Open access peer-reviewed chapter

An Overview of Genus Zanthoxylum with Special Reference to Its Herbal Significance and Application

Written By

Gyanmani Ekka, Shailesh Kumar Jadhav and Afaque Quraishi

Submitted: 07 February 2020 Reviewed: 13 April 2020 Published: 16 May 2020

DOI: 10.5772/intechopen.92459

From the Edited Volume

Herbs and Spices

Edited by Muhammad Akram and Rabia Shabir Ahmad

Chapter metrics overview

644 Chapter Downloads

View Full Metrics

Abstract

The plants of genus Zanthoxylum are effectually utilized in conventional and present-day medicine system to fight many diseases and disorders like pain, seizures, inflammation, cancer, liver and heart malady. Many of its plants—trees and shrubs, are citrus in nature, with curative antimicrobial, antihelminthic, antipyretic, and antiviral activities. More than 100 of its plant species have been identified and recorded for their potential as an herb in modern pharmacopeia. The species of this genus also have potent ethno-pharmacological significance. Many medicinal secondary metabolites like terpenoids, flavonoids, and alkaloids have also been profiled in many Zanthoxylum species. Additionally, fruit of many of the species is also significantly utilized as a major spice under the name “Sichuan pepper” in many countries like China and India. Thus, this unique blend of herb and spice characteristic of the genus needs a detailed description. This chapter highlights the major significant discoveries in the recent decade in this genus, which can add a step in the way of development of herbal medicines. Documentation of such medicinal plants may aid in derivation of plant-based medicines, which is the demand of the hour.

Keywords

  • cancer
  • herb
  • herbal
  • Sichuan pepper
  • Zanthoxylum

1. Introduction

Herbal or phyto-based medicines are in the mainstream of present pharmacological world [1]. Nearly two-thirds of the medicines throughout the world is plant based, the rest being the conventional ones [2]. The main reasons behind the preference of herbal drugs over the synthetic ones are that they have negligible side effects, are cost-effective, and easily available [3]. Also, the knowledge of herbal sources has led to the formation of base for many modern medicines. Some examples mentioned by Vickers and Zellman [4] are aspirin taken from willow bark, digoxin from foxglove, and morphine from opium poppy. Many researchers have also stated the development of resistance to allopathic drugs after a long medicament leading to becoming ineffective [5]. Thus, a documentation of medicinal plants and its herbal aspect becomes the first step toward a “botanical healing.”

Advertisement

2. Zanthoxylum genus

Zanthoxylum genus, belonging to family Rutaceae, comprises of aromatic, therapeutic deciduous shrubs and tree species [6]. Most of the plants in this genus are characterized by the presence of a strong lemon like odor, and prickly spines in the world [7]. The plant is mostly found in the South and Southeast Asian regions at an altitude of 1300–2500 m [7, 8, 9]. It is a plant of the warm temperate climate [10]. The benefits of this genus vary from healing common problems like toothache, gum problems, stomach ache, cough, cold, fever, dyspepsia, expulsion of roundworms, and in the treatment of fatal disease like cancer [11, 12, 13, 14, 15, 16]. It is an anti-inflammatory, antinociceptive, antifertility, adipogenic, hepatoprotective agent; it also has the special ability to improve speaking power, reducing rheumatism, arthritis, asthma, skin diseases, abdominal pain, anorexia, ataxia, and purifying the blood [12, 15, 17, 18, 19, 20, 21, 22, 23, 24]. The plant has phytomolecules that display insecticidal, antiparasitic, nematicidal, larvicidal, and fungicidal activities [13, 15, 25, 26, 27, 28, 29].

Advertisement

3. Zanthoxylum as herbal medicine in ancient pharmacology

Several species under Zanthoxylum genus have been used by various regions of the world since ancient times for benefits of mankind and their live stocks [5, 30, 31]. Z. liebmannianum bark is used for removal of parasites in Mexico [32]. For malaria, Z. rhoifolium, [33, 34] and Z. acutifolium [5] have been preferred from this genus. Nyishi tribe of India utilizes Z. armatum fruit, seed and bark in a traditional “Honyur” mix to treat stomach disorder, fever, and cholera, respectively [10]. Z. chiloperene var. angustifolium Engl. is also known as an antiparasitic agent in Paraguay [35]. Z. integrifolium bark is utilized by YaMei and Lanyu indigenous tribes of Taiwan, to treat dyspepsia and fever [5]. Freitas et al. [36] has reported antitumor and colitis-relief in Z. rhoifolium Lam. Z. monophyllum has found a place in the Venezulan medicine for treating jaundice, and ophthalmia [5]. Roots of Z. zanthoxyloides have been used to treat sickle cell anemia [37]. Z. alatum has been used for treatment of diabetes, toothaches, and abnormal cell growth [38].

Advertisement

4. Zanthoxylum as herbal medicine in modern pharmacology

4.1 Anticancerous activity

Z. leprieurii and Z. zanthoxyloides inhibits cancerous activity of leukemia (HL60) and (MCF7) breast cancer cell line [39]. It also shows moderate anti-cancerous activity (MCF-7), liver (WRL-68), prostrate (PC-3) and colon (CACO2) carcinoma cell lines [37]. In another study, methanolic extract (ME) of the pericarp of Z. armatum revealed the presence of compounds ZP-amide A, C, D, E, hydroxyl α and β sanshool, and Timuramide A, B, C and D [9]. All these compounds inhibited the growth of mouse glioma cells that were deficient of tumor suppressor genes NF1 homolog-Nf1, whereas only few compounds showed activity against cell lacking Trp 53—the genes responsible for encoding tumor suppressor gene p53, at a concentration that is nontoxic to the nontumor cells. Z. alatum Roxb. stem bark petroleum ether extract (PE) possesses various anticancerous lignans, namely sesamin, kobusin, and 4′0 demethyl magnolin [38]. Out of which 4′0 demethyl magnolin, which is a novel compound, gave the best anticancerous output against lungs (A549) and pancreatic (MIA-PaCa) cancer cell lines, in comparison to the standard docetaxel. Z. armatum dried root ethyl acetate extract (EAE) (a good antioxidant) and its two components flavonoids, apigenin and kaempferol-7-O-glucoside, also possess an anticancerous trait against A-549, MIA-PaCa, MCF-7, and CACO2 cancer colon cell lines [40]. In an extensive study, the ME of leaf of Z. armatum induced apoptosis in cervical cancer cell lines (HeLA) at IC50 (60 μg/mL) through Caspase 3-independent and extracellular signal-regulated kinases (ERK)-dependent mitogen-activated protein kinases (MAPK) apoptosis pathways [41]. Karmakar et al. [42] demonstrated that at an IC50 value of 102.30 μg/mL, the ME of the leaves of Z. armatum exhibited toxicity against Ehrlich Ascites cancer cells. The toxic effect was attributed to the presence of phenol and flavonoid compounds in the plant extract. Karmakar et al. [43] stated that the ME of leaves of this plant are capable of apoptosis by regulation of bcl-2/bax protein expressions and DNA damage in cancer cells and determined the presence of flavonoids, rutin, myricetin, and quercetin in the methanolic extract as potent anticancerous phytochemicals. Zanthonitrile [{4-[(3-Methyl-2-buten-1-yl) oxy] phenyl}acetonitrile] isolated from the leaves of the plant eluted by hexane: ethyl acetate solvent has a cytotoxic effect on Ehrlich Ascites Cancer cells with an IC50 value of 57.28 μg/mL [44]. Aqueous extract (AE) of Z. piperitum De Candolle fruit induces c-Jun N-terminal kinase autophagic cell death in colorectal (DLD-1), hepatocarcinoma (HepG2), and CACO2 cancer cell lines but not in A549, MCF7, and colon (WiDr) cells [45]. Alam et al. [46] demonstrated that ME and crude saponins from leaves, fruit, and bark of Z. armatum have a potential of exerting a cytotoxic effect on breast (MDA-MB-468, MCF-71) and colorectal cancer (Caco-21) cell lines using the mechanism of apoptosis. Another compound Tambulin, a flavonoid isolated from the fruit exhibited antiproliferative action on certain cancer cell lines like MCF-7, WRL-68, COLO-205, MDAMB-231, with an IC50 ranging from 37.96 to 48.7 μg/mL [45]. Three compounds isolated from Z. zanthoxyloides fruits ME, hesperidin, skimmianine, and sesamine, possess anticancerous activity up to some level against A549, MCF7, and PC3 cell lines [47]. Pang et al. [48] confirms the anti-proliferative activity of seed oil of Z. bungeanum Maxim. on melanoma (A375) cells by arresting G1 phase and inducing apoptosis. Component analysis revealed the presence of unsaturated fatty acid in the seed oil. The EAE fraction of the fruit of Z. acanthopodium has been found effective for breast cancer cell line (T47D) toxicity [49]. Another isolated compound scoparone, a coumarin from the fruit of Z. leprieurii, at an IC50 of 44.93 μg/mL can be used to design anticancerous agents against human HepG2, with the least amount of toxicity to normal Chang liver cell lines [49]. Fruit of Z. acanthopodium in n-hexane fraction is also, effectively anticancerous toward T47D cell line [50]. The possible mechanism for this is cell cycle inhibition, apoptosis induction, and downregulation of cyclin D1 activity. Geranyl acetate is present in the highest percentage in the effective fruit n-fraction.

4.2 Neuroprotectant activity

Nakamura et al. [51] has reported that Z. bungeanum could reduce scopolamine-induced dementia. Gx-50, an isolate of Z. bungeanum could also aid in Alzheimer’s disease, PE of the same plant can act as an antidepressant [52]. This compound gx-50 has the ability to cross blood–brain barrier and stop the degradation of nerve cells. Qinbunamide isolated from pericarp of Z. bungeanum can activate the nerve growth factor to further activate neurite outgrowth at 20 μM concentration [53]. Three alkaloids berberine, chelerythrine, and columbamine isolated from chloroform extract of Z. schreberi inhibit cholinesterase and butyrylcholinesterase [51]. As these enzymes are responsible for breakdown of acetyl choline, their inhibition leads to increase in number of nerve messengers, especially helpful in case of Alzheimer’s and myasthenia gravis.

4.3 Antiparasitic activity

Z. chiloperone leaves EO and one of its major component canthin-6-one, showed an inhibition of parasitic activity of Trypanosoma cruzi at 10 mg/kg of oral and subcutaneous dose in comparison to standard benznidazole (dose 50 mg/kg) [35]. ME of Z. armatum seeds, at a concentration of 50 mg/mL induced paralysis in Pheretima posthuma (test model) in lesser time than the reference drug piperazine citrate (10 mg/mL) [54]. Z. armatum methanolic leaves extract at the concentrations of 250–1000 μg/mL showed a moderate trypanocidal activity on blood parasite found both in humans and animals—Trypanosoma evansi in an in vitro condition utilizing mice cells as a model [28]. Hexane bark extract of Z. heitzii acts as an inhibitor of P. falciparum at an IC50 0.050 μg/mL [55]. Dihydronitidine one of the major components of the extract also acts as an antimalarial compound at an IC50 value of 0.0089 μg/mL [55]. In another investigation, anti-leishmanial activity was seen in crude extract and its hexane fraction of Z. armatum fruit against Leishmania major [46]. The essential oil (EO) of Z. monophyllum leaves also possess acute toxicity against larvae of Anopheles subpictus (LC50 41.50 μg/mL), Aedes albopictus (LC50 45.35 μg/mL), and Culex tritaeniorhynchus (LC50 49.01 μg/mL). Among its two major compounds Germacrene D-4-ol has better efficiency than α-Cadinol [56]. Also, The EO, along with Germacrene D-4-ol, and α-Cadinol, EO has very low toxicity against Gambusia affinis, an eradicator of malarial larvae. Costa et al. [57], has reported antiparasitic activity of three noval compounds (5,7,8-trimethoxy coumarin, syringaresinol, and dictamine, isolated from the ethanol extract (EE) of roots of Z. tingoassuiba against Leishmania amazonensis and Trypanosoma cruzi, similar to positive control benznidazole and amphotericin. The larval stage of Schistosoma haematobium, a bladder cancer causing parasite can be successfully eliminated by acridone compound arborinine, isolated from fruit of Z. leprieurii at an IC50 value of 6.98 μg/mL [57].

4.4 Antimicrobial activity

Bark EE of Z. fagara, Z. elephantiasis, and Z. martinicense presented antifungal activity against fungi prevalent in domestic animals—Aspergillus flavus, A. niger, Candida albicans, Saccharomyces cerevisiae, Microsporum canis, Trichophyton mentagrophytes [31]. Though, it was unable to inhibit bacteria like Staphylococcus aureus, Escherichia coli, Klebsiella pneumonia, and Psuedomonas aeruginosa [31]. Lipophilic leaf of Z. armatum extract was found effective against Alternaria alternata and Curvularia lunata [58]. The EE of the whole Z. armatum plant proved to be effective against S. aureus (7 mm zone of inhibition), Bacillus subtilis (23 mm the biggest zone of inhibition), B. cereus (6 mm zone of inhibition), and B. thuringiensis (1 mm zone of inhibition) [25]. Barkatullah et al. [18] reported a maximum inhibition of Micrococcus luteus, Pasteurella multocida, E. coli, and B. subtilis by the application of Z. armatum leaf EE. Z. leprieurii and Z. xanthoxyloides EO decreased the effective time required to deactivate 7log cfu/mL of Salmonella enteritidis [59]. Oxychelerythrine, a benzophenanthridine alkaloid extracted from the ME of roots of Z. capense Thunb. altered the sensitivity of S. aureus ATCC 6538 to tested antibiotics (erythromycin, oxacillin, and tetracycline) in a positive way by twofold [60]. According to Alam and Saqib [46], n-hexane, chloroform, and aqueous-methanol fraction of Z. armatum fruits are a potent antifungal source, especially against Trichophyton longifusus, Microsporum canis, A. flavus, Fusarium solani. The presence of alkaloids may be the reason behind such activity. Chen et al. [61] has found antimicrobial activity in the leaves extract of Z. bungeanum Maxim. against E. coli. K. pneumonia, P. aeruginosa, S. enteritidis, Listeria monocytogenes, C. albicans. Mirza et al. [62] observed that Z. armatum aqueous leaves extract-derived copper oxide nanomaterials (100 μl) was more sensitive in all bacterial strains tested (S. aureus, Streptococcus mutans, Streptococcus pyrogenes, Staphylococcus epidermidis, B. cereus, Corynebacterium xerosis, E. coli, K. pneumonia, P. aeruginosa, and Proteus vulgaris) in comparison to its source plant extract (100 μl).

Potent tuberculosis plant: ME of Z. leprieurii at minimum inhibitory concentrations (MIC) of 47.5, 75.3 and 125.0 μg/mL inhibited rifampicin-resistant and isoniazid-resistant strains of Mycobacterium tuberculosis, respectively [63]. Hydroxy-1,3-dimethoxy-10-methyl-9-acridone, 1-hydroxy-3-methoxy-10-methyl-9-acridone, and 3-hydroxy-1, 5,6-trimethoxy-9-acridone isolated from the plant also exhibited potent inhibition of resistant strains [63].

4.5 Antiviral activity

Z. coreanum root extract at an IC50 of 1.0 μg/mL inhibited porcine epidemic diarrhea virus growth [5]. Moreover Z. planispinum also exhibited the similar activity at an IC50 of 6.4 and 7.5 μg/mL respectively. Patino et al. [5] also reported the anti-HIV activity of Z. ailanthoides, Z. integrifoliolum, and Z. scandens.

4.6 Antioxidant activity

Oxypeucedanin, a coumarin compound present in ME of roots of Z. flavum Vahi., possesses significant antioxidant activity with an IC50 value of 8.3 μg/mL in a dichloro-dihydro-fluorescein diacetate (DCFH-DA) assay [32]. Z. armatum fruit EE showed promising activity as a natural source of antioxidants [64]. According to their study, the bioactive compound responsible for such quenching action is flavonoids, especially quercetin. Moreover, the EE of its stem bark tested via 2,2′-diphenyl picrylhydrazyl (DPPH) free radical scavenging activity exhibited significant antioxidant activities [23]. DPPH radical scavenging activity was obtained in the sequence of ME of stems (IC50 54.6 ± 2.9 μg/mL) > dichloromethane extract of stems (IC50 4.7 ± 117.5 μg/mL) > EO of fruits (IC50 5764.7 ± 6.5 μg/mL) of plant Z. limonella Alston [65]. Singh et al. [65] reported that the EO from the seeds of Z. armatum was a potent antioxidant. Ethyl acetate fraction and aqueous fraction of Z. bungeanum showed potent DPPH, 2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid (ABTS) radical scavenging abilities and ferric-reducing antioxidant power (FRAP) [66]. The EO and its constituent oleoresins showed moderate antioxidant activity when evaluated by DPPH radical scavenging, Fe2+ chelating, ferric thiocyanate method, and different lipid peroxidation assays. Leaves extract of Z. bungeanum maxim “You Huajiao” variety from China possess DPPH and ABTS radical scavenging activity. It also possesses the reducing potent power (FRAP) [61]. A compound Ombuin isolated from the fruit of Z. armatum possessed antioxidant capacity [67]. Two new sesquiterpenoid glycosides, dihydrophaseic acid 4′-O-[6″-O-(4″′-hydroxy-3″′, 5″′-dimethoxy) benzoyl)]-b-D-glucopyranoside and dihydrophaseic acid 4′-O-[6″-O-(3″′-methoxy4″′-hydroxy) benzoyl)]-b-D-glucopyranoside, isolated from the ethanolic extract of stems of Z. armatum showed moderate scavenging activity in DPPH free radical assay with IC50 values of 241 and 264 μM, respectively [68]. Aqueous extract of leaves of Z. armatum and copper oxide nanoparticles derived from it were less effective in DPPH radical scavenging activity in comparison to L-ascorbic [40].

4.7 Anti-inflammatory activity

The crude extract of Z. armatum reduced thermal pain significantly at the concentrations of 100 and 400 mg/kg body weight in case of intraperitoneal (i.p.), in comparison to 30 mg/kg body weight i.p. of standard anti-inflammatory drug phenacetin [69]. Also, its root extract exhibited analgesic activity when compared to standard drug indomethacin (40 mg/kg body weight i.p.) [69]. The anti-inflammatory activity of EE of the stem bark of Z. armatum against paw edema in Wistar rats has also been observed by Sati et al. [23]. Thus, Z. armatum may be helpful in the treatment of pain and inflammation symptoms. Eight lignans that may be responsible for this curative quality, namely eudesmin, horsfieldin, fargesin, kobusin, sesamin, asarinin, planispine A, and pinoresinol-di-3,3-dimethylallyl, were recognized by HPLC analysis in its EE of stem and root [70]. Nine terpenylated coumarins, namely 7-[(E)30,70-dimethyl-60-oxo-20,70-octadienyl]oxy-coumarin, schinilenol, schinindiol, collinin, 7-[(E)-70-hydroxy-30,70-dimethy-locta-20,50-dienyloxy]-coumarin, 8-methoxyanisocoumarin, 7-(60R-hydroxy-30,70-dimethyl-20E,70-octadienyloxy)coumarin, (E)-4-methyl-6-(coumarin-70-yloxy)hex-4-enal, and aurapten, along with a 4-quinolone alkaloid and integrifoliodiol, isolated from the leaves of Z. schinifolium by α-glucosidase inhibitory effect showed anti-inflammatory activity [71]. EO from fruits of Z. coreanum Nakai inhibited both the IgE-antigen complex and IL4 production in RBL-2H3 mast cells showing anti-inflammatory activity [60]. 2α-methyl-2β-ethylene-3β-isopropyl-cyclohexan-1β, 3α-diol and phenol-O-β-D-arabinopyranosyl-4′-(3″,7″,11″,15″-tetramethyl) hexadecan-1″-oate noval compound isolated from the methanolic extract of Z. armatum fruit exhibited anti-inflammatory activity by inhibiting pro-inflammatory cytokines like TNF-α and IL-6 in peritoneal macrophages at the concentration of 5 and 10 μg/mL [72].

4.8 Antihyperglycemic activity

Z. armatum bark ME exhibits anti-hyperglycemic activity against streptozotocin-induced diabetic rats at 200 and 400 mg/kg concentration [73]. Stem bark AE of Z. chalybeum Engl. displayed anti-hyperglycemic activity at 10, 100, and 1000 mg/kg body weight concentration against streptozotocin-induced diabetic rats [74]. n-Butanol fraction of Z. alatum EE inhibits can inhibit protein tyrosine phosphatase-1B and stimulates glucose uptake in C2CL2 myotubes in streptozotocin-induced diabetic rats [75]. Z. armatum aqueous leaves extract the activity α amylase, and α and β glucosidase, thus, can act as an antidiabetic agent [75].

4.9 Insecticidal activity

Di-chloromethane extract of Z. usambarense bark displayed insecticidal activity against Masca domestica at 5000 g/ha, but its individual component could not produce any insecticidal results [70]. The EO contained sabinene, D-germacrene, β-mycrene, β-elemene and γ-elemene. The larvicidal potential of EO and its constituent from the seeds of Z. armatum were screened against three mosquito species, Aedes aegypti, Anopheles stephensi, and Culex quinquefasciatus [15]. Among these three species, C. quinquefasciatus showed the highest sensitivity at lowest concentration LC50 and LC95 at 49 and 146 ppm, respectively. However, linalool the most important constituent present at the maximum concentration (57%) in the EO failed to establish any significant larvicidal effect individually [15]. Hieu et al. [76] revealed that mixtures of Z. armatum seed oil (ZA-SO) and its constituents either alone (0.2 mg/cm2) or as a binary mixture (0.01 + 0.99 mg/cm2) with Calophyllum inophyllum nut oil (CI-NO) can be potential repellents against stable fly, Stomoxys calcitrans. An aerosol of the mixture (ZA-SO-2.5% + CI-NO-2.5%) was also found effective as a repellent of stable fly. Among all its constituents tested, only methyl cinnamate exhibited a significant effect [77]. In a different study, three bioactive compounds—piperitone, myrtanol, and citronellal from Z. armatum seed oil were assessed for fumigant toxicity potency against stable fly with a simultaneous comparison of chlorpyrifos and dichlorvos, which are the organophosphorus insecticides [78]. The fumigant toxicity potential of seed oil and all three compounds according to the vapor phase assay was high with LC50 value 0.242–0.456 μg/cm3, but their toxic level was five magnitudes below the organophosphorus insecticides, which reflects that Z. armatum may be further used as a bio-insecticide [78]. Additionally, constituents of EO-cuminaldehyde citronellal, neral, linalool, linalool oxide, terpinen-4-ol, 1,8-cineole, and piperitone induced a significant repellent behavior in the stable fly [76]. The in vitro insecticidal efficacy of the EE of the bark of the plant has been observed against mustard aphid Lipaphis erysimi at the concentrations of 0.5, 1.0, 1.5, and 2.0 mg/L [79]. After 24 h of spray at a concentration of 2.0 mg/L, 100% mortality of the aphid was there, proving that Z. armatum may be a good insecticide [79]. Nanoencapsulated EO of Z. rhoifolium leaves efficiently reduced the number of eggs and nymphs of Bemisia tabaci at 1–5% concentration [80]. Out of 32 constituents identified from the EE of Z. armatum twigs, 1,8-cineole piperitone, and limonene in particular were efficient as an insecticide against Lasioderma serricorne and T. castaneum [81]. While, in another study, the effect of Z. armatum leaves methanolic extract has been checked for antifeedant activity on the adults of “Red flour beetle”–T. castaneum [82]. Another study stated the effects of n-hexane, EE, and ME of leaves of Z. armatum on Plutella xylostella, diamondback moth [83]. In this investigation, the n-hexane fraction exhibited the best larvicidal activity at an LC50 value of 2988.6 ppm. Moreover, two compounds, particularly 2-undecanone and 2-tridecanone identified from the n-hexane fraction of leaf extract of Z. armatum through GC–MS analysis, which may be responsible for the larvicidal activity [83]. Egg laying capacity of Bemisia tabaci, a major tomato pest, can be reduced (85–98%) by using EO of Z. rhoifolium and Z. riedelianum at 1.0–2.0% concentration [84].

4.10 Nematicidal activity

EO of Z. armatum fruit showed more than 90% nematicidal activity 5 mg/mL concentration against Bursaphelenchus xylophilus, whereas its components methyl trans cinnamate and ethyl trans cinnamate also exhibited 100% activity at 0.0625–2.0 and 0.25–2.0 mg/mL, respectively [85]. Z. armatum leaves AE (100–400 mg/kg body weight concentration) decreased the hatching ability of Meloidogyne incognita [13]. The leaves of Z. armatum also work as a nematicide on M. incognita if added directly in the soil at the concentrations of 8, 10, and 20 g/kg of soil [86].

4.11 Hepatoprotective activity

Glycoprotein isolates of Z. piperitum DC fruit inhibited hypoxanthine/xanthine oxidase [87]. It also decreased the level of lactate dehydroenase, thio barbituric acid, while increasing the level of antioxidant enzymes in carbon tetrachloride acute liver damage. The leaf EE of Z. armatum significantly decreased all the symptoms of hepatotoxicity in Wistar albino rats by normalizing the elevated levels of hepatic enzymes, which was induced by carbon tetrachloride [24]. It induced hepatoprotective activity at a concentration of 500 mg/kg of body weight in comparison to standard drug silymarin at a dose of 100 mg/kg body weight. This effect was there due to the radical scavenging activity of the phytochemicals, especially flavonoids present in the plant [24]. In a different study, the EE of bark of Z. armatum significantly expressed hepatoprotective activity at concentrations of 100, 200, and 400 mg/kg when administered orally in Wistar albino rats (where liver damage was instigated by paracetamol) by decreasing the levels of hepatic enzymes, bilirubin and at the same time increasing catalase, superoxide dismutase, and glutathione in comparison to silymarin [88]. In a recent study the ME at a dose of 500 mg/kg exhibited successful hepatoprotective activity with 66.87, 64.84, 67.95, 60.76, and 65.85% protection on aspartate aminotransferase, alanine aminotransferase, alkaline phosphatase, total bilirubin, and total protein enzyme levels of the liver, respectively in Wistar rats [89].

4.12 Gastroprotective activity

Z. rhoifolium Lam. stem bark EE prevents the formation of gastric lesions at 125–500 mg/kg dose by increasing enzymes like catalase (reduces oxidative stress) and also mucous secretion, nitric oxide (repair of gastrointestinal tract injury) [36]. It also helps in opening KATP channel to control H+ pump and acid secretion. Z. bungeanum pericarp extract reduces the level of TNF-α, IL-1β, and IL-12 to reduce the inflammation in J774.1 colon cells [90]. Thus it can be utilized to treat ulcerative colitis.

4.13 Cardiovascular activity

Z. bungeanum EO at the concentration of 5, 10, and 20 mL/kg had a significant effect on deduction of cholesterol, hyperlipidemia, triglyceride, and low-density lipoprotein; it also aided in the induction of high-density lipoprotein [91]. The EO of Z. bungeanum also helped in relaxation of contracted aortic muscles by reducing calcium influx via calcium channels [90]. Z. armatum fruits hydroethanolic extract (dose administered—200 and 400 mg/kg body weight) succeeded in decreasing the elevated levels of cardiac diagnostic marker enzymes (aspartate aminotransferase, alanine aminotransferase, lactate dehydrogenase, creatine kinase-MB, troponin-T), lipid profile and antioxidants (enzymatic and nonenzymatic), to normal conditions. The results were comparable to a positive control verapamil, expected phytochemicals are yet to be identified [92].

4.14 Antiobesity activity

In a study, methyl cinnamate, an important bioactive compound of the Z. armatum suppressed the intracellular lipid accumulation [20]. It was possible at a concentration of 25 μM through Ca2+/calmodulin-dependent protein kinase kinase 2 (CaMKK2)-phospho AMP-activated protein kinase (AMPK) signaling pathway, by downregulating the adipogenic transcription factors, sterol regulatory element binding protein-1 (SREBP-1), peroxisome proliferator-activated receptor γ (PPARγ), and CCAAT/enhancer-binding protein α (C/EBPα), as well as inhibiting the activity of PPARγ and glycerol-3-phosphate dehydrogenase (GPDH) in 3 T3-L1 preadipocyte cells [20].

Advertisement

5. Zanthoxylum as spice

“Sichuan pepper” with its pungent odor and numbing taste belongs to the genus Zanthoxylum [93]. According to Ji et al. [94] five species of Zanthoxylum—Z. armatum, Z. bungeanum, Z. shinifolim, Z. simulans, Z. piperitum, are commonly considered as “spice” species of the family. Not only seeds, fruit, leaves, bark, and even root of this genus is used as spice in China, Japan, and Korea [95]. This pepper is used by the people of China to create a special “Mala” flavor, which literally refers to numbness and spiciness. More than hundred volatile compounds have been isolated, which are responsible for the unique spicy note and fragrance of this genus [96]. Some nonvolatile compounds have also been identified like alkylamides (sanshools, capsaicin) and polyphenolic compounds [97]. The genus has found a place in culinary as “five spice powder,” in cosmetics, as well as in pharmaceutical too [98]. In cosmetics, the variable fragrance provided by the genus—sweet, spearmint, herbal, floral, fruity, rose, citrus—is of great significance [99]. This diversity is due to the presence of more than hundred volatile compounds like 1,8-cineole, 1-terpineol, 2,-nonenal, 2-tridecanone, α-elemol, α-pinene, β-pinene, geraniol, myrcene, neryl acetate, piperitone, rosefuran etc. [100]. The pungent property of the spice of this genus has been attributed to the presence of α-, β-, γ-δ-sanshool and hydroxyl α and β-sanshool [99]. In pharmacology the presence of polyphenolic compounds like flavonoids and glycosides make Sichuan pepper a good antioxidant and anti-inflammatory agent. Zhu et al. [98] has also reported the presence of antibacterial activity against both gram positive and negative bacteria in EO of Z. bungeanum fruit. EO of Z. shinifolium and Z. piperitum (broad spectrum) has been reported to have antiviral properties [94]. Most significantly, Sichuan pepper and its component sanshool amide have displayed inhibitory action on the formation of hetercyclic amines, which are carcinogenic to humans in beef grilling procedure [11, 101]. Utilization of analytical techniques to compare component analysis is lacking in the literature related to spice knowledge of the genus. Also according to Ji et al. [94] if the effect of heat (while cooking) is elaborated on the spice and its individual compound, it would be more beneficial to the herb and spice world.

Advertisement

6. Conclusion and future perspective

Zanthoxylum genus is a stockpile of medicinal plants brewing with therapeutic properties, as gathered from the above references of the recent decade. The readers can benefit with the traditional and current knowledge on the herbal aspect of several species Z. acanthopodium, Z. acutifolium, Z. ailanthoides, Z. armatum, Z. bungeanum, Z. chalybeum, Z. chiloperone, Z. coreanum, Z. elephantiasis, Z. fagara, Z. flavum, Z. heitzii, Z. integrifoliolum, Z. leprieurii, Z. liebmannianum, Z. limonella, Z. martinicense, Z. monophyllum, Z. piperitum, Z. planispinum, Z. rhoifolium, Z. riedelianum, Z. scandens, Z. schinifolium, Z. schreberi, Z. tingoassuiba, Z. usambarense, Z. zanthoxyloides to name a few, of this genus. The information can form the basis of research regarding drug formulations, conservation of medicinal plants, pharmacokinetics, and new drug discoveries, which are plant based in the future.

References

  1. 1. Kamboj VP. Herbal medicine. Current Science. 2000;78:35-39
  2. 2. Newman DJ, Cragg GM. Natural products as sources of new drugs over the last 25 years. Journal of Natural Products. 2007;70:461-477
  3. 3. Pal S, Shukla Y. Herbal medicine: Current status and the future. Asian Pacific Journal of Cancer Prevention. 2002;4:281-288
  4. 4. Vickers A, Zollman C. ABC of complementary medicine: Herbal medicine. British Medical Journal. 1999;319:1050-1053
  5. 5. Patino OJ, Prieto JA, Cuca LE. Zanthoxylum genus as potential source of bioactive compounds. In: Rasooli I, editor. Bioactive Compounds in Phytomedicine. London, United Kingdom: IntechOpen; 2012. DOI: 10.5772/26037
  6. 6. Ahmad A, Misra LN, Gupta MM. Hydroxalk-(4Z)-enoic acids and volatile components from the seeds of Zanthoxylum armatum. Journal of Natural Products. 1993;56:456-460
  7. 7. Datt G, Chauhan JS, Ballabha R. Influence of pre-sowing treatments on seed germination of various accessions of Timroo (Zanthoxylum armatum DC.) in the Garhwal Himalaya. Journal of Applied Research on Medicinal and Aromatic Plants. 2017;7:89-94
  8. 8. Alam F, Saqib QN. Pharmacognostic study and development of quality control parameters for fruit, bark and leaf of Zanthoxylum armatum (Rutaceae). Ancient Science of Life. 2015;34:147-155
  9. 9. Devkota KP, Wilson J, Henrich CJ, McMahon JB, Reilly KM, Beutler JA. Isobutylhydroxyamides from the pericarp of Nepalese Zanthoxylum armatum inhibit NF1-defective tumor cell line growth. Journal of Natural Products. 2013;76:59-63
  10. 10. Shankar R, Rawat MS. Conservation and cultivation of threatened and high valued medicinal plants in north East India. International Journal of. Biodiversity and Conservation. 2013;5:584-591
  11. 11. Kalia NK, Singh B, Sood RP. A new amide from Zanthoxylum armatum. Journal of Natural Products. 1999;62:311-312
  12. 12. Kumar V, Kumar S, Singh B, Kumar N. Quantitative and structural analysis of amides and lignans in Zanthoxylum armatum by UPLC-DAD-ESI-QTOF-MS/MS. Journal of Pharmaceutical and Biomedical Analysis. 2014;94:23-29
  13. 13. Mukhtar T, Kayani MZ, Hussain MA. Nematicidal activities of Cannabis sativa L. and Zanthoxylum alatum Roxb. Against Meloidogyn incognita. Industrial Crops and Products. 2013;42:447-453
  14. 14. Thakur T, Trak TH, Tripathi J. Ethnomedicinal use of some of the plants of Udhampur district of Jammu and Kashmir, India. Indian Journal of Applied and Pure Biology. 2017;32:293-300
  15. 15. Tiwary M, Naik SN, Tewary DK, Mittal PK, Yadav S. Chemical composition and larvicidal activities of the essential oil of Zanthoxylum armatum DC. (Rutaceae) against three mosquito vectors. Journal of Vector Borne Diseases. 2007;44:198-204
  16. 16. Waheed A, Mahmud S, Akhtar M, Nazir T. Studies on the components of essential oil of Zanthoxylum armatum by GC-MS. American Journal of Analytical Chemistry. 2011;2:258-261
  17. 17. Barkatullah IM, Muhammad N. Evaluation of Zanthoxylum armatum DC for in vitro and in vivo pharmacological screening. African Journal of Pharmacy and Pharmacology. 2011;5:1718-1723
  18. 18. Barkatullah IM, Muhammad N, Tahir L. Antimicrobial evaluation, determination of total phenolic and flavonoid contents in Zanthoxylum armatum DC. Journal of Medicinal Plant Research. 2012;6:2105-2110
  19. 19. Bhatia H, Sharma YP, Manhas RK, Kumar K. Ethnomedicinal plants used by the villagers of district Udhampur, J & K. Journal of Ethnopharmacology. 2014;151:1005-1018
  20. 20. Chen Y, Lee M, Hsu C, Wei C, Tsai Y. Methyl cinnamate inhibits adipocyte differentiation via activation of the caMKK2-AMPK pathway in 3T3 L1 preadipocytes. Journal of Agricultural and Food Chemistry. 2012;60:955-963
  21. 21. Fatima A, Ahmad M, Zafar M, Yaseen G, Khan MPZ, Butt MA, et al. Ethnopharmacological relevance of medicinal plants used for treatment of oral diseases in Central Punjab-Pakistan. Journal of Herbal Medicine. 2017;12:88-110
  22. 22. Guo T, Deng Y, Xie H, Yao C, Cai C, Pan S. Antinociceptive and anti- inflammatory activities of ethyl acetate fraction from Z. armatum in mice. Fitoterapia. 2011;82:347-351
  23. 23. Sati SC, Sati MD, Raturi R, Badoni P, Singh H. Anti-inflammatory and antioxidant activities of Zanthoxylum armatum stem bark. Global Journal of Engineering Research. 2011;11:19-21
  24. 24. Verma N, Khosa RL. Hepatoprotective activity of leaves of Z. armatum DC. in CCL4 induced hepatotoxicity in rats. Indian Journal of Biochemistry and Biophysics. 2010;47:124-127
  25. 25. Joshi B, Lekhak S, Sharma A. Antibacterial property of different medicinal plants: Ocimum sanctum, Cinnamomum zeylanicum, Xanthoxylum armatum, Origanum majorana. Kathmandu University Journal of Science, Engineering and Technology. 2009;5:143-150
  26. 26. Ramanujam SN, Ratha BK. Effect of alcohol extract of a natural piscicide – Fruits of Zanthoxylum armatum DC. On Mg2+ − and Na+, K+ ATPase activity in various tissues of a freshwater air breathing fish, Heteropneustes fossilis. Aquaculture. 2008;283:77-82
  27. 27. Raturi R, Badoni PP, Ballabha R. Insecticidal and fungicidal activities of stem bark of Zanthoxylum armatum (Rutaceae). World Journal of Pharmacy and Pharmaceutical Sciences. 2014;3:1838-1843
  28. 28. Shaba P, Pandey NN, Sharma OP, Rao JR, Singh RK. Therapeutic effects of Zanthoxylum alatum leaves and Eugenia caryophyllata buds (fruits) against Trypanosoma evansi. Journal of Animal and Veterinary Advances. 2012;2:91-97
  29. 29. Xu D, Zhuo Z, Wang R, Ye M, Pu B. Modeling the distribution of Zanthoxylum armatum in China with maxEnt modeling. Global Ecology and Conservation. 2019;19:e00691
  30. 30. Adesina SK. The Nigerian Zanthoxylum: Chemical and biological values. African Journal of Traditional, Complementary, and Alternative Medicines. 2005;2:282-301
  31. 31. Dieguez-Hurtado R, Garrido-Garrido G, Prieto-Gonzalez S, Iznaga Y, Gonzalez L, Molina-Torres J, et al. Antifungal activity of some Cuban Zanthoxylum species. Fitoterapia. 2003;74:384-386
  32. 32. Ross SA, Sultana GNN, Burandt CL, ElSohly MA, Marais JPJ, Ferreira D. Syncaparmide, a new antiplasmodial (+)-norepinephrine derivative from Zanthoxylum syncarpum. Journal of Natural Products. 2004;67:88-90
  33. 33. Bertani S, Bourdy G, Landau I, Robinson JC, Esterre PH, Deharo E. Evaluation of French Guiana traditional antimalarial remedies. Journal of Ethnopharmacology. 2005;98:45-54
  34. 34. Jullian V, Bourdy G, Georges S, Maurel S, Sauvain M. Validation of use of a traditional remedy from French Guiana, Zanthoxylum rhoifolium lam. Journal of Ethnopharmacology. 2006;106:348-352
  35. 35. Ferreira ME, Cebrian-Torrejon G, Corrales AS, Vera De Bilbao N, Rolon M, Gomez CV, et al. Zanthoxylum chiloperone leaves extract: First sustainable Chagas disease treatment. Journal of Ethnopharmacology. 2011;133:986-993
  36. 36. Freitas FFBP, Fernandes HB, Piauilino CA, Pereira SS, Carvalho KIM, Chaves MH, et al. Gastroprotective activity of Zanthoxylum rhoifolium lam in animal models. Journal of Ethnopharmacology. 2011;137:700-708
  37. 37. Misra LN, Wouatsa NAV, Kumar S, Kumar V, Tchoumbougnang F. Antibacterial, cytotoxic activities and chemical composition of fruits of two Cameroonian Zanthoxylum species. Journal of Ethnopharmacology. 2013;148:74-80
  38. 38. Mukhija M, Lal Dhar K, Nath Kalia A. Bioactive Lignans from Zanthoxylum alatum Roxb stem bark with cytotoxic potential. Journal of Ethnopharmacology. 2014;152:106-112
  39. 39. Choumessi AT, Loureiro R, Silva AM, Moreira AC, Pieme AC, Tazoacha A, et al. Toxicity evaluation of some traditional African spices on breast cancer cells and isolated rat hepatic mitochondria. Food and Chemical Toxicology. 2012;50:4199-4208
  40. 40. Mukhija M, Singh MP, Dhar KL, Kalia AN. Cytotoxic and antioxidant activity of Zanthoxylum alatum stem bark and its flavonoid constituents. Journal of Pharmacognosy and Phytochemistry. 2015;4:86-92
  41. 41. Singh T, Meitei H, Sharma A, Robinson A, Singh L, Singh T. Anticancer properties and enhancement of therapeutic potential of cisplatin by leaf extract of Zanthoxylum armatum DC. Biological Research. 2015;48:46-55
  42. 42. Karmakar I, Haldar S, Chakraborty M, Dewanjee S, Haldar PK. Antioxidant and cytotoxic activity of different extracts of Zanthoxylum alatum. Free Radicals and Antioxidants. 2015;5:21-28
  43. 43. Karmakar I, Haldar S, Chakraborty M, Chaudhury K, Dewanjee S, Haldar PK. Regulation of apoptosis through bcl-2/bax proteins expression and DNA damage by Zanthoxylum alatum. Pharmaceutical Biology. 2016a;54:503-508
  44. 44. Karmakar I, Haldar S, Chakraborty M, Dewanjee S, Haldar PK. In vitro antioxidant and cytotoxic activity of Zanthonitrile isolated from Zanthoxylum alatum. Journal of Applied Pharmaceutical Science. 2016b;6:119-122
  45. 45. Nozaki R, Kono T, Bochimoto H, Watanabe T, Oketani K, Sakamaki Y, et al. Zanthoxylum fruit extract from Japanese pepper promotes autophagic cell death in cancer cells. Oncotarget. 2016;7:70437-70446
  46. 46. Alam F, Saqib QN. Evaluation of Zanthoxylum armatum Roxb for in vitro biological activities. Journal of Traditional and Complementary Medicine. 2017;7:515-518
  47. 47. Guetchueng ST, Nahar L, Ritchie KJ, Ismail FMD, Evans AR, Sarker SD. Zanthoamides G-I: Three new alkamides from Zanthoxylum zanthoxyloides. Phytochemistry Letters. 2018;26:125-129
  48. 48. Pang W, Liu S, He F, Li X, Saira B, Zheng T, et al. Anticancer activities of Zanthoxylum bungeanum seed oil on malignant melanoma. Journal of Ethnopharmacology. 2018;229:180-189
  49. 49. Satria D, Silalahi J, Haro G, Ilyas S, Hasibuan PAZ. Chemical analysis and cytotoxic activity of N-hexane fraction of Zanthoxylum acanthopodium DC fruits. Rasayan Journal of Chemistry. 2019;12:803-808
  50. 50. Satria D, Silalahi J, Haro G, Ilyas S, Hasibuan PAZ. Cell cycle inhibition of ethylacetate fraction of Zanthoxylum acanthopodium DC fruit against T47D cells. Open Access Macedonian Journal of Medical Sciences. 2019;7:726-729
  51. 51. Nakamura T, Komai N, Isogami I, Ueno K, Ikegami F, One K. Memory and learning-enhancing effect of Daikenchuto, a traditional Japanese herbal medicine, in mice. Journal of Natural Medicines. 2006;60:64-67
  52. 52. Chen SF, Li LF, Chen J, Wang XT, Huang HJ. The antidepressant effect of procyanidins extract from Zanthoxylum bungeanum maxim. On ovariectomized model mice. Journal of Wenzhou Medical University. 2015;45:260-264
  53. 53. Tian JM, Wang Y, Xu YZ, Yu ZC, Wei AZ, Zhang WM, et al. Characterization of isobutylhydroxyamides with NGF-potentiating activity from Zanthoxylum bungeanum. Bioorganic & Medicinal Chemistry Letters. 2016;26:338-342
  54. 54. Mehta DK, Das R, Bhandari A. In vitro anthelmintic activity of seeds of Zanthoxylum armatum DC. Against Pheretima posthuma. International Journal of Green Pharmacy. 2012;6:26-28
  55. 55. Goodman CD, Austarheim I, Mollard V, Mikolo B, Malterud KE, McFadden GI, et al. Natural products from Zanthoxylum heitzii with potent activity against the malaria parasite. Malaria Journal. 2016;15:1-8
  56. 56. Pavela R, Govindarajan M. The essential oil from Zanthoxylum monophyllum a potential mosquito larvicide with low toxicity to the non-target fish Gambusia affinis. Journal of Pest Science. 2017;90:369-378
  57. 57. Costa RS, Souza Filho OP, Junior OCSD, Silva JJ, Le Hyaric M, Santos MAV, et al. In vitro antileishmanial and antitrypanosomal activity of compounds isolated from the roots of Zanthoxylum tingoassuiba. Brazilian Journal of Pharmacognosy. 2018;28:551-558
  58. 58. Guleria S, Kumar A. Antifungal activity of some Himalayan medicinal plants using direct bioautography. Journal of Cell and Molecular Biology. 2006;5:95-98
  59. 59. Kamdem SSL, Belletti N, Tchoumbougnang F, Essia-Ngang JJ, Montanari C, Tabanelli G, et al. Effect of mild heat treatments on the antimicrobial activity of essential oils of Curcuma longa, Xylopia aethiopica, Zanthoxylum xanthoxyloides and Zanthoxylum leprieurii against Salmonella enteritidis. Journal of Essential Oil Research. 2015;27:52-60
  60. 60. Cabral V, Luo X, Junqueira E, Costa SS, Mulhovo S, Duarte A, et al. Enhancing activity of antibiotics against Staphylococcus aureus: Zanthoxylum capense constituents and derivatives. Phytomedicine. 2015;22:469-476
  61. 61. Chen X, Wang W, Wang C, Liu Z, Sun Q, Wang D. Quality evaluation and chemometric discrimination of Zanthoxylum bungeanum maxim leaves based on flavonoids profiles, bioactivity and HPLC-fingerprint in a common garden experiment. Industrial Crops and Products. 2019;134:225-233
  62. 62. Mirza A, Kareem A, Nami S, Rehman S, Bhat S, Nishat N, et al. Copper oxide nanomaterials derived from Zanthoxylum armatum DC and Berberis lycium Royle plant species: Characterization, assessment of free radical scavenging and antibacterial activity. Chemistry & Biodiversity. 2019;16:e1900145
  63. 63. Bunalema L, Fotso GW, Waako P, Tabuti J, Yeboah SO. Potential of Zanthoxylum leprieurii as a source of active compounds against drug resistant Mycobacterium tuberculosis. BMC Complementary and Alternative Medicine. 2017;17:4-9
  64. 64. Batool F, Sabir SM, Rocha JBT, Shah AH, Saify ZS, Ahmed SD. Evaluation of antioxidant and free radical scavenging activities of fruit extract from Zanthoxylum alatum: A commonly used spice from Pakistan. Pakistan Journal of Botany. 2010;42:4299-4311
  65. 65. Singh G, Kapoor IPS, Singh P, Heluani C, Lampasona M, Catalan CAN. Chemistry and antioxidant properties of essential oil and oleoresins extracted from the seeds of Tomer (Zanthoxylum armatum DC.). International Journal of Food Properties. 2013;16:288-300
  66. 66. Zhang YJ, Wang DM, Yang LN, Zhou D, Zhang JF. Purification and characterization of flavonoids from the leaves of Zanthoxylum bungeanum and correlation between their structure and antioxidant activity. PLoS One. 2014;9:e105725
  67. 67. Nooreen Z, Kumar A, Bawankule DU, Tandon S, Ali M, Xuan TD, et al. New chemical constituents from the fruits of Zanthoxylum armatum and its in vitro anti-inflammatory profile. Natural Product Research. 2019;33:665-672
  68. 68. Liu X, Xu L, Liu L, Wang Y, Zhao Y, Kang Q, et al. Combination of essential oil from Zanthoxylum bungeanum maxim. And a microemulsion system: Permeation enhancement effect on drugs with different lipophilicity and its mechanism. Journal of Drug Delivery Science and Technology. 2019;55:101309
  69. 69. Kaur V, Kumar T, Bora SK. Pharmacological evaluation of Zanthoxylum armatum root extract on analgesic and anti-inflammatory activity. Journal of Pharmacy Research. 2011;4:2561-2562
  70. 70. He W, Puyvelde LV, Kimpe ND, Verbruggen L, Anthonissen K, Flaas MVD, et al. Chemical constituents and biological activities of Zanthoxylum usambarense. Phototherapy Research. 2002;16:66-70
  71. 71. Nguyen PH, Zhao BT, Kim O, Le JH, Choi JS, Min BS, et al. Anti-inflammatory terpenylated coumarins from the leaves of Zanthoxylum schinifolium with α-glucosidase inhibitory activity. Journal of Natural Medicines. 2016;70:276-281
  72. 72. Nooreen Z, Singh S, Singh DK, Tandon S, Ahmad A, Luqman S. Characterization and evaluation of bioactive polyphenolic constituents from Zanthoxylum armatum DC., a traditionally used plant. Biomedicine and Pharmacotherapy. 2017;89:366-375
  73. 73. Karki H, Upadhayay K, Pal H, Singh R. Antidiabetic potential of Zanthoxylum armatum bark extract on streptozotocin-induced diabetic rats. International Journal of Green Pharmacy. 2014;8:77-83
  74. 74. Kimani CN, Mbaria JM, Suleiman M, Gakuya D, Kiama SG. Antihyperglycemic activity of Zanthoxylum chalybeum stem bark extract in diabetic rats. Journal of Pharmacognosy. 2015;4:183-189
  75. 75. Rynjah CV, Devi NN, Khongthaw N, Syiem D, Majaw S. Evaluation of the antidiabetic property of aqueous leaves extract of Zanthoxylum armatum DC. Using in vivo and in vitro approaches. Journal of Traditional and Complementary Medicine. 2018;8:134-140
  76. 76. Hieu TT, Kim S, Kwon HW, Ahn Y. Enhanced repellency of binary mixture of Zanthoxylum piperitium pericrap steam distillate or Zanthoxylum armatum seed oil constituents and Calophyllum inophyllum nut oil and their aerosols to Stomyxs calcitrans. Pest Management Science. 2010;66:1191-1198
  77. 77. Hieu TT, Jung J, Kim S, Ahn Y, Kwon HW. Behavioural and electroantennogram response of the stable fly (Stomoxys calcitrans L.) to plant essential oils and their mixtures with attractants. Pest Management Science. 2014;70:163-172
  78. 78. Hieu TT, Kim S, Ahn Y. Toxicity of Zanthoxylum piperitum and Zanthoxylum armatum oil constituents and related compounds to Stomoxys calcitrans. Journal of Medical Entomology. 2012;49:1084-1091
  79. 79. Ross SA, Al-Azeib MA, Krishnavei KS, Fronczek FR, Burandt CL. Alkamides from the leaves of Zanthoxylum syncarpum. Journal of Natural Products. 2005;68:1297-1299
  80. 80. Christofoli M, Costa ECC, Bicalho KU, de Cassia DV, Peixoto MF, Alves CCF, et al. Insecticidal effect of nanoencapsulated essential oils from Zanthoxylum rhoifolium (Rutaceae) in Bemisia tabaci populations. Industrial Crops and Products. 2015;70:301-308
  81. 81. Wang C, You C, Yang K, Guo S, Geng Z, Fan L, et al. Antifeedant activities of methanol extracts of four Zanthoxylum species and benzophenanthridines from stem bark of Zanthoxylum schinifolium against Tribolium castaneum. Industrial Crops and Products. 2015b;74:407-411
  82. 82. Wang C, Zhang W, You C, Guo S, Geng Z, Fan L, et al. Insecticidal constituents of essential oil derived from Zanthoxylum armatum against two stored product insects. Journal of Oleo Science. 2015a;64:861-868
  83. 83. Kumar V, Reddy SGE, Chauhan U, Kumar N, Singh B. Chemical composition and larvicidal activity of Zanthoxylum armatum against diamondback moth Plutella xylostella. Natural Product Research. 2016;30:689-692
  84. 84. Costa ECC, Christofoli M, de Costa GCS, Peixoto MF, Fernandes JB, Forim MR, et al. Essential oil repellent action of plants of the genus Zanthoxylum against Bemisia tabaci biotype B. (Homoptera: Aleyrodidae). Scientia Horticulturae. 2017;226:327-332
  85. 85. Kim J, Seo S, Park K. Nematicidal activity of plant essential oil and components from Gaultheria fragrantissima and Zanthoxylum alatum against the pine wood nematode, Bursaphelenchus xylophilus. Nematology. 2011;13:87-93
  86. 86. Kayani MZ, Mukhtar T, Hussain MA. Evaluation of nematicidal effects of Cannabis sativa L. and Zanthoxylum alatum Roxb. Against root-knot nematodes, Meloidogyne incognita. Crop Protection. 2012;39:52-56
  87. 87. Lee SJ, Lim KT. Glycoprotein of Zanthoxylum piperitum DC has a hepatoprotective effect via anti-oxidative character in vivo and in vitro. Toxicology In Vitro. 2008;22:376-385
  88. 88. Ranawat LS, Patel J. Antioxidant and hepatoprotective activity of ethanolic extracts of bark of Zanthoxylum armatum DC in paracetamol-induced hepatotoxicity. International Journal of Pharmaceutical Sciences and Drug Research. 2013;5:120-124
  89. 89. Talluri MR, Gummadi VP, Battu GR, Killari KN. Evaluation of Hepatoprotective activity of Zanthoxylum armatum on paracetamol-induced liver toxicity in rats. Indian Journal of Pharmaceutical Sciences. 2018;81:138-145
  90. 90. Zhang Z, Liu J, Shen P, Cao Y, Lu X, Gao X, et al. Zanthoxylum bungeanum pericarp extract prevents dextran sulfate sodium-induced experimental colitis in mice via the regulation of TLR4 and TLR4-related signaling pathways. International Immunopharmacology. 2016;41:127-135
  91. 91. Heng L, Li C, Jia M, Yao XJ, Mei QB. Study on therapeutic effects of seed soil of Zanthoxylum bungeanum maxim. On experimental hyperlipidemia in rat. Chinese Medical Journal. 2005;30:1012-1013
  92. 92. Mangalanathan M, Devendhiran T, Uthamaramasamy S, Kumarasamy K, Mohanraj K, Devendhiran K, et al. Efficacy of Zanthoxylum armatum fruit on isoproterenol induced myocardial infarction in rats. South Asian Journal of Engineering and Technology. 2019;8:4-11
  93. 93. Jiang L, Kubota K. Formation by mechanical stimulus of the flavor compounds in young leaves of Japanese pepper (Xanthoxylum piperitum DC). Journal of Agricultural and Food Chemistry. 2001;49:1353-1357
  94. 94. Ji Y, Li S, Ho C. Chemical composition, sensory properties and application of Sichuan pepper (Zanthoxylum genus). Food Science and Human Wellness. 2019;8:115-125
  95. 95. Wu G, Wu H. Analgesia synergism of essential oil from pericarp of Zanthoxylum schinifolium and verapamil. Evidence-based Complementary and Alternative Medicine. 2014;2014:505876
  96. 96. Oh M, Chung MS. Effects of oils and essential oils from seeds of Zanthoxylum schinifolium against foodborne viral surrogates. Evidence-Based Complementary and Alternative Medicine. 2014;2014:1-6
  97. 97. Ravindran PN, Divakaran M, Pillai GS. Other herbs and spices: Achiote to Szechuan pepper. In: Handbook of Herbs and Spices. Vol. 2. Cambridge, United Kingdom: Woodhead Publishing Series in Food Science, Technology and Nutrition; 2012. pp. 534-556
  98. 98. Zhu RX, Zhong K, Zeng WC, He XY, Gu XQ, Zhao ZF, et al. Essential oil composition and antibacterial activity of Zanthoxylum bungeanum. African Journal of Microbiology Research. 2011;5:4631-4637
  99. 99. Sugai E, Morimitsu Y, Kubota K. Quantitative analysis of sanshool compounds in Japanese pepper (Xanthoxylum piperitum DC.) and their pungent characteristics. Bioscience, Biotechnology, and Biochemistry. 2005a;69:1958-1962
  100. 100. Sugai E, Morimitsu Y, Iwasaki Y, Morita A, Watanabe T, Kubota K. Pungent qualities of sanshool-related compounds evaluated by a sensory test and activation of rat TRPV1. Bioscience, Biotechnology, and Biochemistry. 2005b;69:1951-1957
  101. 101. Zeng M, Wang J, Zhang M, Chen J, He Z, Qin F, et al. Inhibitory effects of Sichuan pepper (Zanthoxylum bungeanum) and sanshoamide extract on heterocyclic amine formation in grilled ground beef patties. Food Chemistry. 2018;239:111-118

Written By

Gyanmani Ekka, Shailesh Kumar Jadhav and Afaque Quraishi

Submitted: 07 February 2020 Reviewed: 13 April 2020 Published: 16 May 2020