoparticles via nanoprecipitation technique Improvement in entrapment efficiency.[164]
Solid lipid nanoparticles Effective against hepatosteatosis via lipogenesis inhibition and lipolysis
induction.[165]
Janus magnetic mesoporous silica nanoparticles based on Fe3O4 head Safe and effective against hepatocellular carcinoma.[166]
for magnetic targeting and a mesoporous SiO2 body for delivery of
berberine.
PEG-lipid-PLGA hybrid nanoparticles by solvent evaporation method Improvement in relative oral bioavailability, affinity, liposolubility, drug-
loading efficiency and sustained/controlled release action.[167]
Spherical shaped sliver based nano-particles was synthesized by the Improves acetaminophen-induced liver and kidney damage probably via
biofabrication technique proinflammatory factor & NF-kB factors inhibition.[168]
Brij-S20-modified nanocrystals Improvement in the oral bioavailability having poor water solubility
and Pgp-mediated efflux.[169]
Janus gold mesoporous silica nanocarriers using folic acid Triple-therapies strategies for liver cancer.[170]
Sonication, coating and extrusion based on red blood cell membrane- RBGPs hold the potential to achieve sustained-release and long circula-
camouflaged BH-loaded gelatin nanoparticles (RBGPs) tion to avoid side effects associated by high plasma concentration.[171]
PLGA nanoparticles by nanoprecipitation and encapsulation Liver protection is higher as compare to normal drug.[172]
Natural polysaccharide (chitosan-alginate)-based nanoparticles by Effective for oral delivery of poorly soluble and permeable drugs.[173]
ionic gelation method
Nanoparticles developed by anti-solvent precipitation methods Improved in liver protection and high blood concentration.[174]
Nanostructured lipid carriers using modified solvent injection method Mitigate hepatic I/R-induced lesion via modulation of HMGB1/TLR4/
NF-κB signaling and autophagy.[175]
Advance drug delivery and combinational drug approaches 11

Table 2 Continued

Drug delivery approaches/methods Outcome

Self-emulsifying or micro-emulsifying microsphere/drug


Membrane emulsification technology Suitable for poorly water soluble drug.[176]
Efficient self-micro-emulsification region was identified by pseudo Bioavailability improvement.[177]
ternary phase diagrams
The optimal self-microemulsion formula was composed of OP, To get small particle size and stable formulation.[178]
propanediol, cinnamon oil and total alkaloid from Rhizoma Coptidis
with the ratio of 4:8:3:6.
Solid dispersion
Solid dispersion by solvent evaporation technique Promising strategies for oral bioavailability improvements.[179]
ASD with hydrogenated phosphatidylcholine Improvement in intestinal absorption, permeability and bioavailabil-
ity.[180]

Downloaded from https://academic.oup.com/rpsppr/article/2/1/rqad002/6991356 by guest on 30 March 2023


Taste-masked microcapsules containing disintegrating tablets
Orally disintegrating tablets containing microcapsule and coated Taste masking to improve patient compliance.[181]
with Eudragit E100.

Ischemia/reperfusion injuries Cynara scolymus, Cichorium intybus and Taraxacum


Berberine (100  mg/kg/ oral) was investigated on ischemia/ officinalis,[189] evodiamine,[190–192] ferulic acid,[193] insulin sensi-
reperfusion (IR) hepatic injury after orthotopic liver trans- tizer,[194] lysergol,[195] metformin,[196] plant stanols,[197] puerarin
plantation on rats and found to promote liver transplant is- and baicalin,[198] red yeast rice and policosanols,[199] red yeast
chemia/reperfusion injury partly via Sirt1/FoxO3α mediated rice and Morus alba leaves extract,[200] red yeast rice and
autophagy activation.[146] Moreover, berberine (18.6 mM) was chitosan,[201] red rice, liposomal berberine and curcumin,[202]
found to preserve mitochondrial function and bioenergetics, resveratrol,[203, 204] resveratrol and glibenclamide,[205] Rhizoma
protecting the liver from the damaging effects caused by I/R, Coptidis with Cortex Cinnamomi,[206] S allyl cysteine,[207]
when tested as a part of preservation solution for rat model silymarin,[208, 209] simvastatin,[210] sitagliptin,[211] and sodium
of ex vivo liver transplant.[147] Similarly, berberine (100 mg/ caprate.[212–214] Among advance drug delivery including com-
kg/oral) in another study was investigated on hepatic cold is- binational approach such as baicalin complex,[215] baicalin
chemia rat model via reducing apoptosis, possibly involving and gardenia fruit complex,[216] baicalin nanocrystals,[217]
PI3K/Akt/mTOR signalling pathway modulation.[148] carboxymethylcellulose and hyaluronic acid hydrogel,[218]
carboxymethyl hexanoyl chitosan/hyaluronic acid pol-
ymer gel,[219] cellulose nanofiber based hydrogel,[220] citric
Berberine-Based Advance Drug Delivery acid based salt cocrystal,[221] Coptidis rhizoma extract based
System nano-drug delivery,[222] cucurbitacin B based phospho-
In Table 2 details on berberine-based advance drug delivery lipid complex,[223] doxorubicin based liposomes,[224] fumaric
system was discussed. Various drug delivery approaches acid based co-crystal,[225] 2-hydroxypropyl-β-cyclodextrin
such as dendrimer,[149] erythrocyte-hemoglobin self-as- (HPβCD) complex,[226] magnolol based complex,[227] modi-
sembly system,[150] hydrogel/beads,[151–153] liposomes,[154, 155] fied β-cyclodextrin encapsulation,[228] modified sodium algi-
memory fibers,[156] microparticles,[157] microemulsion,[158–160] nate and NIPAM based nanogels,[229] O-hexadecyl-dextran
nanoparticles/nano-crystals/nanocarriers,[161–175] self- based nanoparticles,[230] ibuprofen-based co-crystal,[231]
emulsifying or micro-emulsifying microsphere/drug,[176–178] mangiferin salt,[232] polylactide-poly (ethylene glycol)
solid dispersion[179, 180] and taste-masked microcapsules con- diblock copolymers based self-assembled filomicelles,[233]
taining disintegrating tablets[181] were investigated to improve sodium N-[8-(2-hydroxybenzoyl) amino] caprylate based
the solubility, bioavailability and permeability related issues. microsphere,[234] sulfonato-β-cyclodextrin based supra-
The major outcome of these delivery systems is to overcome molecular nanoparticle,[235] vanillin-cross-linked chitosan
the solubility issue, bioavailability hurdle and permeability microcarriers,[236] and wogonoside based complex.[215]
followed by improvement in efficacy and safety. Similarly, structure modification using a core molecules of
berberine such as berberrubine,[237] demethyleneberberine,[238,
239]
demethylenetetrahydroberberine,[240] oxyberberine[241] and
Berberine-Based Combinational Drug 9-O-benzoyl- substitution.[242]
Approaches
Novel combination, structure modifications and advance
drug delivery system using berberine for improvement of sol- Clinical Trial for Hepatoprotection
ubility, bioavailability and permeability was shown in Table 3. Table 4 describes the various clinical trial conducted on ber-
Berberine was investigated by combination with bicyclol,[182] berine in terms of investigations of hepatoprotective action
chitosan and chitosan hydrochloride,[55] chlorogenic acid on human beings. In brief, berberine was tested for liver func-
and tocotrienols,[183] coptisine, palmatine, epiberberine and tions,[47, 210, 243] hepatitis B and hepatitis C,[244] hypercholester-
jatrorrhizine,[184] Coptis chinensis,[185] Coptidis rhizoma ex- olemia,[245] NASH,[183, 246, 247] and NALFD.[248–254] The major
tract,[186] curcumin,[187] curcumin with dextran coating,[188] mechanisms included are
12 Satish Sardana et al.

Table 3 Combinations/structure modifications and drug delivery approaches including berberine for improving hepatoprotective action

Combinations/structure modifications and drug delivery approaches Outcome

Combinations
Bicyclol Alleviates NAFLD.[182]
Chitosan and chitosan hydrochloride Improvement in intestinal absorption.[55]
Chlorogenic acid and tocotrienols Improve metabolic system and liver parameters on overweight
subjects.[183]
Coptisine, palmatine, epiberberine and jatrorrhizine Synergetic action for lowering efficacy on cholesterol.[184]
Coptis chinensis Improvement in detoxification.[185]
Coptidis rhizoma extract (metabolites) Improvement in pharmacokinetics.[186]
Curcumin Exhibited improved ameliorative action on rats with NAFLD than
lovastatin.[187]

Downloaded from https://academic.oup.com/rpsppr/article/2/1/rqad002/6991356 by guest on 30 March 2023


Curcumin with dextran coating Promote oral absorptions with prolonged circulation and synchro-
nized biodistribution with improved ameliorative effects on NAFLD
in mice.[188]
Cynara scolymus, Cichorium intybus and Taraxacum officinalis Normalize CCl4-induced rat liver toxicity.[189]
Evodiamine Improved apoptosis on HHC.[190]
Evodiamine Cholesterol absorption inhibition in intestine during HFD.[191]
Evodiamine NAFLD via gut microbiota and intestinal barrier safety.[192]
Ferulic acid Cleansing promoters for detoxification.[193]
Insulin sensitizer NAFLD with significant influence on oxidative stress.[194]
Lysergol Bioavailability enhancement.[195]
Metformin Improvement in NAFLD with type 2 diabetes mellitus.[196]
Plant stanols Reduces plasma cholesterol synergistically in rats.[197]
Puerarin and baicalin Protects from NAFLD via upregulation of hepatic IR expression and
PPAR-γ levels.[198]
Red yeast rice and policosanols Reduces cholesterol levels via improved endothelial function and insu-
lin sensitivity.[199]
Red yeast rice and Morus alba leaves extract (LopiGLIK) Significantly reduced levels of serum CLC (−11.4%) without changing
PCSK9 plasma levels.[200]
Red yeast rice and chitosan Primary prevention by improving Non-HDL cholesterol levels on
dyslipidemia individuals.[201]
Red rice (fermented), liposomal berberine and curcumin Improve lipid profiles and reduces inflammatory markers.[202]
Resveratrol Improves the lipid-lowering efficacy.[203]
Resveratrol Exhibit chemotherapeutic effect in transglutaminase 2 and
hepatocarcinoma.[204]
Resveratrol and glibenclamide Modifies activities of xenobiotic-metabolizing enzyme.[205]
Rhizoma Coptidis with Cortex Cinnamomi Improvement in tissue distribution pattern of berberine in rats.[206]
S allyl cysteine Ameliorated hepatocarcinoma induced by DEN+CCl4.[207]
Silymarin (Berberol) Effective in reducing glycosylated hemoglobin.[208]
Silymarin Supplement for improving liver function.[209]
Simvastatin Improves the lipid-lowering efficacy.[210]
Sitagliptin Improving insulin resistance in NAFLD.[211]
Sodium caprate Improvement in intestine absorption.[212]
Sodium caprate Intestine absorption improvement in rats.[213]
Sodium caprate Inhibiting hepatic gluconeogenesis via AMPK pathway.[214]
Drug delivery approaches
Baicalin complex Improves bioavailability.[215]
Baicalin and Gardenia fruit complex Solubility enhancement.[216]
Baicalin nanocrystals Orally promote co-absorption of both the components.[217]
Carboxymethylcellulose and hyaluronic acid hydrogel Improvement in anti-inflammatory release system.[218]
Carboxymethyl hexanoyl chitosan/ hyaluronic acid polymer gel Sustained release.[219]
Cellulose nanofiber based hydrogel Controlled drug delivery.[220]
Citric acid based salt cocrystal Improving solid-state properties of berberine chloride.[221]
Coptidis rhizoma extract based nano-drug Improving pharmacokinetics.[222]
delivery
Advance drug delivery and combinational drug approaches 13

Table 3 Continued

Combinations/structure modifications and drug delivery approaches Outcome

Cucurbitacin B based phospholipid complex Improve therapeutic effectiveness associated with bile duct-drug target
delivery system.[223]
Doxorubicin based liposomes Enables superior therapeutic index than Doxil.[224]
Fumaric acid based co-crystal Solubility and Stability improvements.[225]
2-Hydroxypropyl-β-cyclodextrin (HPβCD) complex Supports to develop oral solution dosage.[226]
Magnolol based complex Improves absorption and metabolism time.[227]
Modified β-cyclodextrin encapsulation Shows a higher affinity for binding to kit22.[228]
Modified sodium alginate and NIPAM based nanogels Improves drug loading and release.[229]
O-hexadecyl-dextran based nanoparticles Improved high glucose stress induced apoptosis.[230]
Ibuprofen-based co-crystal Improves obesity via protein kinases TBK1 and IKKɛ inhibition.[231]

Downloaded from https://academic.oup.com/rpsppr/article/2/1/rqad002/6991356 by guest on 30 March 2023


Mangiferin salt Improved lipid modulation and metabolisms of glucose in HepG2
cells.[232]
Polylactide-poly (ethylene glycol) diblock copolymers-based Sustained delivery.[233]
self-assembled filomicelles
Sodium N-[8-(2-hydroxybenzoyl) amino] caprylate based microsphere Improve oral delivery.[234]
Sulfonato-β-cyclodextrin-based supramolecular nanoparticle Improvement in both controlled and sustained release action.[235]
Vanillin-cross-linked chitosan microcarriers. Carriers for both drugs and cells with improved therapeutic matrix.[236]
Wogonoside based complex Improves bioavailability.[215]
Structure modifications
Berberrubine Alleviates NAFLD via glucose modulation glucose and metabolism of
lipid and also restoring gut microbiota.[237]
Demethyleneberberine Exhibit antioxidant action, inhibits mitochondrial dysfunction and stea-
tosis caused by alcoholic liver disease mouse model.[238]
Demethyleneberberine Attenuates NAFLD via AMPK activation and oxidative stress inhibi-
tion.[239]
Demethylenetetrahydroberberine Alleviates NAFLD via NLRP3 inflammasome inhibition and oxidative
stress in mice.[240]
Oxyberberine Attenuating adipocyte inflammation and hepatic insulin pathway via
AMPK activation.[241]
9-O-benzoyl- substitution Exerts triglyceride-lowering action via AMPK pathway.[242]

• Enzymes like CYP3A4 are inhibited in liver by Ber. But • Decreasing serum levels of both ceramide and ceramide-
no significant effect was observed. 1-phosphate, respectively. Levels of Spingomyelin (with
• No significant effect on Liver/ Liver transaminases was C39 chains) were reduced, while that of Sphinomyelines
observed during the study. (with C35 chains) was enhanced.
• Combination therapy proved efficacious and safe, in • No safety concerns were reported related to liver.
hypercholesteraemic patients. • Significant improvement in lipid fat content, liver-related
• Reduced FBG and triglyceride levels in patients effected enzymes (ALT, GGT) and weight less.
by T2DM and IFG levels. Found to be safe in patients • Sex-dependent effects on NAFLD patients.
with hyperglycaemic patients with liver damage. • The effect of berberine was less significant on fasting
• Enhanced levels of serum adiponectin, in patients of blood glucose, lipid profile or liver enzymes in NAFLD
NAFLD effected with insulin. patients.
• Berberine enhanced the conditions in patients. The ad-
ministration of berberine lowered liver enzymes.
• Better insulin sensitivity, liver functions and lipid treat-
ment of treatment groups enhanced significantly. The Patents
combination was efficacious in type 2 diabetes effected Table 5 describes the patents on berberine in terms of
by NAFLD. hepatoprotective and related action. Among patients where
• Liver and metabolic parameters improved significantly hepatoprotection using berberine includes hepatitis B virus
after administration of dosage. infection treatment formula, synthetic analogues for fatty
• 25% more effectiveness was observed in the treatment liver and hepatic steatosis, glycyrrhetinate enantiomeric salt,
group with berberine. polyherbal medicinal pill, sigma 1 receptor binding com-
• Reduced in HFC and body weight. Enhancement in lipid pound, treatment of hepatic oxidative stress, cirrhosis, chronic
profiles, glucose and serum triglycerides while reduced liver disease and chronic hepatitis,[255] weight-loss formula,[256]
ALT and AST. and remedies for hepatic glycogenesis and lipid metabolism
14 Satish Sardana et al.

Table 4 Clinical investigation of berberine on human for hepatoprotection

Model Number of patient Study Dosage/duration Outcome


design

Liver functions dur- 104 patients (52: Ber- Random- All treated after transplant with Cyclosporine Enzymes like CYP3A4 inhibition
ing renal-transplant berine treatment and ized, Con- + Azathioprine (25–50 mg/day) + Prednisone in liver without any significant
recipients treatment 52: without berberine trolled (5–15 mg/day). For Cyclosporine [0–6 months effect.[47]
treatment Clinical – 6–8 mg/kg/day, 6–12 months – 4–6 mg/kg/
Trial day, over 1 year – 2–5 mg/kg/day and berberine
dose – 0.2 gm (3 times/day) – Tablets orally
Risk of berberine on 40 patients (Berberine Random Berberine: 500 mg tablets, Combination – No significant effect on Liver/
liver with moder- group: 8 Males + 12 Assign- Tablets contained Ber – 500 mg + Policosanol Liver transaminases was observed
ate mixed hyper- Females) Combination ment – 10 mg + 200 mg Red yeast Extract rice (3 mg during study.[243]
lipidemia groups: 8 males + 12 Monacolin) + Folic Acid – 2mg + Coenzyme
Females Q10 – 2 mg + Astaxanthin – 0.5 mg

Downloaded from https://academic.oup.com/rpsppr/article/2/1/rqad002/6991356 by guest on 30 March 2023


Safety in liver and 63 patients hypercho- RCT Dose Berberine: 1gm/day for 2 months; Combination therapy proved
kidney functions on lesterolemia (Ber- Simvastatin: 20 mg/day- for or 2 months. efficacious and safe, in
hypercholesteraemic berine: 20 patients; In combination – Berberine (2 times/day) = hypercholesteraemic patients.[209]
patients simvastatin: 20 Simvastatin (Once) – for 2 months
patients and berberine
+ simvastatin: 20)
Hepatitis B and 35 Patients with IFG RCT Berberine: 2 g/day for 1 months Reduced FBG and triglyceride
hepatitis C (18 Patients: Hepatitis levels in patients effected by
B – (6 with T2DM, 12 T2DM and IFG levels. Found to
with IFG); 17 Patients: be safe in patients with hyper-
Hepatitis C – (7 with glycaemic patients with liver
T2DM, 10 with IFG). damage.[244]
NAFLD 68 Patients (Treatment Clinical Berberine: 500 mg/day for 3 months Enhanced levels of serum
group: 38 and control trial adiponectin, in patients of
group: 30) NAFLD effected with insulin.[248]
Type 2 Diabetes 60 Patients in 2 RCT Berberine (500 mg/day: for 12 weeks) and Berberine enhanced the conditions
with NAFLD groups Xuezhikang for 12 weeks. in patients. The administration
of berberine lowered liver en-
zymes.[249]
NAFLD due to 78 Patients (Treatment Clinical Metformin: control group and metformin + Better insulin sensitivity, liver
type-2 diabetes group: 38 and control trial berberine: treatment group (Berberine: 500 mg/ functions and lipid treatment of
group: 40) day for 16 Weeks) treatment groups enhanced signif-
icantly. The combination was effi-
cacious in type 2 diabetes effected
by NAFLD.[250]
Liver steatosis 40 patients – Sub- Double Berberine and chlorogenic acid (Trixy®) Liver and Metabolic parameters
jected to placebo blind, improved significantly after ad-
for 8 weeks-Wash cross-over ministration of dosage.[182]
Out Period 2 weeks designed
– Resubjected to RCT
alternative therapy of
8 Weeks
NASH based com- 96 patients (Selected Random Treatment group: Ber (500 mg/day) + Yi-Gan- 25% more effectiveness was ob-
plicated metabolize randomly between trial Ling (3 months) and Control group – Acarbose served in treatment group with
syndrome treatment and control) + Yi-Gan-Ling (3 months) berberine.[246]
NAFLD LSI – 53/62; LSI + Pio Parallel- Pioglitazone-15 mg/day and Berberine-0.5 g Reduced in HFC and body
(NCT00633282) – 55/60 and LSI + Ber controlled, tid: 16 weeks (Administered 30 min before weight. Enhancement in lipid
– 47/62. Male-98 and open-label meals) profiles, glucose and serum trigly-
Female-86 RCT cerides while reduced ALT and
AST.[251]
NAFLD 41 – (LSI + Berberine) Parallel- 500 mg tid: 16 weeks (Administered 30 min Decreasing serum levels of
(NCT00633282). and 39-LSI. Male-46 controlled, before meals) both ceramide and ceramide-1-
and Female-34 open-label phosphate. Levels of Spingomyelin
RCT (with C39 chains) reduced, while
that of Sphinomyelines (With C35
chains) was enhanced.[252]
Hypercholester- 30 Patients Ran- Nutraceutical combination NC in Red Rice No Safety concerns were reported
olemia in HIV- domized extract 200 mg - (Having 3 mg Monacolin); related to liver.[245]
infected patients (2-Arm) Policosanol - 10 mg; Ber – 500 mg;
(NCT03470376) Cross- Astaxanthin – 0.5 mg; Folic Acid – 0.2 mg and
Over trial Coenzyme Q10 – 2 mg. Therapy for 3 months
(Once daily)
Advance drug delivery and combinational drug approaches 15

Table 4 Continued

Model Number of patient Study Dosage/duration Outcome


design

NASH with diabe- Placebo-32/33, Dou- Berberine ursodeoxycholate (BUDCA) – 500 Significant improvement in lipid
tes (NCT03656744) BUDCA 500 and ble-blind, and 1000 mg: 2 times/day-18 Weeks fat content, liver related enzymes
1000 mg. Male-28 placebo- (ALT, GGT) and weight less.[247]
and Female-72 controlled
RCT
NAFLD with LSI - 53; LSI + Pio- Parallel- Pioglitazone – 15 mg/day and Berberine 0.5 g Sex-dependent effects on NAFLD
or without 47; LSI + Ber - 55. controlled, tid: for 16 weeks patients.[253]
type 2 diabetes Male – 85 and Female open-label
(NCT00633282) - 70 RCT
NAFLD 50 patients with Open- Berberine group-6.25 gm/day orally: for 45 The effect of berberine was less
confirmed fatty liver label days (5L water including 100 g dried berberine significant on lipid profile, fasting

Downloaded from https://academic.oup.com/rpsppr/article/2/1/rqad002/6991356 by guest on 30 March 2023


(Intervention: 25 and double- boiled at 167 °F until 4L) blood glucose or liver enzymes in
control: 25) blinded NAFLD patients.[254]
RCT

Table 5 Patents on hepatoprotective potential of berberine

Patent number Objective Outcome

CN101642454 Hepatitis B virus infection Inhibit replication of intracellular DNA.[255]


treatment formula
CN102370930, Synthesis of berberine analogs Able to reduces lipid levels by increasing hepatic expression via AMPK
CN102327426 and and medication for fatty liver activation to improve the oxidation of fatty acid and reducing accu-
CN101579340 and hepatic steatosis mulation of hepatic triglyceride to release in the form of VLDL.[255]
CN101367800 Glycyrrhetinate enantiomeric Useful for chronic inflammation and hepatitis.[255]
salt
CN101347525 Polyherbal medicinal pill Useful for inflammation and cancer.[255]
WO2010101649 Sigma 1 receptor binding Protect uninfected human cells from Hepatitis C virus infection by
compound reducing susceptibility followed by inhibition of viral spread among
cellular population.[255]
WO2010040058 Treatment of hepatic oxidative Retard the liver fibrosis progression, chronic liver disease, cirrhosis
stress, cirrhosis, chronic liver and chronic hepatitis.[255]
disease and chronic hepatitis.
N103393679A Weight-loss formula Downregulated lipidated LC3-II (an autophagy marker, in a time and
dose-dependent manner). Further, reduce LC3-II accumulation via in-
hibition of autophagic flux in C3H10T1/2 adipocytes.[256]
CN109224071A Improving the development of Reduced HNF4α, miR-122, (FAS1), SREBP-1, ACCα, G6P, PEPCK
hepatic glycogenesis and lipid and CPT-1 expression levels in type 2 diabetes or palmitic acid incu-
metabolism disorders bated in HepG2 cells.[257]
CN112138007A 8-oxoberberine Protect hepatocytes by fat accumulation reduction, also able to signifi-
cantly decreased ALT, TG and AST levels in cells.[257]
CN105693805A Bile acids and berberine for Improves liver functions.[257]
treatment of liver diseases and
cholestatic disorders
CN105693813A Combination of berberine Reduce cholesterol content, promotes obvious phospholipid reduction
with glycyrrhetinic acid and inhibition of the inflammatory reaction.[257]

disorders, 8-oxoberberine based synthetic compound, treat- In this article, evidence collected from various sources is
ment of liver diseases and cholestatic disorders including bile scrutinized to accomplish hepatoprotective action of berbe-
acids and combination with glycyrrhetinic acid.[257] rine and its mechanisms, recent drug delivery system, com-
bination approaches, clinical trial and patent redirecting
towards the improvement of safety and efficacy of berberine
Conclusion and Future Direction on liver protections to meet the objective. Taken all together,
Berberine was found to be safe and effective natural product berberine herewith represents a very promising therapeutic
for long-term treatment and management of various dis- molecules for the protection of liver injuries through several
eases and pathological conditions including liver protection. damages via regulation of lipid and glucose metabolism, in-
However, the poor bioavailability of berberine (less than 1%) flammatory response and oxidative stress. The promising ap-
limits clinical use and therefore several strategies have been plications of berberine can be found to be further enhanced
recently tested to improve the bioavailability of berberine. due to poor pharmacokinetics by advance drug delivery
16 Satish Sardana et al.

approaches, combined treatment and structure modifications. 5. Vuddanda PR, Chakraborty S, Singh S. Berberine: a potential phy-
In addition to clinical efficacy, the safety of berberine is also tochemical with multispectrum therapeutic activities. Expert Opin
a key factor that affects its potential application using all Investig Drugs 2010; 19: 1297–307. doi:10.1517/13543784.2010.
the approaches as applied for improving solubility, bioavail- 517745
6. Chopra RN, Dikshit BB, Chowhan JS. Berberine and berberine-
ability and permeability. Therefore, in conclusion, restricted
containing plants in pharmacology and therapeutics. Ind Med Gaz
bioavailability and permeability can be improved by both 1932; 67: 194–7.
advance drug delivery system and combinational drug ap- 7. Ikram M. A review on the chemical and pharmacolog-
proaches as evidenced from the present work. Additionally, ical aspects of genus Berberis. Planta Med 1975; 28: 353–8.
berberine-based advance delivery system including in combi- doi:10.1055/s-0028-1097869.
nation with bioavailability and permeability enhancer may 8. Luo J, Yan D, Yang M et al. Multicomponent therapeutics of berbe-
provide an added advantage in the near future to meet the rine alkaloids. Evid Based Complement Alternat Med 2013; 2013:
objectives. Further, well-designed, large-scale, long-term and 545898. doi:10.1155/2013/545898
high-quality multicenter clinical trials are necessary to eval- 9. Kumar A, Ekavali, Chopra K et al. Current knowledge and phar-
uate the efficacy and safety of modified berberine to get sev- macological profile of berberine: an update. Eur J Pharmacol 2015;

Downloaded from https://academic.oup.com/rpsppr/article/2/1/rqad002/6991356 by guest on 30 March 2023


761: 288–97. doi:10.1016/j.ejphar.2015.05.068
eral value-added products.
10. Cicero AFG, Baggioni A. Berberine and its role in chronic disease.
Adv Exp Med Biol 2016; 928: 27–45. doi:10.1007/978-3-319-
Supplementary Material 41334-1_2
11. Yuan Z, Wang Y, Chai Y et al. Berberine action targets and its
Supplementary data are available at RPS Pharmacy and absorption behavior: how to use old drug for new mechanisms.
Pharmacology Reports online. Zhongguo Zhong Yao Za Zhi 2016; 41: 2362–70. doi:10.4268/
cjcmm20161231
12. Tabeshpour J, Imenshahidi M, Hosseinzadeh H. A review of the
Conflict of interest effects of Berberis vulgaris and its major component, berberine,
in metabolic syndrome. Iran J Basic Med Sci 2017; 20: 557–68.
The author(s) declare that there are no conflicts of interest.
doi:10.22038/IJBMS.2017.8682
13. Neag MA, Mocan A, Echeverría J et al. Berberine: botanical occur-
Acknowledgments rence, traditional uses, extraction methods, and relevance in cardi-
ovascular, metabolic, hepatic, and renal disorders. Front Pharmacol
All authors acknowledge the administrative and technical sup- 2018; 9: 557. doi:10.3389/fphar.2018.00557
port provided by Amity University Haryana, Gurugram and 14. Imenshahidi M, Hosseinzadeh H. Berberine and barberry (Ber-
DIT University, Dehradun, India for writing the current article. beris vulgaris): a clinical review. Phytother Res 2019; 33: 504–23.
doi:10.1002/ptr.6252
15. Ashrafizadeh M, Fekri HS, Ahmadi Z et al. Therapeutic and bio-
Funding logical activities of berberine: the involvement of Nrf2 signaling
pathway. J Cell Biochem 2020; 121: 1575–85. doi:10.1002/
This work was not funded by any of the agencies.
jcb.29392
16. Gao Y, Wang F, Song Y et al. The status of and trends in the phar-
Authors contributions: macology of berberine: a bibliometric review [1985-2018]. Chin
Med 2020; 15: 7. doi:10.1186/s13020-020-0288-z
Satish Sardana: Supervision; Rupa Gupta: Data curation 17. Och A, Podgórski R, Nowak R. Biological activity of berberine-a
and writing the original draft; Kumud Madan: Supervision summary update. Toxins (Basel) 2020; 12: 713. doi:10.3390/
and visualization; Dheeraj Bisht: Resources and meth- toxins12110713
odology; Vijay Singh Rana: Formal analysis; Samir 18. Ai X, Yu P, Peng L et al. Berberine: a review of its pharmacoki-
Bhargava: Resources and methodology; Neeraj K. Sethiya: netics properties and therapeutic potentials in diverse vascular
Conceptualization, methodology, visualization and revi- diseases. Front Pharmacol 2021; 12: 762654. doi:10.3389/
fphar.2021.762654
sion.
19. Xu X, Yi H, Wu J et al. Therapeutic effect of berberine on met-
abolic diseases: Both pharmacological data and clinical evi-
dence. Biomed Pharmacother 2021; 133: 110984. doi:10.1016/j.
Data availability statement
biopha.2020.110984
Data sharing is not applicable to this article as no datasets 20. Behl T, Singh S, Sharma N et al. Expatiating the pharmacological
were generated or analysed during the current study. and nanotechnological aspects of the alkaloidal drug berberine:
current and future trends. Molecules 2022; 27: 3705. doi:10.3390/
molecules27123705
References
21. Fang X, Wu H, Wei J et al. Research progress on the pharmacolog-
1. Xia X, Weng J. Targeting metabolic syndrome: candidate nat- ical effects of berberine targeting mitochondria. Front Endocrinol
ural agents. J Diabetes 2010; 2: 243–9. doi:10.1111/j.1753- 2022; 13: 982145. doi:10.3389/fendo.2022.982145
0407.2010.00090.x 22. Yarmohammadi F, Hayes W, Karimi G. The therapeutic effects of
2. Song D, Hao J, Fan D. Biological properties and clinical applica- berberine against different diseases: a review on the involvement of
tions of berberine. Front Med 2020; 14: 564–82. doi:10.1007/ the endoplasmic reticulum stress. Phytother Res 2022; 36: 3215–
s11684-019-0724-6 31. doi:10.1002/ptr.7539
3. Birdsall TC, Kelly GS. Berberine: therapeutic potential of an alka- 23. Bansod S, Saifi MA, Godugu C. Molecular updates on berberine in
loid found in several medicinal plants. Altern Med Rev 1997; 2: liver diseases: bench to bedside. Phytother Res 2021; 35: 5459–76.
94–103. doi:10.1002/ptr.7181
4. Habtemariam S. Berberine pharmacology and the gut microbi- 24. Zhou M, Deng Y, Liu M et al. The pharmacological activity of ber-
ota: a hidden therapeutic link. Pharmacol Res 2020; 155: 104722. berine, a review for liver protection. Eur J Pharmacol 2021; 890:
doi:10.1016/j.phrs.2020.104722 173655. doi:10.1016/j.ejphar.2020.173655
Advance drug delivery and combinational drug approaches 17

25. Moreira ES, Ames-Sibin AP, Bonetti CI et al. The short-term ef- 44. Chen C, Yu Z, Li Y et al. Effects of berberine in the gastrointestinal
fects of berberine in the liver: Narrow margins between bene- tract-a review of actions and therapeutic implications. Am J Chin
fits and toxicity. Toxicol Lett 2022; 368: 56–65. doi:10.1016/j. Med 2014; 42: 1053–70. doi:10.1142/S0192415X14500669
toxlet.2022.08.005 45. Chan E. Displacement of bilirubin from albumin by berberine. Biol
26. Liu Y, Zhang L, Song H et al. Update on berberine in nonalcoholic Neonat 1993; 63: 201–8. doi:10.1159/000243932
Fatty liver disease. Evid Based Complement Alternat Med 2013; 46. Tan YZ, Wu AC, Tan BY et al. Study on the interactions of berbe-
2013: 308134. doi:10.1155/2013/308134 rine displace other drug from their plasma proteins binding sites.
27. Zhu X, Bian H, Gao X. The potential mechanisms of berberine in Chin Pharmacol Bull 2002; 18: 576–8.
the treatment of nonalcoholic fatty liver disease. Molecules 2016; 47. Wu X, Lu Q, Xin H et al. Effects of berberine on the blood con-
21: 1336. doi:10.3390/molecules21101336 centration of cyclosporine A in renal transplanted recipients: clin-
28. Wei X, Wang C, Hao S et al. The therapeutic effect of berberine in ical and pharmacokinetic study. Eur J Clin Pharmacol 2005; 61:
the treatment of nonalcoholic fatty liver disease: a meta-analysis. 567–72. doi:10.1007/s00228-005-0952-3
Evid Based Complement Alternat Med 2016; 2016: 3593951. 48. Xin HW, Wu XC, Li Q et al. The effects of berberine on the
doi:10.1155/2016/3593951 pharmacokinetics of cyclosporine A in healthy volunteers.
29. Cicero AFG, Colletti A, Bellentani S. Nutraceutical approach to Methods Find Exp Clin Pharmacol 2006; 28: 25–9. doi:10.1358/

Downloaded from https://academic.oup.com/rpsppr/article/2/1/rqad002/6991356 by guest on 30 March 2023


Non-Alcoholic Fatty Liver Disease (NAFLD): the available clinical mf.2006.28.1.962774
evidence. Nutrients 2018; 10: 1153. doi:10.3390/nu10091153 49. Linn YC, Lu J, Lim LC et al. Berberine-induced haemolysis re-
30. Ren S, Ma X, Wang R et al. Preclinical evidence of berberine on visited: safety of Rhizoma coptidis and cortex phellodendri in
non-alcoholic fatty liver disease: a systematic review and meta- chronic haematological diseases. Phytother Res 2012; 26: 682–6.
analysis of animal studies. Front Pharmacol 2021; 12: 742465. doi:10.1002/ptr.3617
doi:10.3389/fphar.2021.742465 50. Lao-Ong T, Chatuphonprasert W, Jarukamjorn K. Berbe-
31. Wang Y, Tai Y, Zhao D et al. Berberine prevents disease progression rine disturbs the expression of sex-hormone regulated genes
of nonalcoholic Steatohepatitis through modulating multiple path- in βnaphthoflavone-induced mice. J Biol Sci 2013; 13: 271–6.
ways. Cells 2021; 10: 210. doi:10.3390/cells10020210 doi:10.3923/jbs.2013.271.276
32. Koperska A, Wesołek A, Moszak M et al. Berberine in non-alcoholic 51. Mahmoudi M, Rabe SZT, Balali-Mood M et al. Immunotoxicity in-
fatty liver disease-a review. Nutrients 2022; 14: 3459. doi:10.3390/ duced in mice by subacute exposure to berberine. J Immunotoxicol
nu14173459 2016; 13: 255–62. doi:10.3109/1547691X.2015.1058306
33. Liu X, Wang L, Tan S et al. Therapeutic effects of berberine on 52. Marcheluzzo S, Faggian M, Zancato M et al. Analysis of monacolins
liver fibrosis are associated with lipid metabolism and intes- and berberine in food supplements for lipid control: an overview
tinal flora. Front Pharmacol 2022; 13: 814871. doi:10.3389/ of products sold on the italian market. Molecules 2021; 26: 2222.
fphar.2022.814871 doi:10.3390/molecules26082222
34. Asbaghi O, Ghanbari N, Shekari M et al. The effect of berbe- 53. Singh N, Sharma B. Toxicological effects of berberine and
rine supplementation on obesity parameters, inflammation and sanguinarine. Front Mol Biosci 2018; 5: 21. doi:10.3389/
liver function enzymes: a systematic review and meta-analysis of fmolb.2018.00021
randomized controlled trials. Clin Nutr ESPEN 2020; 38: 43–9. 54. Liu X, Li W, Zhang H et al. Biodistribution and pharmacokinetic pro-
doi:10.1016/j.clnesp.2020.04.010 file of berberine and its metabolites in hepatocytes. Phytomedicine
35. Mirhadi E, Rezaee M, Malaekeh-Nikouei B. Nano strategies for ber- 2022; 104: 154288. doi:10.1016/j.phymed.2022.154288
berine delivery, a natural alkaloid of Berberis. Biomed Pharmacother 55. Chen W, Fan D, Meng L et al. Enhancing effects of chitosan and
2018; 104: 465–73. doi:10.1016/j.biopha.2018.05.067 chitosan hydrochloride on intestinal absorption of berberine in
36. Godugu C, Patel AR, Doddapaneni R et al. Approaches to improve rats. Drug Dev Ind Pharm 2012; 38: 104–10. doi:10.3109/036390
the oral bioavailability and effects of novel anticancer drugs berbe- 45.2011.592531
rine and betulinic acid. PLoS One 2014; 9: e89919. doi:10.1371/ 56. Chen W, Miao Y, Fan D et al. Bioavailability study of berberine
journal.pone.0089919 and the enhancing effects of TPGS on intestinal absorption in rats.
37. Iqbal MJ, Quispe C, Javed Z et al. Nanotechnology-based strategies AAPS PharmSciTech 2011; 12: 705–11. doi:10.1208/s12249-011-
for berberine delivery system in cancer treatment: pulling strings 9632-z
to keep berberine in power. Front Mol Biosci 2021; 7: 624494. 57. Liu CS, Zheng YR, Zhang YF et al. Research progress on berberine
doi:10.3389/fmolb.2020.624494 with a special focus on its oral bioavailability. Fitoterapia 2016;
38. Khan S, Hussain A, Attar F et al. A review of the berberine nat- 109: 274–82. doi:10.1016/j.fitote.2016.02.001
ural polysaccharide nanostructures as potential anticancer and 58. Pan GY, Wang GJ, Liu XD et al. The involvement of P-glycoprotein
antibacterial agents. Biomed Pharmacother 2022; 146: 112531. in berberine absorption. Pharmacol Toxicol 2002; 91: 193–7.
doi:10.1016/j.biopha.2021.112531 doi:10.1034/j.1600-0773.2002.t01-1-910403.x
39. Majidzadeh H, Araj-Khodaei M, Ghaffari M et al. Nano-based de- 59. Tsai PL, Tsai TH. Hepatobiliary excretion of berberine. Drug
livery systems for berberine: a modern anti-cancer herbal medicine. Metab Dispos 2003; 32: 405–12. doi:10.1124/dmd.32.4.405
Colloids Surf B Biointerfaces 2020; 194: 111188. doi:10.1016/j. 60. Zuo F, Nakamura N, Akao T et al. Pharmacokinetics of berberine
colsurfb.2020.111188 and its main metabolites in conventional and pseudo germ-free
40. Mujtaba MA, Akhter MH, Alam MS et al. An updated review on rats determined by liquid chromatography/ion trap mass spec-
therapeutic potential and recent advances in drug delivery of ber- trometry. Drug Metab Dispos 2006; 34: 2064–72. doi:10.1124/
berine: current status and future prospect. Curr Pharm Biotechnol dmd.106.011361
2022; 23: 60–71. doi:10.2174/1389201022666210208152113 61. Chen CM, Chang HC. Determination of berberine in plasma, urine
41. Guan S, Wang B, Li W et al. Effects of berberine on expression and bile by high performance liquid chromatograpy. J Chromatogr
of LOX-1 and SR-BI in human macrophage-derived foam cells in- 1995; 665: 117–23. doi:10.1016/0378-4347(94)00517-9
duced by ox-LDL. Am J Chin Med 2010; 38: 1161–9. doi:10.1142/ 62. Tan XS, Ma JY, Feng R et al. Tissue distribution of berberine and
s0192415x10008548 its metabolites after oral administration in rats. PLoS One 2013; 8:
42. Kupeli E, Kosar M, Yesilada E et al. A comparative study on e77969. doi:10.1371/journal.pone.0077969
the anti-inflammatory, antinociceptive and antipyretic effects of 63. Vivoli E, Cappon A, Milani S et al. NLRP3 inflammasome as a
isoquinoline alkaloids from the roots of Turkish berberis species. target of berberine in experimental murine liver injury: interfer-
Life Sci 2002; 72: 645–57. doi:10.1016/s0024-3205(02)02200-2 ence with P2X7 signalling. Clin Sci (Lond) 2016; 130: 1793–806.
43. Kheir MM, Wang Y, Hua L et al. Acute toxicity of berberine and doi:10.1042/CS20160400
its correlation with the blood concentration in mice. Food Chem 64. Zhao Z, Wei Q, Hua W et al. Hepatoprotective effects of ber-
Toxicol 2010; 48: 1105–10. doi:10.1016/j.fct.2010.01.033 berine on acetaminophen-induced hepatotoxicity in mice.
18 Satish Sardana et al.

Biomed Pharmacother 2018; 103: 1319–26. doi:10.1016/j. 83. Hasanein P, Ghafari-Vahed M, Khodadadi I. Effects of isoquinoline
biopha.2018.04.175 alkaloid berberine on lipid peroxidation, antioxidant defense
65. Khodayar MJ, Javadipour M, Keshtzar E et al. Role of berberine system, and liver damage induced by lead acetate in rats. Redox
against arsenic induced oxidative damage in isolated rat liver mito- Rep 2017; 22: 42–50. doi:10.1080/13510002.2016.1140406
chondria. J Environ Biol 2016; 37: 285–90. 84. Mahmoud AM, Hozayen WG, Ramadan SM. Berberine ameli-
66. Ye X, Feng Y, Tong Y et al. Hepatoprotective effects of Coptidis orates methotrexate-induced liver injury by activating Nrf2/
rhizoma aqueous extract on carbon tetrachloride-induced acute HO-1 pathway and PPARγ, and suppressing oxidative stress
liver hepatotoxicity in rats. J Ethnopharmacol 2009; 124: 130–6. and apoptosis in rats. Biomed Pharmacother 2017; 94: 280–91.
doi:10.1016/j.jep.2009.04.003 doi:10.1016/j.biopha.2017.07.101
67. Feng Y, Siu K, Ye X et al. Hepatoprotective effects of berberine 85. Mehrzadi S, Fatemi I, Esmaeilizadeh M et al. Hepatoprotective
on carbon tetrachloride-induced acute hepatotoxicity in rats. Chin effect of berberine against methotrexate induced liver toxicity
Med 2010; 5: 33. doi:10.1186/1749-8546-5-33 in rats. Biomed Pharmacother 2018; 97: 233–9. doi:10.1016/j.
68. Domitrović R, Jakovac H, Blagojević G. Hepatoprotective activity biopha.2017.10.113
of berberine is mediated by inhibition of TNF-α, COX-2, and iNOS 86. Shalkami AS, Hassanein EHM, Sayed AM et al. Hepatoprotective
expression in CCl (4)-intoxicated mice. Toxicology 2011; 280: 33– effects of phytochemicals berberine and umbelliferone against

Downloaded from https://academic.oup.com/rpsppr/article/2/1/rqad002/6991356 by guest on 30 March 2023


43. doi:10.1016/j.tox.2010.11.005 methotrexate-induced hepatic intoxication: experimental studies
69. Domitrović R, Jakovac H, Marchesi VV et al. Resolution of liver and in silico evidence. Environ Sci Pollut Res Int 2021; 28:
fibrosis by isoquinoline alkaloid berberine in CCl4-intoxicated mice 67593–607. doi:10.1007/s11356-021-15358-4
is mediated by suppression of oxidative stress and upregulation of 87. Eftekhari A, Hasanzadeh A, Khalilov R et al. Hepatoprotective
MMP-2 expression. J Med Food 2013; 16: 518–28. doi:10.1089/ role of berberine against paraquat-induced liver toxicity in rat.
jmf.2012.0175 Environ Sci Pollut Res Int 2020; 27: 4969–75. doi:10.1007/
70. Li J, Pan Y, Kan M et al. Hepatoprotective effects of berberine on s11356-019-07232-1
liver fibrosis via activation of AMP-activated protein kinase. Life 88. Guo N, Yan A, Gao X et al. Berberine sensitizes rapamycin-
Sci 2014; 98: 24–30. doi:10.1016/j.lfs.2013.12.211 mediated human hepatoma cell death in vitro. Mol Med Rep
71. Ghareeb DA, Khalil S, Hafez HS et al. Berberine reduces neu- 2014; 10: 3132–8. doi:10.3892/mmr.2014.2608
rotoxicity related to nonalcoholic steatohepatitis in rats. 89. Hwang JM, Wang CJ, Chou FP et al. Inhibitory effect of berberine
Evid Based Complement Alternat Med 2015; 2015: 361847. on tert-butyl hydroperoxide-induced oxidative damage in rat liver.
doi:10.1155/2015/361847 Arch Toxicol 2002; 76: 664–70. doi:10.1007/s00204-002-0351-9
72. Han C, Sun T, Xv G et al. Berberine ameliorates CCl4-induced 90. Zhou J, Zhou S. Protective effect of berberine on antioxidant en-
liver injury in rats through regulation of the Nrf2-Keap1-ARE zymes and positive transcription elongation factor b expression
and p53 signaling pathways. Mol Med Rep 2019; 20: 3095–102. in diabetic rat liver. Fitoterapia 2011; 82: 184–9. doi:10.1016/j.
doi:10.3892/mmr.2019.10551 fitote.2010.08.019
73. Gholampour F, Masoudi R, Khaledi M et al. Berberis integerrima 91. Zhou JY, Zhou SW, Zhang KB et al. Chronic effects of berberine
hydro-alcoholic root extract and its constituent berberine protect on blood, liver glucolipid metabolism and liver PPARs expression
against cisplatin-induced nephro- and hepato-toxicity. Am J Med in diabetic hyperlipidemic rats. Biol Pharm Bull 2008; 31: 1169–
Sci 2022; 364: 76–87. doi:10.1016/j.amjms.2021.10.037 76. doi:10.1248/bpb.31.1169
74. Germoush MO, Mahmoud AM. Berberine mitigates 92. Eissa LA, Kenawy HI, El-Karef A et al. Antioxidant and anti-in-
cyclophosphamide-induced hepatotoxicity by modulating antiox- flammatory activities of berberine attenuate hepatic fibrosis in-
idant status and inflammatory cytokines. J Cancer Res Clin Oncol duced by thioacetamide injection in rat. Chem Biol Interact 2018;
2014; 140: 1103–9. doi:10.1007/s00432-014-1665-8 294: 91–100. doi:10.1016/j.cbi.2018.08.016
75. Qin C, Zhang H, Zhao L et al. Microbiota transplantation reveals 93. Zverinsky IV, Zverinskaya HG, Sutsko IP et al. Effects of ber-
beneficial impact of berberine on hepatotoxicity by improving gut berine on the recovery of rat liver xenobiotic-metabolizing en-
homeostasis. Sci China Life Sci 2018; 61: 1537–44. doi:10.1007/ zymes after partial hepatectomy. Biomed Khim 2015; 61: 381–3.
s11427-017-9202-0 doi:10.18097/pbmc20156103381
76. Zhao X, Zhang J, Tong N et al. Protective effects of berberine 94. Yang L, Yu S, Yang Y et al. Berberine improves liver injury induced
on doxorubicin-induced hepatotoxicity in mice. Biol Pharm Bull glucose and lipid metabolic disorders via alleviating ER stress of
2012; 35: 796–800. doi:10.1248/bpb.35.796 hepatocytes and modulating gut microbiota in mice. Bioorg Med
77. Chen X, Zhang Y, Zhu Z et al. Protective effect of berberine on Chem 2021; 55: 116598. doi:10.1016/j.bmc.2021.116598
doxorubicin-induced acute hepatorenal toxicity in rats. Mol Med 95. Janbaz KH, Gilani AH. Studies on preventive and curative ef-
Rep 2016; 13: 3953–60. doi:10.3892/mmr.2016.5017 fects of berberine on chemical-induced hepatotoxicity in
78. Sun B, Yang Y, He M et al. Hepatoprotective role of berberine on rodents. Fitoterapia 2000; 71: 25–33. doi:10.1016/s0367-
doxorubicin induced hepatotoxicity – involvement of Cyp. Curr 326x(99)00098-2
Drug Metab 2022; 21: 541–7. doi:10.2174/13892002216662006 96. Yi J, Wu S, Tan S et al. Berberine alleviates liver fibrosis through
20203648 inducing ferrous redox to activate ROS-mediated hepatic stellate
79. Ke X, Zhang R, Li P et al. Hydrochloride Berberine ameliorates cells ferroptosis. Cell Death Discov 2021; 7: 374. doi:10.1038/
alcohol-induced liver injury by regulating inflammation and lipid s41420-021-00768-7
metabolism. Biochem Biophys Res Commun 2022; 610: 49–55. 97. Zhang B, Xu D, Guo Y et al. Protection by and anti-oxidant mech-
doi:10.1016/j.bbrc.2022.04.009 anism of berberine against rat liver fibrosis induced by multiple
80. Li S, Wang N, Tan H et al. Modulation of gut microbiota medi- hepatotoxic factors. Clin Exp Pharmacol Physio 2008; 35: 303–9.
ates berberine-induced expansion of immuno-suppressive cells to doi:10.1111/j.1440-1681.2007.04819.x
against alcoholic liver disease. Clin Transl Med 2020; 10: e112. 98. Dkhil MA. Role of berberine in ameliorating Schistosoma
doi:10.1002/ctm2.112 mansoni-induced hepatic injury in mice. Biol Res 2014; 47: 8.
81. Zhang P, Ma D, Wang Y et al. Berberine protects liver from ethanol- doi:10.1186/0717-6287-47-8
induced oxidative stress and steatosis in mice. Food Chem Toxicol 99. Xing L, Zhang L, Liu T et al. Berberine reducing insulin resist-
2014; 74: 225–32. doi:10.1016/j.fct.2014.10.005 ance by up-regulating IRS-2 mRNA expression in nonalcoholic
82. Gholampour F, Keikha S. Berberine protects the liver and kidney fatty liver disease (NAFLD) rat liver. Eur J Pharmacol 2011; 668:
against functional disorders and histological damages induced 467–71. doi:10.1016/j.ejphar.2011.07.036
by ferrous sulfate. Iran J Basic Med Sci 2018; 21: 476–82. 100. Yang QH, Hu SP, Zhang YP et al. Effect of berberine on expres-
doi:10.22038/IJBMS.2018.25199.6241 sions of uncoupling protein-2 mRNA and protein in hepatic tissue
Advance drug delivery and combinational drug approaches 19

of non-alcoholic fatty liver disease in rats. Chin J Integr Med 119. Teodoro JS, Duarte FV, Gomes AP et al. Berberine reverts hepatic
2011; 17: 205–11. doi:10.1007/s11655-011-0668-4 mitochondrial dysfunction in high-fat fed rats: a possible role for
101. Zhang Y, Chang X, Song X et al. Berberine reverses abnormal SirT3 activation. Mitochondrion 2013; 13: 637–46. doi:10.1016/j.
expression of L-type pyruvate kinase by DNA demethylation and mito.2013.09.002
histone acetylation in the livers of the non-alcoholic fatty disease 120. Cao Y, Pan Q, Cai W et al. Modulation of gut microbiota by ber-
rat. Int J Clin Exp Med 2015; 8: 7535–43. berine improves steatohepatitis in high-fat diet-fed BALB/C mice.
102. Guo T, Woo S, Guo X et al. Berberine ameliorates hepatic steatosis Arch Iran Med 2016; 19: 197–203.
and suppresses liver and adipose tissue inflammation in mice with 121. Xia Q, Wu F, Wu W et al. Berberine reduces hepatic ceramide
diet-induced obesity. Sci Rep 2016; 6: 22612–22. doi:10.1038/ levels to improve insulin resistance in HFD-fed mice by inhibiting
srep22612 HIF-2α. Biomed Pharmacother 2022; 50: 112955. doi:10.1016/j.
103. Zhang Z, Li B, Meng X et al. Berberine prevents progression from biopha.2022.112955
hepatic steatosis to steatohepatitis and fibrosis by reducing endo- 122. Yuan X, Wang J, Tang X et al. Berberine ameliorates nonalco-
plasmic reticulum stress. Sci Rep 2016; 6: 20848. doi:10.1038/ holic fatty liver disease by a global modulation of hepatic mRNA
srep20848 and lncRNA expression profiles. J Transl Med 2015; 13: 24.
104. Li D, Zheng J, Hu Y et al. Amelioration of intestinal barrier dys- doi:10.1186/s12967-015-0383-6

Downloaded from https://academic.oup.com/rpsppr/article/2/1/rqad002/6991356 by guest on 30 March 2023


function by berberine in the treatment of nonalcoholic fatty liver 123. Yang J, Ma X, Li L et al. Berberine ameliorates non-alcoholic
disease in rats. Pharmacogn Mag 2017; 13: 677–82. doi:10.4103/ steatohepatitis in ApoE -/- mice. Exp Ther Med 2017; 14: 4134–
pm.pm_584_16 40. doi:10.3892/etm.2017.5051
105. Sun R, Yang N, Kong B et al. Orally administered berberine 124. Luo Y, Tian G, Zhuang Z et al. Berberine prevents non-alcoholic
modulates hepatic lipid metabolism by altering microbial bile steatohepatitis-derived hepatocellular carcinoma by inhibiting in-
acid metabolism and the intestinal FXR signaling pathway. Mol flammation and angiogenesis in mice. Am J Transl Res 2019; 11:
Pharmacol 2017; 91: 110–22. doi:10.1124/mol.116.106617 2668–82.
106. Zhao L, Cang Z, Sun H et al. Berberine improves glucogenesis and 125. Lu Z, Lu F, Wu L et al. Berberine attenuates non-alcoholic
lipid metabolism in nonalcoholic fatty liver disease. BMC Endocr steatohepatitis by regulating chemerin/CMKLR1 signalling
Disord 2017; 17: 13. doi:10.1186/s12902-017-0165-7 pathway and Treg/Th17 ratio. Naunyn Schmiedebergs Arch
107. Zhang YY, Yan JJ, Zhang P et al. Berberine maintains gut micro- Pharmacol 2021; 394: 383–90. doi:10.1007/s00210-020-01914-
biota homeostasis and ameliorates liver inflammation in experi- 1
mental non-alcoholic fatty liver disease. Chin J Gastroenter 2018; 126. Sun X, Zhang X, Hu H et al. Berberine inhibits hepatic stellate cell
23: 209–15. doi:10.3969/j.issn.1008-7125.2018.04.004 proliferation and prevents experimental liver fibrosis. Biol Pharm
108. Deng Y, Tang K, Chen R et al. Berberine attenuates hepatic oxi- Bull 2009; 32: 1533–7. doi:10.1248/bpb.32.1533
dative stress in rats with non-alcoholic fatty liver disease via the 127. Wang G, Lv Q, Dong Q et al. Berbamine induces Fas-mediated
Nrf2/ARE signalling pathway. Exp Ther Med 2019; 17: 2091–8. apoptosis in human hepatocellular carcinoma HepG2 cells and in-
doi:10.3892/etm.2019.7208 hibits its tumor growth in nude mice. J Asian Nat Prod Res 2009;
109. Zhu X, Bian H, Wang L et al. Berberine attenuates nonalcoholic 11: 219–28. doi:10.1080/10286020802675076
hepatic steatosis through the AMPK-SREBP-1c-SCD1 pathway. 128. Yin CM, Wang Q, Wang HL. Impact of emodin and berberine on
Free Radic Biol Med 2019; 141: 192–204. doi:10.1016/j. the mRNA expression of leptin receptor in HepG2 cells treated
freeradbiomed.2019.06.019 with high concentrations of leptin. World Chin J Digestol 2009;
110. Xu X, Zhu X, Bai J et al. Berberine alleviates nonalcoholic fatty 17: 1122–5. doi:10.11569/wcjd.v17.i11.1122
liver induced by a high-fat diet in mice by activating SIRT3. 129. Yu R, Zhang Z, Wang B et al. Berberine-induced apoptotic and
FASEB J 2019; 33: 7289–300. doi:10.1096/fj.201802316r autophagic death of HepG2 cells requires AMPK activation.
111. Zhang Y, Deng Y, Tang K et al. Berberine ameliorates high-fat Cancer Cell Int 2014; 14: 49. doi:10.1186/1475-2867-14-49
diet-induced non-alcoholic fatty liver disease in rats via activation 130. Zhou Y, Cao S, Wang Y et al. Berberine metabolites could induce
of SIRT3/AMPK/ACC pathway. Curr Med Sci 2019; 39: 37–43. low density lipoprotein receptor up-regulation to exert lipid-
doi:10.1007/s11596-019-1997-3 lowering effects in human hepatoma cells. Fitoterapia 2014; 92:
112. Lu Z, He B, Chen Z et al. Anti-inflammatory activity of berberine 230–7. doi:10.1016/j.fitote.2013.11.010
in non-alcoholic fatty liver disease via the Angptl2 pathway. BMC 131. Li J, Li O, Kan M et al. Berberine induces apoptosis by sup-
Immunol 2020; 21: 28. doi:10.1186/s12865-020-00358-9 pressing the arachidonic acid metabolic pathway in hepatocellular
113. Wang L, Jia Z, Wang B et al. Berberine inhibits liver damage in carcinoma. Mol Med Rep 2013; 12: 4572–7. doi:10.3892/
rats with non-alcoholic fatty liver disease by regulating TLR4/ mmr.2015.3926
MyD88/NF-κB pathway. Turk J Gastroenterol 2020; 31: 902–9. 132. Chen J, Wu F, Luo H et al. Berberine upregulates miR-22-3p to
doi:10.5152/tjg.2020.19568 suppress hepatocellular carcinoma cell proliferation by targeting
114. Chen P, Li Y, Xiao L. Berberine ameliorates nonalcoholic fatty Sp1. Am J Transl Res 2016; 8: 4932–41.
liver disease by decreasing the liver lipid content via reversing the 133. Li M, Zhang M, Zhang Z et al. Induction of apoptosis by berbe-
abnormal expression of MTTP and LDLR. Exp Ther Med 2021; rine in hepatocellular carcinoma HepG2 cells via downregulation
22: 1109. doi:10.3892/etm.2021.10543 of NF-κB. Oncol Res 2017; 25: 233–9. doi:10.3727/0965040
115. Wang L, Xu B, Sagada G et al. Dietary berberine regulates lipid me- 16X14742891049073
tabolism in muscle and liver of black sea bream (Acanthopagrus 134. Liu YN, Zhang ZX. Effect of berberine on a cellular model of
schlegelii) fed normal or high-lipid diets. Br J Nutr 2021; 125: non-alcoholic fatty liver disease. Int J Clin Exp Med 2017; 10:
481–93. doi:10.1017/S0007114520003025 16360–6.
116. Yu M, Alimujiang M, Hu L et al. Berberine alleviates lipid metabo- 135. Rafiei H, Omidian K, Bandy B. Comparison of dietary polyphe-
lism disorders via inhibition of mitochondrial complex I in gut and nols for protection against molecular mechanisms underlying
liver. Int J Biol Sci 2021; 17: 1693–707. doi:10.7150/ijbs.54604 nonalcoholic fatty liver disease in a cell model of steatosis. Mol
117. Dai L, Lu S, Shen T et al. Methyltransferase SETD2 mediates Nutr Food Res 2017; 61: 1600781. doi:10.1002/mnfr.201600781
hepatoprotection of berberine against steatosis. Ann Transl Med 136. Ramesh G, Das S, Sadashiva SRB. Berberine, a natural alka-
2022; 10: 552552.–552. doi:10.21037/atm-22-1753 loid sensitizes human hepatocarcinoma to ionizing radiation by
118. Chang X, Yan H, Fei J et al. Berberine reduces methylation of blocking autophagy and cell cycle arrest resulting in senescence. J
the MTTP promoter and alleviates fatty liver induced by a high- Pharm Pharmacol 2020; 72: 1893–908. doi:10.1111/jphp.13354
fat diet in rats. J Lipid Res 2010; 51: 2504–15. doi:10.1194/jlr. 137. Shan M, Dai Y, Ren X et al. Berberine mitigates nonalcoholic
m001958 hepatic steatosis by downregulating SIRT1-FoxO1-SREBP2
20 Satish Sardana et al.

pathway for cholesterol synthesis. J Integr Med 2021; 19: 545–54. sheath wet-spinning technology. J Biomed Nanotechnol 2019; 15:
doi:10.1016/j.joim.2021.09.003 1432–42. doi:10.1166/jbn.2019.2784
138. Vishnoi K, Ke R, Saini KS et al. Berberine represses β-catenin trans- 157. Choudhury D, Jala A, Murty US et al. In vitro and in vivo evalu-
lation involving 4E-BPs in hepatocellular carcinoma cells. Mol ations of berberine-loaded microparticles filled in-house 3D
Pharmacol 2021; 99: 1–16. doi:10.1124/molpharm.120.000029 printed hollow capsular device for improved oral bioavailability.
139. Jie S, Li H, Tian Y et al. Berberine inhibits angiogenic potential AAPS PharmSciTech 2022; 23: 89. doi:10.1208/s12249-022-
of Hep G2 cell line through VEGF down-regulation in vitro. J 02241-9
Gastroenterol Hepatol 2011; 26: 179–85. doi:10.1111/j.1440- 158. Gui SY, Wu L, Peng DY et al. Preparation and evaluation of a
1746.2010.06389.x microemulsion for oral delivery of berberine. Pharmazie 2008;
140. Li Q, Zhang L, Zu Y et al. Generation of reactive oxygen spe- 63: 516–9.
cies by a novel berberine-bile acid analog mediates apoptosis 159. Gui S, Wu L, Pan J et al. Study on preparation of berberine
in hepatocarcinoma SMMC-7721 cells. Biochem Biophys Res microemulsion and its absorption in intestine. Zhongguo Zhong
Commun 2013; 433: 432–7. doi:10.1016/j.bbrc.2013.02.104 Yao Za Zhi 2009; 34: 398–401.
141. Wang X, Wang N, Li H et al. Up-regulation of PAI-1 and down- 160. Cheng L, Wang Y, Liu S et al. Comparative study on pharmacoki-
regulation of uPA are involved in suppression of invasiveness netics of six major alkaloids in Zuojin Wan microemulsion based gel

Downloaded from https://academic.oup.com/rpsppr/article/2/1/rqad002/6991356 by guest on 30 March 2023


and motility of hepatocellular carcinoma cells by a natural com- and hydrogel. Zhongguo Zhong Yao Za Zhi 2011; 36: 3327–31.
pound berberine. Int J Mol Sci 2016; 17: 577. doi:10.3390/ 161. Pandey S, Mewada A, Thakur M et al. Biogenic gold nanoparticles
ijms17040577 as fotillas to fire berberine hydrochloride using folic acid as mo-
142. Li F, Dong X, Lin P et al. Regulation of Akt/FoxO3a/Skp2 axis lecular road map. Mater Sci Eng C Mater Biol Appl 2013; 33:
is critically involved in berberine-induced cell cycle arrest in 3716–22. doi:10.1016/j.msec.2013.05.007
hepatocellular carcinoma cells. Int J Mol Sci 2018; 19: 327. 162. Nguyen TX, Huang L, Liu L et al. Chitosan-coated nano-liposomes
doi:10.3390/ijms19020327 for the oral delivery of berberine hydrochloride. J Mater Chem B
143. You X, Cao X, Lin Y. Berberine enhances the radiosensitivity of 2014; 2: 7149–59. doi:10.1039/c4tb00876f
hepatoma cells by Nrf2 pathway. Front Biosci 2019; 24: 1190– 163. Wu S, Don T, Lin C et al. Delivery of berberine using chitosan/
202. doi:10.2741/4775 fucoidan-taurine conjugate nanoparticles for treatment of defec-
144. Hung T, Jassey A, Liu C et al. Berberine inhibits hepatitis C virus tive intestinal epithelial tight junction barrier. Mar Drugs 2014;
entry by targeting the viral E2 glycoprotein. Phytomedicine 2019; 12: 5677–97. doi:10.3390/md12115677
53: 62–9. doi:10.1016/j.phymed.2018.09.025 164. Vuddanda PR, Mishra A, Singh SK et al. Development of poly-
145. Tai C, Jassey A, Liu C et al. Targeting autophagy augments BBR- meric nanoparticles with highly entrapped herbal hydrophilic
mediated cell death in human hepatoma cells harboring hepatitis drug using nanoprecipitation technique: an approach of quality
C virus RNA. Cells 2020; 9: 908. doi:10.3390/cells9040908 by design. Pharm Dev Technol 2015; 20: 579–87. doi:10.3109/10
146. Lin Y, Sheng M, Weng Y et al. Berberine protects against ischemia/ 837450.2014.908302
reperfusion injury after orthotopic liver transplantation via 165. Xue M, Zhang L, Yang M et al. Berberine-loaded solid lipid
activating Sirt1/FoxO3α induced autophagy. Biochem Biophys Res nanoparticles are concentrated in the liver and ameliorate
Commun 2017; 483: 885–91. doi:10.1016/j.bbrc.2017.01.028 hepatosteatosis in db/db mice. Int J Nanomedicine 2015; 10:
147. Martins RM, Rolo AP, Teodoro JS et al. Addition of berberine to 5049–57. doi:10.2147/IJN.S84565
preservation solution in an animal model of ex vivo liver trans- 166. Wang Z, Wang Y, Chang Z et al. Berberine-loaded Janus
plant preserves mitochondrial function and bioenergetics from the nanocarriers for magnetic field-enhanced therapy against
damage induced by ischemia/reperfusion. Int J Mol Sci 2018; 19: hepatocellular carcinoma. Chem Biol Drug Des 2017; 89: 464–9.
284. doi:10.3390/ijms19010284 doi:10.1111/cbdd.12866
148. Sheng M, Zhou Y, Yu W et al. Protective effect of Berberine pre- 167. Yu F, Ao M, Zheng X et al. PEG-lipid-PLGA hybrid nanoparticles
treatment in hepatic ischemia/reperfusion injury of rat. Transplant loaded with berberine-phospholipid complex to facilitate the oral
Proc 2015; 47: 275–82. doi:10.1016/j.transproceed.2015.01.010 delivery efficiency. Drug Deliv 2017; 24: 825–33. doi:10.1080/10
149. Gupta L, Sharma AK, Gothwal A et al. Dendrimer encapsulated 717544.2017.1321062
and conjugated delivery of berberine: a novel approach mitigating 168. Wu Y, Huang X, Yang M et al. Ameliorative effect of berbe-
toxicity and improving in vivo pharmacokinetics. Int J Pharm rine coated bio-active nanoparticles in acetaminophen induced
2017; 528: 88–99. doi:10.1016/j.ijpharm.2017.04.073 hepato-renal damage in diabetic rats. J Photochem Photobiol B
150. Yu Q, Li M, Chen H et al. The discovery of berberine erythrocyte- 2018; 189: 250–7. doi:10.1016/j.jphotobiol.2018.10.015
hemoglobin self-assembly delivery system: a neglected carrier un- 169. Xiong W, Sang W, Linghu KG et al. Dual-functional Brij-S20-
derlying its pharmacokinetics. Drug Deliv 2022; 29: 856–70. doi: modified nanocrystal formulation enhances the intestinal trans-
10.1080/10717544.2022.2036870 port and oral bioavailability of berberine. Int J Nanomedicine
151. Liu C, Xu Y, Ouyang H et al. Preparation and in vitro evaluation 2018; 13: 3781–93. doi:10.2147/IJN.S163763
of self-assembled beads drug delivery system of berberine hydro- 170. Li X, Wang Z, Wang X et al. Berberine-loaded Janus gold
chloride. Zhongguo Zhong Yao Za Zhi 2013; 38: 1924–8. mesoporous silica nanocarriers for chemo/radio/photothermal
152. Gao J, Fan D, Song P et al. Preparation and application of therapy of liver cancer and radiation-induced injury inhibition. Int
pH-responsive composite hydrogel beads as potential delivery car- J Nanomedicine 2019; 14: 3967–82. doi:10.2147/IJN.S206044
rier candidates for controlled release of berberine hydrochloride. R 171. Su J, Zhang R, Lian Y et al. Preparation and characterization of
Soc Open Sci 2020; 7: 200676200676. doi:10.1098/rsos.200676 erythrocyte membrane-camouflaged berberine hydrochloride-
153. Yan C, Liang J, Fang H et al. Fabrication and evaluation of silk loaded gelatin nanoparticles. Pharmaceutics 2019; 11: 93.
sericin-derived hydrogel for the release of the model drug berbe- doi:10.3390/pharmaceutics11020093
rine. Gels 2021; 7: 23. doi:10.3390/gels7010023 172. Ahrari A, Najafzadehvarzi H, Taravati A et al. The inhibitory
154. Xue Y, Weng G, He J et al. Preparation of oral berberine bisulfate effect of PLGA-encapsulated berberine on hepatotoxicity and
liposomes. Zhongguo Zhong Yao Za Zhi 1995; 20: 730–1. α-smooth muscle actin (α-SMA) gene expression. Life Sci 2021;
155. Lin Y, Kuo J, Hsu C et al. Optimizing manufacture of liposomal 284: 119884. doi:10.1016/j.lfs.2021.119884
berberine with evaluation of its antihepatoma effects in a murine 173. Kohli K, Mujtaba A, Malik R et al. Development of natural
xenograft model. Int J Pharm 2013; 441: 381–8. doi:10.1016/j. polysaccharide-based nanoparticles of berberine to enhance
ijpharm.2012.11.017 oral bioavailability: formulation, optimization, ex vivo, and in
156. Tang J, Zhao R, Yin X et al. Programmable release of berberine vivo assessment. Polymers (Basel) 2021; 13: 3833. doi:10.3390/
chloride hydrate from shape memory fibers prepared from core- polym13213833
Advance drug delivery and combinational drug approaches 21

174. Sahibzada MUK, Zahoor M, Sadiq A et al. Bioavailability and diet induced hyperlipidemic rats. Lipids Health Dis 2017; 16:
hepatoprotection enhancement of berberine and its nanoparticles 239. doi:10.1186/s12944-017-0628-x
prepared by liquid antisolvent method. Saudi J Biol Sci 2021; 28: 192. Dai Y, Zhu W, Zhou J et al. The combination of berberine and
327–32. doi:10.1016/j.sjbs.2020.10.006 evodiamine ameliorates high-fat diet-induced non-alcoholic fatty
175. Gendy AM, Elnagar MR, Allam MM et al. Berberine-loaded liver disease associated with modulation of gut microbiota in
nanostructured lipid carriers mitigate warm hepatic ischemia/ rats. Braz J Med Biol Res 2022; 55: e12096. doi:10.1590/1414-
reperfusion-induced lesion through modulation of HMGB1/ 431X2022e12096
TLR4/NF-κB signaling and autophagy. Biomed Pharmacother 193. DiNicolantonio JJ, McCarty MF, Assanga SI et al. Ferulic acid
2022; 145: 112122. doi:10.1016/j.biopha.2021.112122 and berberine, via Sirt1 and AMPK, may act as cell cleansing
176. Li H, Yi T. Preparation and in vitro characterization of berberine promoters of healthy longevity. Open Heart 2022; 9: e001801.
hydrochloride--self-emulsifying microsphere by membrane emul- doi:10.1136/openhrt-2021-001801
sification technology. Yao Xue Xue Bao 2013; 48: 554–9. 194. Du H, Wang Z. Effect of berberine combined with insulin sensi-
177. Zhu J, Tang D, Feng L et al. Development of self-microemulsifying tizer on nonalcoholic fatty liver disease and its influence on oxida-
drug delivery system for oral bioavailability enhancement of ber- tive stress. Chin J Integr Tradit West Med Dig 2017; 25: 875–9.
berine hydrochloride. Drug Dev Ind Pharm 2013; 39: 499–506. 195. Patil S, Dash RP, Anandjiwala S et al. Pharmacokinetic study of

Downloaded from https://academic.oup.com/rpsppr/article/2/1/rqad002/6991356 by guest on 30 March 2023


doi:10.3109/03639045.2012.683875 berberine from rasont and implication of lysergol for its bioavail-
178. Zhang B, Lu W, Chen W. Study on self-microemulsifying drug de- ability enhancement. J Liq Chromatogr Relat Technol 2013; 36:
livery system of Jiaotai pill active components. Zhong Yao Cai 336–48. doi:10.1080/10826076.2012.657731
2008; 31: 1068–71. 196. Cui JF. Study on the efficacy and safety of berberine combined
179. Zhang Z, Chen Y, Deng J et al. Solid dispersion of berberine- with metformin in the treatment of nonalcoholic fatty liver with
phospholipid complex/TPGS 1000/SiO2: preparation, charac- type 2 diabetes mellitus. Nei Mongol J Tradit Chin Med 2016; 35:
terization and in vivo studies. Int J Pharm 2014; 465: 306–16. 67–8. doi:10.16040/j.cnki.cn15-1101.2016.05.070
doi:10.1016/j.ijpharm.2014.01.023 197. Jia X, Chen Y, Zidichouski J et al. Co-administration of berbe-
180. Shi C, Tong Q, Fang J et al. Preparation, characterization and in rine and plant stanols synergistically reduces plasma cholesterol
vivo studies of amorphous solid dispersion of berberine with hy- in rats. Atherosclerosis 2008; 201: 101–7. doi:10.1016/j.athero-
drogenated phosphatidylcholine. Eur J Pharm Sci 2015; 74: 11–7. sclerosis.2008.03.008
doi:10.1016/j.ejps.2015.04.001 198. Zhao W, Liu L, Wang Y et al. Effects of a combination of puerarin,
181. Hu X, Li Y, Zhang E et al. Preparation and evaluation of orally baicalin and berberine on the expression of proliferator-activated
disintegrating tablets containing taste-masked microcapsules of receptor-γ and insulin receptor in a rat model of nonalcoholic
berberine hydrochloride. AAPS PharmSciTech 2013; 14: 29–37. fatty liver disease. Exp Ther Med 2016; 11: 183–90. doi:10.3892/
doi:10.1208/s12249-012-9880-6 etm.2015.2846
182. Li H, Liu N, Li J et al. Combined use of bicyclol and berberine 199. Affuso F, Ruvolo A, Micillo F et al. Effects of a nutraceutical com-
alleviates mouse nonalcoholic fatty liver disease. Front Pharmacol bination (berberine, red yeast rice and policosanols) on lipid levels
2022; 13: 843872. doi:10.3389/fphar.2022.843872 and endothelial function randomized, double-blind, placebo-
183. Cicero AFG, Rosticci M, Parini A et al. Short-term effects of a com- controlled study. Nutr Metab Cardiovasc Dis 2010; 20: 656–61.
bined nutraceutical of insulin-sensitivity, lipid level and indexes doi:10.1016/j.numecd.2009.05.017
of liver steatosis: a double-blind, randomized, cross-over clinical 200. Adorni MP, Ferri N, Marchianò S et al. Effect of a novel nutra-
trial. Nutr J 2015; 14: 30. doi:10.1186/s12937-015-0019-y ceutical combination on serum lipoprotein functional profile and
184. Kou S, Han B, Wang Y et al. Synergetic cholesterol-lowering ef- circulating PCSK9. Ther Clin Risk Manag 2017; 13: 1555–62.
fects of main alkaloids from Rhizoma Coptidis in HepG2 cells doi:10.2147/TCRM.S144121
and hypercholesterolemia hamsters. Life Sci 2016; 151: 50–60. 201. Spigoni V, Aldigeri R, Antonini M et al. Effects of a new nutra-
doi:10.1016/j.lfs.2016.02.046 ceutical formulation (Berberine, Red Yeast Rice and Chitosan) on
185. Su G, Wang H, Gao Y et al. ¹H-NMR-based metabonomics of the Non-HDL cholesterol levels in individuals with dyslipidemia: re-
protective effect of Coptis chinensis and berberine on cinnabar- sults from a randomized, double blind, placebo-controlled study.
induced hepatotoxicity and nephrotoxicity in rats. Molecules Int J Mol Sci 2017; 18: 1498. doi:10.3390/ijms18071498
2017; 22: 18551855. doi:10.3390/molecules22111855 202. Biagi M, Minoretti P, Bertona M et al. Effects of a nutraceutical
186. Zhao J, Zhou T, Lu J et al. Intra-herb interactions: primary metab- combination of fermented red rice, liposomal berberine, and
olites in Coptidis rhizoma extract improved the pharmacokinetics curcumin on lipid and inflammatory parameters in patients with
of oral berberine hydrochloride in mice. Front Pharmacol 2021; mild-to-moderate hypercholesterolemia: an 8-week, open-label,
12: 675368. doi:10.3389/fphar.2021.675368 single-arm pilot study. Arch Med Sci Atheroscler Dis 2018; 3:
187. Feng WW, Kuang SY, Tu C et al. Natural products berberine and 137e137–141. doi:10.5114/amsad.2018.79597
curcumin exhibited better ameliorative effects on rats with non- 203. Zhu X, Yang J, Zhu W et al. Combination of berberine with res-
alcohol fatty liver disease than lovastatin. Biomed Pharmacother veratrol improves the lipid-lowering efficacy. Int J Mol Sci 2018;
2018; 99: 325–33. doi:10.1016/j.biopha.2018.01.071 19: 3903. doi:10.3390/ijms19123903
188. Chen Y, Jiang Z, Xu J et al. Improving the ameliorative effects of 204. D’Arcy MS, Pike CVS, Coussons PJ. A novel combined resvera-
berberine and curcumin combination via dextran-coated bilosomes trol/berberine phytochemotheraputic using the HePG2 cell line as
on non-alcohol fatty liver disease in mice. J Nanobiotechnology a model for the treatment of hepatocarcinoma. Cell Biol Int 2021;
2021; 9: 230. doi:10.1186/s12951-021-00979-1 45: 2499–509. doi:10.1002/cbin.11695
189. Huseini HF, Mahmoudabady AZ, Badi NH et al. The protective 205. Bozcaarmutlu A, Sapmaz C, Bozdoğan O et al. The effect of
effect of medicinal herbs extracts including Cynara scolymus L., co-administration of berberine, resveratrol, and glibenclamide
Cichorium intybus L. Taraxacum officinal L. and Berberis vulgaris on xenobiotic metabolizing enzyme activities in diabetic rat liver.
L. in single and in combination form in CCl4 induced rat liver tox- Drug Chem Toxicol 2022; 45: 990–8. doi:10.1080/01480545.20
icity. J Med Plants 2012; 11: 78–85. 20.1802475
190. Wang XN, Han X, Xu LN et al. Enhancement of apoptosis of 206. Zhang R, Li K, Yang J et al. Effect of Rhizoma coptidis
human hepatocellular carcinoma SMMC-7721 cells through coadministration with Cortex cinnamomi on tissue distribution
synergy of berberine and evodiamine. Phytomedicine 2008; 15: of berberine in rats. Zhongguo Zhong Yao Za Zhi 2010; 35:
1062–8. doi:10.1016/j.phymed.2008.05.002 2180–3.
191. Zhou X, Ren F, Wei H et al. Combination of berberine and 207. Sengupta D, Chowdhury KD, Sarkar A et al. Berberine and S
evodiamine inhibits intestinal cholesterol absorption in high fat allyl cysteine mediated amelioration of DEN+CCl4 induced
22 Satish Sardana et al.

hepatocarcinoma. Biochim Biophys Acta 2014; 1840: 219–44. 225. Yang D, Cao J, Jiao L et al. Solubility and stability advantages
doi:10.1016/j.bbagen.2013.08.020 of a new cocrystal of berberine chloride with fumaric acid. ACS
208. Di Pierro F, Putignano P, Villanova N et al. Preliminary study Omega 2020; 5: 8283–92. doi:10.1021/acsomega.0c00692
about the possible glycemic clinical advantage in using a fixed 226. Battu SK, Repka MA, Maddineni S et al. Physicochemical charac-
combination of Berberis aristata and Silybum marianum stand- terization of berberine chloride: a perspective in the development
ardized extracts versus only Berberis aristata in patients with of a solution dosage form for oral delivery. AAPS PharmSciTech
type 2 diabetes. Clin Pharmacol: Adv Appl 2013; 5: 167–74. 2010; 11: 1466–75. doi:10.1208/s12249-010-9520-y
doi:10.2147/CPAA.S54308 227. Li Y, Yuan X, Rong X et al. Design, synthesis and biological eval-
209. Mohtashaminia F, Amini MR, Sheikhhossein F et al. Effects uation of a hybrid compound berberine and magnolol for im-
berberine-silymarin on liver enzymes: a systematic review and provement of glucose and lipid metabolism. RSC Adv 2016; 6:
meta-analysis of randomized controlled trials. Clin Nutr ESPEN 81924–31. doi:10.1039/c6ra15100k
2022; 49: 181–6. doi:10.1016/j.clnesp.2022.01.037 228. Suganthi S, Sivaraj R, Enoch IVMV. Molecular encapsulation of
210. Kong W, Wei J, Zuo Z et al. Combination of simvastatin with ber- berberine by a modified β-cyclodextrin and binding of host: guest
berine improves the lipid-lowering efficacy. Metabolism 2008; 57: complex to G-quadruplex DNA. Nucleosides Nucleotides Nucleic
1029–37. doi:10.1016/j.metabol.2008.01.037 Acids 2019; 38: 858–73. doi:10.1080/15257770.2019.1618469

Downloaded from https://academic.oup.com/rpsppr/article/2/1/rqad002/6991356 by guest on 30 March 2023


211. Mehrdoost S, Yaghmaei P, Jafary H et al. The therapeutic ef- 229. Chang S, Qin D, Yan R et al. Temperature and pH dual responsive
fects of berberine plus sitagliptin in a rat model of fatty liver nanogels of modified sodium alginate and NIPAM for berberine
disease. Iran J Basic Med Sci 2021; 24: 451–9. doi:10.22038/ loading and release. ACS Omega 2021; 6: 1119–28. doi:10.1021/
ijbms.2021.52239.11822 acsomega.0c03965
212. Lv XY, Li J, Zhang M et al. Enhancement of sodium caprate on 230. Kapoor R, Singh S, Tripathi M et al. O-hexadecyl-dextran en-
intestine absorption and antidiabetic action of berberine. AAPS trapped berberine nanoparticles abrogate high glucose stress in-
PharmSciTech 2010; 11: 372–82. doi:10.1208/s12249-010- duced apoptosis in primary rat hepatocytes. PLoS One 2014; 9:
9386-z e89124. doi:10.1371/journal.pone.0089124
213. Du HZ, Lv XY, Du GW et al. Absorption enhancement of sodium 231. Wang M, Xu R, Liu X et al. A co-crystal berberine-ibuprofen
caprate on berberine in intestine of rats. J Jilin Univ Med Ed 2011; improves obesity by inhibiting the protein kinases TBK1 and
37: 35–40. IKKɛ. Commun Biol 2022; 5: 807. doi:10.1038/s42003-022-
214. Zhang M, Lv X, Li J et al. Sodium caprate augments the hy- 03776-0
poglycemic effect of berberine via AMPK in inhibiting hepatic 232. Wang C, Jiang J, Wu W et al. The compound of mangiferin-
gluconeogenesis. Mol Cell Endocrinol 2012; 363: 122–30. berberine salt has potent activities in modulating lipid and glu-
doi:10.1016/j.mce.2012.08.006 cose metabolisms in HepG2 cells. Biomed Res Int 2016; 2016:
215. Wang J, Tanaka T, Zhang H et al. Formation and conformation of 18753436–14. doi:10.1155/2016/8753436
baicalin-berberine and wogonoside-berberine complexes. Chem 233. Liu X, Wang Y, Yun P et al. Self-assembled filomicelles prepared
Pharm Bull (Tokyo) 2012; 60: 706–11. doi:10.1248/cpb.60.706 from polylactide-poly (ethylene glycol) diblock copolymers for
216. Okoshi K, Uekusa Y, Narukawa Y et al. Solubility enhancement of sustained delivery of cycloprotoberberine derivatives. Saudi
berberine-baicalin complex by the constituents of Gardenia fruit. Pharm J 2018; 26: 342–8. doi:10.1016/j.jsps.2018.01.008
J Nat Med 2021; 75: 76–83. doi:10.1007/s11418-020-01446-1 234. Li Y, Zhu C. Development and in vitro and in vivo evaluation
217. Li Z, Liu Y, Wang J et al. Baicalin-berberine complex nanocrystals of microspheres containing sodium N-[8-(2-hydroxybenzoyl)
orally promote the co-absorption of two components. Drug amino] caprylate for the oral delivery of berberine hydrochloride.
Deliv Transl Res 2022; 12: 3017–28. doi:10.1007/s13346-022- Molecules 2020; 25: 1957. doi:10.3390/molecules25081957
01167-w 235. Chen X, Chen Y, Hou X et al. Sulfonato-β-cyclodextrin mediated
218. Huang Y, Huang K, Yang B et al. Fabrication of novel hydrogel supramolecular nanoparticle for controlled release of berberine.
with berberine-enriched carboxymethylcellulose and hyaluronic ACS Appl Mater Interfaces 2018; 10: 24987–92. doi:10.1021/
acid as an anti-inflammatory barrier membrane. Biomed Res Int acsami.8b08651
2016; 2016: 13640182–9. doi:10.1155/2016/3640182 236. Zou Q, Li J, Li Y. Preparation and characterization of vanillin-
219. Lu K, Lin Y, Lu H et al. A novel injectable in situ forming gel based cross-linked chitosan therapeutic bioactive microcarriers.
on carboxymethyl hexanoyl chitosan/hyaluronic acid polymer Int J Biol Macromol 2015; 79: 736–47. doi:10.1016/j.
blending for sustained release of berberine. Carbohydr Polym ijbiomac.2015.05.037
2019; 206: 66464–673. doi:10.1016/j.carbpol.2018.11.050 237. Yang S, Cao S, Li C et al. Berberrubine, a main metabolite of
220. Zong S, Wen H, Lv H et al. Intelligent hydrogel with both redox berberine, alleviates non-alcoholic fatty liver disease via modu-
and thermo-response based on cellulose nanofiber for con- lating glucose and lipid metabolism and restoring gut mi-
trolled drug delivery. Carbohydr Polym 2022; 278: 118943. crobiota. Front Pharmacol 2022; 13: 913378. doi:10.3389/
doi:10.1016/j.carbpol.2021.118943 fphar.2022.913378
221. Lu Q, Dun J, Chen J et al. Improving solid-state properties of ber- 238. Zhang P, Qiang X, Zhang M et al. Demethyleneberberine, a nat-
berine chloride through forming a salt cocrystal with citric acid. Int ural mitochondria-targeted antioxidant, inhibits mitochondrial
J Pharm 2019; 554: 14–20. doi:10.1016/j.ijpharm.2018.10.062 dysfunction, oxidative stress, and steatosis in alcoholic liver dis-
222. Zhao J, Zhao Q, Lu J et al. Natural nano-drug delivery system ease mouse model. J Pharmacol Exp Ther 2015; 352: 139–47.
in coptidis rhizoma extract with modified berberine hydrochlo- doi:10.1124/jpet.114.219832
ride pharmacokinetics. Int J Nanomedicine 2021; 16: 6297–311. 239. Qiang X, Xu L, Zhang M et al. Demethyleneberberine attenuates
doi:10.2147/IJN.S323685 non-alcoholic fatty liver disease with activation of AMPK and in-
223. Cheng L, Xu P, Shen B et al. Improve bile duct-targeted drug hibition of oxidative stress. Biochem Biophys Res Commun 2016;
delivery and therapeutic efficacy for cholangiocarcinoma by 472: 603–9. doi:10.1016/j.bbrc.2016.03.019
cucurbitacin B loaded phospholipid complex modified with 240. Zhang Y, Wen J, Liu D et al. Demethylenetetrahydroberberine al-
berberine hydrochloride. Int J Pharm 2015; 489: 148–57. leviates nonalcoholic fatty liver disease by inhibiting the NLRP3
doi:10.1016/j.ijpharm.2015.04.024 inflammasome and oxidative stress in mice. Life Sci 2021; 281:
224. Zhang R, Zhang Y, Zhang Y et al. Ratiometric delivery of dox- 119778. doi:10.1016/j.lfs.2021.119778
orubicin and berberine by liposome enables superior thera- 241. Li Q, Dou Y, Huang Z et al. Therapeutic effect of oxyberberine on
peutic index than Doxil Ⓡ. Asian J Pharm Sci 2020; 15: 385–96. obese non-alcoholic fatty liver disease rats. Phytomedicine 2021;
doi:10.1016/j.ajps.2019.04.007 85: 153550. doi:10.1016/j.phymed.2021.153550
Advance drug delivery and combinational drug approaches 23

242. Cao S, Yu S, Cheng L et al. 9-O-benzoyl-substituted berberine 256. Jin Y, Khadka DB, Cho W. Pharmacological effects of berberine
exerts a triglyceride-lowering effect through AMPK signaling and its derivatives: a patent update. Expert Opin Ther Pat 2016;
pathway in human hepatoma HepG2 cells. Environ Toxicol 26: 229–43. doi:10.1517/13543776.2016.1118060
Pharmacol 2018; 64: 11–7. doi:10.1016/j.etap.2018.09.007 257. Kong Y, Li L, Zhao L et al. A patent review of berberine and its
243. Cicero AFG, Rovati LC, Setnikar I. Eulipidemic effects of derivatives with various pharmacological activities (2016–2020).
berberine administered alone or in combination with other Expert Opin Ther Pat 2022; 32: 211–23. doi:10.1080/13543776.
natural cholesterol-lowering agents, A single-blind clin- 2021.1974001
ical investigation. Arzneimittelforschung 2007; 57: 26–30. 258. He Q, Mei D, Sha S et al. ERK-dependent mTOR pathway is in-
doi:10.1055/s-0031-1296582 volved in berberine-induced autophagy in hepatic steatosis. J Mol
244. Zhang H, Wei J, Xue R et al. Berberine lowers blood glucose in Endocrinol 2016; 57: 251–60. doi:10.1530/jme-16-0139
type 2 diabetes mellitus patients through increasing insulin re- 259. Lu K, Wang L, Zhang D et al. Berberine attenuates oxidative stress
ceptor expression. Metabolism 2010; 59: 285–92. doi:10.1016/j. and hepatocytes apoptosis via protecting mitochondria in blunt
metabol.2009.07.029 snout bream Megalobrama amblycephala fed high-fat diets. Fish
245. Pirro M, Francisci D, Bianconi V et al. Nutraceutical treatment for Physiol Biochem 2017; 43: 65–76. doi:10.1007/s10695-016-0268-5
hypercholesterolemia in HIV-infected patients: the NU-TRY(HIV) 260. Sun Y, Yuan X, Zhang F et al. Berberine ameliorates fatty acid-

Downloaded from https://academic.oup.com/rpsppr/article/2/1/rqad002/6991356 by guest on 30 March 2023


randomized cross-over trial. Atherosclerosis 2019; 280: 51–7. induced oxidative stress in human hepatoma cells. Sci Rep 2017;
doi:10.1016/j.atherosclerosis.2018.11.026 7: 11340. doi:10.1038/s41598-017-11860-3
246. Li HL. Observation of the clinical effects of Berberine combined 261. Sun H, Liu Q, Hu H et al. Berberine ameliorates blockade of
with Yi-gan-Ling in the treatment of metabolize syndrome with autophagic flux in the liver by regulating cholesterol metabolism
nonalcoholic steatohepatitis. Anhui Med Pharm J 2015; 19: 363– and inhibiting COX2-prostaglandin synthesis. Cell Death Dis
6. 2018; 9: 824. doi:10.1038/s41419-018-0890-5
247. Harrison SA, Gunn N, Neff GW et al. A phase 2, proof of con- 262. Sun Y, Xia M, Yan H et al. Berberine attenuates hepatic steatosis
cept, randomised controlled trial of berberine ursodeoxycholate and enhances energy expenditure in mice by inducing autophagy
in patients with presumed non-alcoholic steatohepatitis and type and fibroblast growth factor 21. Br J Pharmacol 2018; 175: 374–
2 diabetes. Nat Commun 2021; 12: 5503. doi:10.1038/s41467- 87. doi:10.1111/bph.14079
021-25701-5 263. Ma C, Shi X, Wu Y et al. Berberine attenuates atheroscle-
248. Bai RM, Zheng BB, Zhang RD. Effects of berberine on insulin rotic lesions and hepatic steatosis in ApoE -/- mice by down-
resistance and serum adiponectin of nonalcoholic fatty liver pa- regulating PCSK9 via ERK1/2 pathway. Ann Transl Med 2021; 9:
tients. Pract Geriatr 2011; 25: 423–6. 15171517.–1517. doi:10.21037/atm-20-8106
249. Xie XM, Meng XJ, Zhou XJ. The effency of Berberine in newly 264. Wang F, Zhou H, Zhao G et al. Inhibitory effects of berberine on
diagnosed type 2 diabetes mellitus with nonalcoholic fatty liver ion channels of rat hepatocytes. World J Gastroenterol 2004; 10:
disease patients and the influence of blood rheology. China J Chin 2842–285. doi:10.3748/wjg.v10.i19.2842
Mater Med 2011; 36: 3032–5. 265. Yuebin G, Yan Z, Rui L et al. Berberine regulated Gck, G6pc, Pck1
250. Cao YF, Cai-Wei W, Zhang LL et al. Clinical observation on the and Srebp-1c expression and activated AMP-activated protein ki-
Berberine plus metformin in treatment of type 2 diabetes compli- nase in primary rat hepatocytes. Int J Biol Sci 2011; 7: 673–84.
cated by nonalcoholic fatty liver disease. Mod Prevent Med 2912; doi:10.7150/ijbs.7.673
39: 4885–7. 266. Li CH, Tang SC, Wong CH et al. Berberine induces miR-373 ex-
251. Yan H, Xia M, Wang Y et al. Efficacy of berberine in patients with pression in hepatocytes to inactivate hepatic steatosis associated
non-alcoholic fatty liver disease. PLoS One 2015; 10: e0134172. AKT-S6 kinase pathway. Eur J Pharmacol 2018; 825: 107–18.
doi:10.1371/journal.pone.0134172 doi:10.1016/j.ejphar.2018.02.035
252. Chang X, Wang Z, Zhang J et al. Lipid profiling of the thera- 267. Liang H, Wang Y. Berberine alleviates hepatic lipid accumu-
peutic effects of berberine in patients with nonalcoholic fatty liver lation by increasing ABCA1 through the protein kinase C δ
disease. J Transl Med 2016; 14: 266. doi:10.1186/s12967-016- pathway. Biochem Biophys Res Commun 2018; 498: 473–80.
0982-x doi:10.1016/j.bbrc.2018.03.003
253. Yan H, Wu W, Chang X et al. Gender differences in the efficacy of 268. Saxena S, Shukla S, Kakkar P. Berberine induced modulation
pioglitazone treatment in nonalcoholic fatty liver disease patients of PHLPP2-Akt-MST1 kinase signaling is coupled with mito-
with abnormal glucose metabolism. Biol Sex Differ 2021; 12: 1. chondrial impairment and hepatoma cell death. Toxicol Appl
doi:10.1186/s13293-020-00344-1 Pharmacol 2018; 347: 92–103. doi:10.1016/j.taap.2018.03.033
254. Nejati L, Movahedi A, Salari G et al. The effect of berberine 269. Xu L, Zheng X, Wang Y et al. Berberine protects acute liver failure
on lipid profile, liver enzymes, and fasting blood glucose in pa- in mice through inhibiting inflammation and mitochondria-
tients with Non-alcoholic Fatty Liver Disease (NAFLD): a ran- dependent apoptosis. Eur J Pharmacol 2018; 819: 161–8.
domized controlled trial. Med J Islam Repub Iran 2022; 36: 39. doi:10.1016/j.ejphar.2017.11.013
doi:10.47176/mjiri.36.39 270. Zhou F, Bai M, Zhang Y et al. Berberine-induced activation of
255. Singh IP, Mahajan S. Berberine and its derivatives: a patent review AMPK increases hepatic FGF21 expression via NUR77. Biochem
(2009–2012). Expert Opin Ther Pat 2013; 23: 215–31. doi:10.15 Biophys Res Commun 2018; 495: 1936–41. doi:10.1016/j.
17/13543776.2013.746314 bbrc.2017.12.070

You might also like