You are on page 1of 11

Journal of Herbal Medicine 11 (2018) 1–11

Contents lists available at ScienceDirect

Journal of Herbal Medicine


journal homepage: www.elsevier.com/locate/hermed

Review article

Nutritional and medicinal applications of Moringa oleifera Lam.—Review of T


current status and future possibilities

Swati Guptaa, Rohit Jaina, Sumita Kachhwahab, S.L. Kotharic,
a
Department of Biosciences, Manipal University Jaipur, India
b
Department of Botany, and Bioinformatics Infrastructure Facility, University of Rajasthan, Jaipur, India
c
Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, India

A R T I C L E I N F O A B S T R A C T

Keywords: Moringa oleifera Lam. (vern. Drumstick) is a mineral rich, medicinally important tree species of family
Moringa oleifera Moringaceae. It has a wide range of culinary applications and has bioremediation, nutritional and medicinal
Alzheimer’s properties. Conventionally, seeds of the tree have been used as natural coagulants and flocculents in waste water
Cancer treatment. Its leaves and bark act as biosorbent for remediation of heavy metals and dyes. The tree provides
Moringa gum
remedies for a range of diseases and disorders by dint of its unique combination of numerous phytochemicals.
Antioxidant
Gum exudates of the tree are of immense medicinal importance due to their applications in biodegradable drug
delivery systems and in treatment of asthma, dysentery and intestinal cancer. The multidimensional utilities of
M. oleifera may cause overexploitation of this tree, posing danger to the existing natural variability in the near
future. Therefore, there is a need for conservation of the species for ethnobotanical, pharmacological, nu-
traceutical and biodiversity purposes. Development of tissue culture propagation methods will assist in pre-
serving some of the germplasm of the species. In the present review, consolidated analysis of the role of bio-
technology in conservation and genetic enhancement of nutritional, medicinal and commercial value of the tree
has been placed in perspective, together with an up to date review on phytochemical analysis of the plant, and its
utility has been discussed to invite the attention of the scientific community to further consider the study of this
miracle tree species and its nutritional and pharmaceutical properties.

1. Introduction Moringa oleifera Lam. (syn. M. pterygosperma Gaertn.) is a minerals


and vitamin rich, nutritious and medicinally important tree species of
Plants provide the richest resource of natural compounds, which are the Hindustan center of crop origin, belonging to the family
used directly or indirectly for a wide range of applications for the Moringaceae, and commonly called Drumstick, Ben Oil or Horseradish
wellbeing of human population and domestic animals (Khanuja, 2012). tree (Arora et al., 2013; Ramachandran et al., 1980).
These compounds are primarily phytomolecules synthesized through Moringaceae is a monogeneric family including the single genus
primary and secondary metabolic pathways in plants (Arora et al., Moringa with 10–12 species. The plant M. oleifera is characterized by
2013). tripinnate leaves, yellow or white petioles, hanging 3-sided pods and
The dependence of humans on plants for their basic requirements whitish grey corky bark (Fig. 1a–c). Other characteristics include bi-
like food, medicine, clothes and shelter is as old as mankind itself sexual, stalked, white or creamy axillary flowers, globular winged
(Goyal et al., 2007), and still in the modern age the majority of com- seeds, pendulous ribbed capsules and soft, fissured tuberous taproots
mercial products including pharmaceutical and healthcare, food and (Bhandari, 1995; Ramachandran et al., 1980; Seshadri and Nambiar,
beverages, textiles, cosmetics and aromas are obtained from plants 2003). The tree also exudes gum through lysigenous traumatic gum
(Khanuja, 2012). Therefore, plants are and will remain economically, ducts developed in the bark (Bhandari, 1995; Subrahmanyam and Shah,
industrially, environmentally, spiritually, historically, and aesthetically 1988). The shape and color of the gum exudates varies from stalactite
important for survival, sustenance and prosperity of life on the Earth pieces to tears and yellow to reddish brown or black (Fig. 1d) in color
(Arora et al., 2013). (Bhandari, 1995; Morton, 1991; Panda et al., 2006; Ramachandran

Abbreviations: Kn, Kinetin; BAP, 6-benzylaminopurine; IBA, Indole-3-butyric acid; IAA, Indole-3-acetic acid; NAA, α-naphthaleneacetic acid; TDZ, Thidiazuron; 2,4-D, 2,4-di-
chlorophenoxyacetic acid; TRIA, triacontanol; PGR(s), plant growth regulator(s); MS, Murashige and Skoog

Corresponding author.
E-mail address: slkothari28@gmail.com (S.L. Kothari).

http://dx.doi.org/10.1016/j.hermed.2017.07.003
Received 1 November 2016; Received in revised form 17 March 2017; Accepted 31 July 2017
Available online 02 August 2017
2210-8033/ © 2017 Elsevier GmbH. All rights reserved.
S. Gupta et al. Journal of Herbal Medicine 11 (2018) 1–11

Fig 1. Moringa oleifera Lam. a. Tree growing in natural habitat. b. Tree in flowering stage. c. Tree in fruiting stage. d. Gum secretion from the cut marks on the stem. (For interpretation of
the references to color in the text, the reader is referred to the web version of this article.)

et al., 1980). These gum exudates are water insoluble due to their used as vegetables (Farooq et al., 2012; Ramachandran et al., 1980;
mucilaginous texture and belong to the tragacanth or hog gum series Stevens et al., 2013). Culinary practices are affected by varying tradi-
(Ramachandran et al., 1980). tions and taste preferences (Seshadri and Nambiar, 2003). Traditional
It is a fast growing, medium sized deciduous tree, propagated as a dishes across the world include the use of tender young plants, young
perennial plant from cuttings and seeds (Ramachandran et al., 1980; and mature leaflets and flowers in soup and sauce preparations
Seshadri and Nambiar, 2003). It mainly grows in semi-arid, tropical and (Morton, 1991; Stevens et al., 2013). Leaves are also used in salad
subtropical regions, although the drought resistance properties of the preparation with groundnut (Fahey, 2005; Morton, 1991; Stevens et al.,
plant makes it more suitable for drier regions (Farooq et al., 2012; 2013), herbal tea preparations, porridge, complementary baby foods,
Leone et al., 2015; Seshadri and Nambiar, 2003; Thurber and Fahey, spice and as garnish. The fresh leaves serve as a good snack when
2009). The roots are susceptible to water-logging and tend to rot in such chewed raw (Ramachandran et al., 1980; Stevens et al., 2013).
conditions. It is also able to tolerate a wide range of soil types, the In addition, seeds obtained from tender and mature pods are po-
optimum being a well-drained sandy or loamy soil (pH 5–9) (Patel pularly used in pickling (Ramachandran et al., 1980). Seeds obtained
et al., 2010a,b; Ramachandran et al., 1980). from young pods are eaten green, and mature seeds are either roasted
M. oleifera is valued for its multiple economic, medicinal and neu- or fried before consumption and taste like peanuts (Fahey, 2005;
traceutical properties worldwide. This plant has been honored as Morton, 1991; Ramachandran et al., 1980). The extremely pungent
“Botanical of the Year – 2007” by the National Institute of Health (NIH). roots are mixed with vinegar and salt and serve as a substitute to the
The Africans used to call it “Never Die” or “Miracle Tree” for its ability popular condiment “horseradish” (Morton, 1991).
to treat more than 300 diseases. This review focuses the important medicinal and neutraceutical
All the plant parts including immature pods, leaves, mature seeds, properties and also the biotechnology of this high value plant M. olei-
flowers and roots have long been consumed by humans for a variety of fera.
applications. The immature pods, leaves, flowers and mature seeds are

2
S. Gupta et al. Journal of Herbal Medicine 11 (2018) 1–11

2. Methodology reported to be comparable to that of the most commonly used coagu-


lant, aluminium sulphate and as a consequence use of M. oleifera as a
A thorough and critical survey of the literature related to Moringa substitute to these chemical coagulants is encouraged for turbid water
research was conducted up to March 2017. Various online and offline treatments (Pritchard et al., 2010), along with the use of its flower
resources were taken into consideration. The primary source of data extracts for disinfection of the waste water (Moura et al., 2011). Fur-
collection for the review included research papers and review articles ther, Broin et al. (2002) cloned cDNA encoding one of the flocculating
published by reputed publishers such as Springer, Elsevier, Routledge proteins present in M. oleifera seeds and the resulting recombinant
and Taylor & Francis imprints, BMC and PLOS One. Online databases proteins were used against clay particles, as well as Gram positive and
including NCBI, Scopus and Science Direct were also referred to collect Gram negative bacteria. Besides being used as a natural coagulant,
the data on M. oleifera. The paid articles were accessed through the flocculent and microbicidal, the tree has also been explored for its
Bioinformatics Infrastructure Facility Center sponsored by DBT, New potential to remediate toxic compounds usually found in industrial soil
Delhi located at the University of Rajasthan campus. Some other ‘grey and waste water.
literature’ sources such as webpages, conference proceedings, book The presence of toxins in effluents, due to improper disposal prac-
chapters and theses were also reviewed to access the maximum possible tices, lead to soil and water pollution and disturbance in the food chain
information about the potential utilities, extent of research carried out associated with a particular ecosystem. Hence, strong emphasis has
and current bottlenecks in the M. oleifera research. The Flora of the been made over the past few years for the development of efficient
Indian Desert (Bhandari, 1995), a comprehensive book based on prac- methods for removal of toxicants. Use of M. oleifera as a natural bio-
tical survey of the Desert zone of India, was referred to for collection of sorbent for remediation of toxicants is one novel approach, in which
information about the geographical distribution, taxonomy and mor- researchers have explored different parts of the tree for their remedial
phology of the tree. Herbarium “RUBL”, located at University of Ra- efficiency using different parameters. Biosorbents developed by che-
jasthan Campus was also visited to collect data regarding the mor- mical modification of leaves serve as an excellent substitute to the
phological and identification characteristics of the tree. The importance conventionally used adsorbents for removal of heavy metals, particu-
of this plant in Ayurvedic, Unani, Siddha and folk medicinal formula- larly Cd(II), Cu(III), Ni(II) and Pb(II) from the aqueous solution (Reddy
tions was further validated through the Indian Medicinal Plant Data- et al., 2010a, 2012). In a similar study, Reddy et al. (2010b) demon-
base, NMPB (http://www.medicinalplants.in). strated that M. oleifera bark can serve as an efficient biosorbent for
Literature search was conducted using Google Scholar and PubMed biosorption of Pb2+, and concluded that such biosorbents can serve as
using the following keywords: Moringa oleifera; drumstick; sajina; efficient substitutes to the existing synthetic adsorbents such as acti-
Moringa pterygosperma; metabolite profiling of Moringa; metabolite vated carbon and ion exchange resins. Efficient removal of azo and
profiling of drumstick; phytochemical analysis of Moringa; in vitro anthroquinonic dyes and organic compounds such as benzene, toluene,
propagation; micropropagation; uses of drumstick tree; and importance ethylbenzene and cumene using seed extracts and pods, respectively
of Moringa oleifera. The commercial importance of the tree was re- has also been illustrated (Akhtar et al., 2007; Beltrán-Heredia and
viewed by surveying different market places and online stores for the Sánchez Martín, 2008).
availability of Moringa oleifera products. These studies not only highlight the potential of M. oleifera as an
In this review the challenges associated with commercialization of efficient and environmentally friendly coagulant and biosorbent for
the products of drumstick tree, along with the scope of biotechnological bioremediation of polluted water and land, but open new gateways for
interventions as a remedy, are discussed. The review thus highlights the developing commercially viable coagulants, flocculants and biosorbents
need for future research pertaining to the application of biotechnology for remediation of xenobiotics.
for enhancement of commercial value of the tree.
3.2. Nutritional value
3. Advances in M. oleifera research/multidimensional potential
and utilities of M. oleifera M. oleifera has been used as a dietary supplement due to its rich
nutritional content. It is an outstanding indigenous source of vitamins,
3.1. Bioremediation potential proteins and minerals (Fahey, 2005). The tree contains digestible pro-
teins, iron, magnesium, calcium, vitamins (B6, B2 and C), and car-
Use of M. oleifera as a water purifier has been a common practice for otenoids. M. oleifera is one of the richest natural sources of provitamin
decades, and research in this direction revealed the significant bior- A (Maheshwari et al., 2014; Mehta et al., 2011; Seshadri and Nambiar,
emediation potential of different components of the tree. 2003).
During the early 1990s, the efficiency and properties of the drum- Every part of this tree has been found to possess many nutrients.
stick tree as a natural coagulant were assessed and it was found that M. Flowers, leaves, young shoots and immature pods are good sources of
oleifera seeds, while being biodegradable and non-toxic, can act as a methionine and are also rich in phosphorus, calcium and iron. The
viable substitute to industrial coagulants such as alum (Sutherland refined seed oil is an acceptable substitute to olive oil, due to the pre-
et al., 1994). This coagulating potential is attributed to dimeric cationic sence of all the essential fatty acids as in olive oil (Morton, 1991). The
proteins (13 kDa) and flocculating proteins (about 6.5 kDa) as isolated lipid composition of seeds is even greater than that of soyabean, making
and purified from aqueous extracts of M. oleifera plant parts it nutritionally important. The high content of essential amino acids in
(Gassenschmidt et al., 1995; Ndabigengesere et al., 1995). In fact, the the seeds makes it an excellent substitute to legumes, which are poor in
purified proteins were reported to be more effective as the coagulation sulphur containing amino acids (Ferreira et al., 2008).
process is mediated through adsorption and neutralization of colloidal The leaves of the plant are also significant in terms of their use as a
charges (Ndabigengesere et al., 1995). These promising results led to nutritional supplement to combat malnutrition (Fahey, 2005; Seshadri
extensive research on the isolation and purification of efficient coagu- and Nambiar, 2003; Thurber and Fahey, 2009).
lants from M. oleifera extracts, and as a result a variety of coagulating According to Botany of the Plant Industry as well as Trees of Life (an
and flocculating components were isolated from different plant parts. NGO), “leaves of M. oleifera possess: 4 times more calcium and two
Okuda et al. (2001) successfully purified an active coagulant from the times more protein than milk, 7 times more Vit. C than oranges, 3 times
salt solution of M. oleifera seeds crude extract, while hemaggulatinating more potassium and iron than banana and spinach respectively and 4
proteins and a lectin coagulant were isolated and purified from M. times more Vit. A than carrots” (Thurber and Fahey, 2009) – and
oleifera tissue extracts and seeds, respectively by Santos et al. (2009). therefore, this plant is unique as it is very rare to find such a diversified
The coagulant activity of different extracts and purified proteins was nutrient profile in a single plant species (Razis et al., 2014).

3
S. Gupta et al. Journal of Herbal Medicine 11 (2018) 1–11

This tree serves as a sustainable and economically sound nutrient Moyo et al., 2012b; Ogbunugafor et al., 2012; Siddhuraju and Becker,
rich food supplement, for those suffering from malnutrition, especially 2003; Vongsak et al., 2013). Similarly, the antioxidant activities of
in developing countries. In addition, it is also being used to boost food aqueous extracts of roots (Satish et al., 2013) and methanolic extracts of
security, foster rural development and support sustainable land. stems and pods (Gupta et al., 2012; Kumbhare et al., 2012) have also
been ascertained using both in vitro as well as in vivo assays.
3.3. Pharmaceutical value Sreelatha and Padma (2010), reported that the diverse antioxidant
profile of leaves also corresponds to the cryoprotective nature of the
The medicinal virtues of M. oleifera are well known through its use plant. The plant parts can also be used as natural preservative for fat
as a traditional medicine for a number of diseases and disorders since (Patel et al., 2010a,b).
ancient times (Ramachandran et al., 1980). This tree has been popu-
larly used as a folk medicine for anaemia, arthritis and rheumatism, 3.3.2. Antitumor activity
asthma, constipation, diarrhea, stomach pain, ulcers, intestinal spasms, Extracts of the stem and seeds are reported to exhibit cytotoxic,
headache and sore gums (Mehta et al., 2011; Pandey et al., 2012; Razis anticancer and antitumor activities (Araújo et al., 2013; Onsare and
et al., 2014). The tree possesses abortifacient, anti-rheumatic, anti-in- Arora, 2015; Shaban et al., 2012). Similarly, leaf extracts prepared in
flammatory, bactericidal and diuretic activities and also serves as an- different solvents have also been found to exhibit anticancer, cytotoxic
tidote, emetic, purgative, stimulant, tonic, vermifuge (anthelmintic (Berkovich et al., 2013; Nair and Varalakshmi, 2011), antiproliferative
medicine) and vesicant (Fatima et al., 2014; Mehta et al., 2011; Patel (Pamok et al., 2012), antimyelomic (Parvathy and Umamaheshwari,
et al., 2010a,b). The gum exudates are not only reported to be used in 2007), antileukemia, antihepatocarcinoma (Khalafalla et al., 2010) and
treating various chronic disorders, but are also being used as a potential chemoprotective (Anwar et al., 2007; Murakami et al., 1998) activities.
substitute to synthetic binders and suspending agents in drug delivery Phytochemical analysis revealed that these activities are credited to
systems (Jarald et al., 2012; Panda and Ansari, 2013; Panda et al., 3-O-6x-oleoyl-β-D-glucopyranosyl-β-sitosterol, β-sitosterol-3-O-β-D-glu-
2008; Varma et al., 2014). Various performulation studies ascertained copyranoside, 4-(α-L-rhamnosyloxy) phenylacetonitrile, 4-hydro-
the suitability of M. oleifera gum as a mucoadhesive polymer, disin- xyphenylacetonitrile and 4-hydroxyphenyl-acetamide in seeds
tegrant, binder (Patel et al., 2012) and as a sustained release polymer (Guevara et al., 1999; Villasenor et al., 1989); cis-9-hexadecenal, quinic
(Varma et al., 2014), while the drug dissolution and disintegration acid, 3,5-dihydroxy-6-methyl-2,3-dihydro-4H-pyran-4-1, 9-octadece-
studies showed that the rate of drug release can adequately be mon- namide, methyl octadecenoate in flowers (Inbathamizh and Padmini,
itored by varying the gum concentration in the tablet formulation 2012) and quercetin, kaempferol (Krishnamurthy et al., 2015; Sreelatha
(Basawaraj et al., 2010; Panda et al., 2008). Later, in vitro toxicity as- et al., 2011), 4-(4′-O-acetyl-α-L-rhamnosyloxy) benzyl isothiocyanate,
says of Moringa gum confirmed its non-toxic nature, thus rendering it O-ethyl-4-(α-L-rhamnosyloxy) benzyl carbamate, 4-(L-rhamnosyloxy)
safe for both topical and non-topical formulations (Jarald et al., 2012; benzyl isothiocyanate, niaziminin and niazimicin (Anwar et al., 2007;
Panda et al., 2006). Shah et al. (2011), evaluated the efficiency of Murakami et al., 1998) in leaves.
Moringa gum as a pharmaceutical excipient based on various physico- Recently, it was found that the antitumor activities of leaf extracts is
chemical parameters and concluded that the gum obtained from M. mediated through the antioxidant and apoptosis inducing activities of
oleifera serves as an excellent natural source of pharmaceutical ex- the plant (Jung, 2014; Tiloke et al., 2013).
cipient. Therefore, Moringa gum exudates can efficiently serve as a
natural substitute to the toxic and synthetic excipients, leading to en- 3.3.3. Antimicrobial activity
hanced therapeutic efficiency of the drug by improvising its stability, Antibacterial activities of different components of the plant in-
rate of dissolution and disintegration. Similarly, Moringa coagulant can cluding leaves (Abalaka et al., 2012; Doughari et al., 2007; Kekuda
be used as an efficient amorphous state stabilizer in many drug for- et al., 2010; Mandal et al., 2014; Moyo et al., 2012a; Oluduro, 2012;
mulations such as ibuprofen, meloxicam and felodipine (Bhende and Rahman et al., 2009; Thilza et al., 2010; Valarmathy et al., 2010), seeds
Jadhav, 2012). The salt extract of Moringa seeds has also been parti- (Auwal et al., 2013; Jabeen et al., 2008; Oluduro et al., 2010; Viera
cularly used with Povidone (polyvinylpyrrolidone, PVP) to improve et al., 2010) and pods (Arora and Onsare, 2014) have been reported
solubility of the partially soluble NSAIDS (Noolkar et al., 2013). against E. coli, S. typhi, P. aeruginosa, E. cloace, P. vulgaris, S. aureus, M.
Recent studies indicate that the tree gains its exceptional ability to kristinae, E. aerogenes, Shigella, B. cereus, Streptococcus-B-haemolytica, B.
treat such a wide range of medical conditions from its rich phyto- subtilis, K. pneumonia, B. megaterium, S. lutea, B. sterothermophilus, S.
chemical composition. An overview of some of the important medicinal pyogenes, V. cholera, S. entridis, enteropathogens and wound bacteria.
properties and associated phytochemicals is as follows: Extracts of seeds (Chuang et al., 2007; Jabeen et al., 2008; Oluduro
et al., 2010) and leaves (Chuang et al., 2007; Kekuda et al., 2010;
3.3.1. Antioxidant activity Oluduro, 2012) are also reported to exhibit significant anti-fungal ac-
The antioxidant activity of different parts of M. oleifera is mainly tivity against T. mentagrophyte, Pullarium sp., A. flavus, Penicillium sp., A.
attributed to the presence of ascorbic acid (Vit. C), β-carotene (Kumar niger, A. oryzae, A. terreus, A. nidulans, F. solani, R. solani, C. clados-
et al., 2012; Mahajan and Mehta, 2007), quercetin, kaempferol (Gupta porioides, P. sclerotigenum and Dermatophytes (T. rubrum, E. xoccosum,
et al., 2012), and phenolic acids (Anwar et al., 2007; Mahajan and M. canis).
Mehta, 2007; Sreelatha et al., 2011). Also, isothiocyanates, polyphenols Significant antimicrobial activity of root extracts against E. coli, S.
and rutin from leaves (Bajpai et al., 2005; Tumer et al., 2015); toco- aureus, P. aeruginosa, P. mirabilis, Penicillium sp., Mucor sp., A. niger and
pherols, myricetin and lectins from seeds (Mahajan and Mehta, 2007; C. albicans have also been reported.
Santos et al., 2005; Singh et al., 2013); procyanidins from bark In addition, the antiviral activities of seeds against HSV-1 (Ali et al.,
(Atawodi et al., 2010) and palmitic acid, phytosterols and 9-octadece- 2004) and that of leaves against Epstin Barr Virus (EBV), HIV and HSV-
namide from the flowers (Inbathamizh and Padmini, 2012) have been 1 (Lipipun et al., 2003; Nworu et al., 2013; Sudha et al., 2010) have also
regarded as potent antioxidants. Monopalmitic acid, oleic acid, tri-oleic been tested successfully.
triglycerides present in seed oil are also reported to exhibit antioxidant Further investigations showed that these antimicrobial activities are
activity (Lalas and Tsaknis, 2002; Mahajan and Mehta, 2007). possibly attributed to the presence of 4-(α-L-rhamnopyranosyloxy)
Antioxidant activities of different extracts obtained from different benzyl isothiocyanate, methyl N-4-(α-L-rhamnopyranosyloxy)benzyl
plant parts have been reported in several studies. Extracts of leaves in carbamate, 4-(α-D-glucopyranosyl-1→4-α-L-rhamnopyranosyloxy)-
organic solvents such as methanol and acetone have been found to benzyl thiocarboxamide (Oluduro et al., 2010; Pandey et al., 2012) and
exhibit antioxidant activities (Atawodi et al., 2010; Charoensin, 2014; 4-(α-L-rhamnopyranosyloxy)benzyl glucosinolate (Goyal et al., 2007),

4
S. Gupta et al. Journal of Herbal Medicine 11 (2018) 1–11

together with proanthocyanidins and glucomoringine (Maldini et al., 3.3.7. Neuroprotective effect
2014; Singh et al., 2013). The presence of cardiac glycosides in pods Hannan et al. (2014), revealed that leaves exert a neuroprotective
(Arora and Onsare, 2014), kaempferol, rhamnetin, kaempferitin, iso- effect by promoting neuronal survival and neurite outgrowth. Leaf ex-
quercitrin and pterygospermin in flowers, spirochin and anthonine in tracts are also reported to exhibit a protective effect against Alzheimer’s
roots (Farooq et al., 2012; Mehta et al., 2011; Raj et al., 2011) and disease by altering brain monoamine levels and electrical activity
aglycon of deoxy-niazimicine (N-benzyl,S-ethyl thioformate) in bark (Ganguly and Guha, 2008). Further, it was found that the CNS de-
(Nikkon et al., 2003) also contribute to their antimicrobial nature. pressant action of leaves is mediated through its analgesic and antic-
onvulsive activities (Bakre et al., 2013; Gupta et al., 1999; Ray et al.,
2003).
3.3.4. Anti-inflammatory activity
The presence of potent anti-inflammatory agents including 4-[(α-L- 3.3.8. Treatment of metabolic disorders
rhamnosyloxy)benzyl] isothiocyanate, 4-[(4′-O-acetyl-α-L-rhamnosy- Leaf extracts have been reported to possess hypocholesterolemic
loxy)benzyl]isothiocyanate (Morton, 1991; Stohs and Hartman, 2015; (Ghasi et al., 2000), antihyperglycemic, antihyperlipidemic and hepa-
Waterman et al., 2014), quercetin, kaempferol glucosides, (Coppin toprotective activities (Divi et al., 2012; Jain et al., 2010; Jaiswal et al.,
et al., 2013), 4-(2-O-acetyl- α-L-rhamnosyloxy)benzyl isothiocyanate, 4- 2009; Kumar and Pari, 2003; Ndong et al., 2007; Pari and Kumar,
(3-O-acetyl-α-rhamnosyloxy)benzyl isothiocyanate (Cheenpracha et al., 2002).
2010), 3,5-dihydroxy-6-methyl-2,3-dihydro-4H-pyran-4-1, 9-octadece- Later, Tahiliani and Kar (2000) and Chumark et al. (2008) affirmed
namide (Inbathamizh and Padmini, 2012), aurantiamide acetate and the presence of anti-hyperthyroidism and anti-atherosclerotic activities
1,3-dibenzyl urea (Maheshwari et al., 2014; Pandey et al., 2012; in leaves. Also, extracts of roots have been found to possess estrogenic,
Sashidhara et al., 2009) in different parts of M. oleifera have been re- anti-estrogenic, progestational and anti-progestational activities (Bose,
ported. The reported anti-inflammatory and anti-arthritic activities of 2007; Shukla et al., 1988) along with liver and kidney protective effects
seed extracts are ascribed to the presence of glycosides (Gupta et al., (Mazumder et al., 1999).
2005; Hamza, 2010; Mahajan et al., 2007). Phytochemical analysis showed that β-sitosterol and 4-[α-(L-rham-
Further, Sulaiman et al. (2008), demonstrated that anti-in- nosyloxy) benzyl]-o-methyl thiocarbamate (trans) are potent choles-
flammatory activity of leaves is mediated through inhibition of in- terol lowering, anti-diarrheal, antispasmodic, hypolipidemic agents
flammation signaling pathways. The role of leaf and root extracts in (Anwar et al., 2007; Jain et al., 2010; Maheshwari et al., 2014), while
treatment of inflammation (Ezeamuzie et al., 1996; Pal et al., 1995) and quercetin-3-glycoside is responsible for the antidiabetic (Farooq et al.,
that of pod extracts in amelioration of inflammation associated dis- 2012), antidyslipidemic (Mbikay, 2012) and hypoglycemic
orders such as cancer, asthma, allergic rhinitis, atopic dermatitis and (Maheshwari et al., 2014) effects. Also, moringine and moringinine are
rheumatoid arthritis (Lee et al., 2013; Muangnoi et al., 2012) has also reported to have a role in antihypoglycemic properties of the plant (Kar
been studied. et al., 1999; Maheshwari et al., 2014; Mehta et al., 2011). The presence
of insulin like plant proteins in leaf extracts has been recently reported
3.3.5. Cardio-protective activity for treatment of diabetes (Paula et al., 2016).
The presence of O-[2′-hydroxy-3′-(2″-heptenyloxy)]propyl un- Stohs and Hartman (2015), reported the presence of a novel com-
decanoate, O-ethyl-4-(α-L-(rhamnosyloxy)-benzyl carbamate, methyl p- pound, niaziridin, which enhances the gastrointestinal absorption of
hydroxybenzoate, (Faizi et al., 1998; Pandey et al., 2012; Stohs and vitamins and nutrients. The presence of quercetin and kaempferol in
Hartman, 2015) and 4-(α -L-rhamnosyloxy benzyl)-O-methyl thio- flowers and pods has been reported to be associated with their anti-
carbamate (Farooq et al., 2012; Pandey et al., 2012) correspond to the diabetic and hepatoprotective activities (Farooq et al., 2012; Gupta
hypotensive nature of the plant as a whole, while N- α-L-rhamnopyr- et al., 2012).
anosyl vincosamide from the bark is reported to exhibit cardioprotec-
tive effects (Morton, 1991). 3.3.9. Other activities
The hypotensive and bradycardiac activities of leaves are attributed M. oleifera has also been reported to have wound healing activities.
to niazinin, niazimicin, niaziminin, niazimin, niazirin, niazicin, niazir- Potent wound healing agents such as vicenin-2 (Muhammad et al.,
inin, 4-(4′-O-acetyl-α-rhamnosyloxy) benzyl isothiocyanate (Bose, 2013), quercetin, kaempferol and phytosterols (Hukkeri et al., 2006)
2007; Faizi et al., 1994a,b), and glucomoringine is reported to have a have been reported in ethyl acetate extract of the leaves. Recently,
hypertensive effect (Dangi et al., 2002; Maheshwari et al., 2014). Lambole and Kumar (2012) reported dexamethasone present in bark as
Leaves are also reported to exert a positive effect on the circulatory an efficient wound healer. It mediates the activity by suppressing anti-
system and reduce mortality and morbidity as a consequence of cor- healing agents. The wound healing activity has also been reported in
onary heart diseases, due to the presence of gossypetin, quercetagenin the seeds of M. oleifera (Bhatnagar et al., 2013; Parwani et al., 2016).
and proanthocyanidins (Kumar et al., 2012; Seshadri and Nambiar, Leaves, seeds and roots of the plant also possess various other activities,
2003). Nandave et al. (2009), demonstrated that the cardioprotective such as schizonticidal, antiulcerogenic (Dahiru et al., 2006; Patel et al.,
activity of leaves is mediated through its antioxidant, antiperoxidative 2010a,b), larvicidal (De Oliveira et al., 2011; Prabhu et al., 2011),
and myocardial preservative effects. Later, Abdulazeez et al. (2016) antiurolithiatic and anti-ulcerative colitis (Gholap et al., 2012; Karadi
reported that the blood pressure effect of alkaloids and flavonoids et al., 2006), whereas the gum exudates of plant are reported to exhibit
present in seeds and leaves is due to their inhibitory effect on angio- macrofilaricidal activity (Kushwaha et al., 2011).
tensin converting enzymes. Extracts of the leaves contribute significant radioprotective effects
as estimated by Rao et al. (2001) using comparative scoring of aber-
rations in bone marrow metaphase chromosomes between mice pre-
3.3.6. Immunomodulatory activity treated with leaf extracts and those without any pre-treatment prior to
Leaf extracts have been found to exhibit both immunosuppressive irradiation. Seeds are reported to exhibit antipyretic (Hukkeri et al.,
and immunostimulatory activities (Rachmawati and Rifa’i, 2014). The 2006), aphrodisiac (Zade et al., 2013), antihypersentitivity and anti-
immunomodulatory effect of leaves is mediated through reduction in anaphylactic (Mahajan and Mehta, 2007) activities along with being a
cyclophosphamide induced immunosuppression by stimulating both potent antitoxidant to fluoride toxicity (Ranjan et al., 2009).
cellular and humoral immunity (Gupta et al., 2010), which is attributed The anti-asthamatic and anti-rhinitis activities of seed extracts were
to the presence of compounds like isothiocyanates and glycoside cya- attributed to the presence of moringine and moringinine, which play a
nides (Sudha et al., 2010). significant role in bronchiole relaxation and treatment of respiratory

5
S. Gupta et al. Journal of Herbal Medicine 11 (2018) 1–11

tract infections (Agrawal and Mehta, 2008; Mahajan and Mehta, 2008). 5. Need for M. oleifera biotechnology
Mehta et al. (2003), reported the presence of anti-atherosclerotic ac-
tivity in pods, while the protective effect of the phenolic compounds of In the past few years, Moringa has gained the attention of both
the tree against oxidative stress was recently reported by Ramabulana scientific and business communities due to its economic importance in
et al. (2016). terms of food, medicines, waste management and sustainable agri-
Evidently, drumstick tree serves as a good source of neutraceuticals culture practices. The extensive research and attempt for commercia-
which are very rare to find altogether in a single plant in sufficient lization of drumstick tree and its products has led to an urgent need for
quantities. Thus, the tree can be regarded as medicinally important for its conservation – from dietary, pharmacological, ethnobotanical and
prevention as well as cure. biotechnological perspectives.
One solution to the increasing demand for M. oleifera based products
is biotechnological approaches, which include in vitro propagation for
3.4. Other economic utilities
mass multiplication, enhanced biosynthesis of secondary metabolites
through metabolite profiling, pathway engineering and recombinant
Apart from the above discussed properties, the tree has also been
technology.
studied for some other economic utilities such as feed value and a
source of natural gas. M. oleifera has emerged as an important cattle
5.1. Problems associated with M. oleifera propagation
feed that can be used as a substitute to conventional animal feeds such
as sunflower seed cake and alfalfa (Babiker et al., 2016; Sarwatt et al.,
The Drumstick tree is easily propagated by stem cuttings as well as
2002) due to its high nutritional value. Use of Moringa silage as a feed
seeds, but propagation by cutting reduces the growth of the mother
has been reported to exhibit a significantly positive effect on milk
plant and also affects the yield and sometimes can cause death of the
quality and quantity in dairy cows (Mendieta-Araica et al., 2011), and
mother plant (Islam et al., 2005). Plants obtained by seed propagation
milk yield and growth performance in goats and lambs (Babiker et al.,
vary in genotypes leading to variations in phenotypes and nutritional
2016). Further, Aregheore (2002) suggested that use of the tree foliage
values (Riyathong et al., 2010). The tree also has low seed viability and
as a part of dairy forage for goats serves as an economical source of
low seed germination rates, which in turn results in slow population
protein supplementation. The cost of beef production and nutritional
growth rates (Stephenson and Fahey, 2004).
quality of roughages was estimated to be optimum when M. oleifera was
The need for large areas of agricultrural land for mass cultivation of
used as a feed, thereby serving as an economical forage for cattle (Roy
M. oleifera, to meet the increasing demand of industries for production
et al., 2016).
of finished goods is somewhat impractical in a society where scarcity of
The tree has also been reported to be an efficient feedstock for
cultivable land is a major concern due to increasing population and
biofuels such as biogas and biodiesel (Kafuku and Mbarawa, 2010;
industrialization.
Rashid et al., 2008). The vegetable oil obtained from the seeds of tree
M. oleifera is highly susceptible to infestations with flies, aphids,
has been reported to be of superior quality than that of Jatropha curcas
thrips and other insects, leading to foliar damage which in turn results
(Kibazohi and Sangwan, 2011). These findings provide new horizons
in decline in total biomass and thereby making it unfit for consumption
that could be investigated through sustainable development and green
as food and medicine. Additionally, the frost sensitive nature of the tree
technology.
makes it almost impossible to grow it in open lands of temperate zones
In addition to culinary uses, the oil obtained from the seeds has also
(Förster et al., 2013).
been used for a variety of purposes such as lighting, hair dressing, lu-
Being the sole cultivable species of family Moringaceae, the tree has
brication in watches, soap making and perfume production (Morton,
to meet the growing demands of local population for its use as food,
1991). The seed cake remaining after oil extraction is commonly used
medicine and water clarifiers which may lead to gradual decline in its
as a biofertilizer due to its rich nutrient content.
biodiversity due to reducing natural tree resources (Steinitz et al., 2009;
Pontual et al. (2012) reported the caseinolytic and milk coagulating
Stephenson and Fahey, 2004).
activities of proteases present in flowers, while in a recent similar study,
Apart from these issues, one of the major drawbacks that usually
milk clotting enzymes were purified and characterized from seeds and
holds back the commercialization of such medicinal plant species is
were successfully employed in cheese making, thus suggesting an al-
fluctuations in metabolic constituents due to inconsistent environ-
ternative to conventional animal rennets (Ahmed, 2016).
mental conditions, harvesting conditions and other variations which in
Gum exudates obtained from excisions on the bark have been used
turn causes difficulties in standardization of commercial formulations.
in leather tanning and calico printing (Morton, 1991).
Moreover, the geographical distribution of the tree also results in var-
Lee et al. (2016), recently developed an antimicrobial, antioxidative
iations in morphological, metabolic and genetic characteristics as a
and biodegradable packaging material from puffer fish skin gelatin
consequence of different climatic conditions prevailing in different
containing M. oleifera leaf extract.
zones (Bhatia et al., 2013; Chatterjee et al., 2010).
Therefore, the tree is not only important from medicinal and nu-
tritional perspectives, but also has a vast range of other potential uti-
5.2. Biotechnological strategies: potential solution
lities.
Biotechnology is the only field of science that not only aids in
4. Commercial value of the plant gaining insights into the actual mechanisms of living bodies but also
provides numerous techniques which can be employed for their further
The considerable medicinal potential has led to the development of improvisation. Plant biotechnology is one such specialized field of
various commercial formulations, such as Shrigu, Rumalaya, Septilin biotechnology in which biotechnological tools and techniques enable
(The Himalaya Drug Company, Bangaluru, India), Livospin (Herbals one to explore plant systems and to understand mechanisms associated
APS Pvt. Ltd., Patna, India) and Orthoherb (Walter Bushnell Ltd. with different activities followed by their improvisation such as en-
Mumbai, India), while Ratnagiri Rasa, Sarasvata Ghrta, and Sudarsana hanced growth and yield, rapid mass multiplication, increased meta-
churna are some of its ayurvedic formulations. bolite production and secretion. Plant tissue culture, metabolite pro-
The gum exudates of the tree are valued in the Indian as well as filing, genetic and pathway engineering are some of the common tools
global markets for its dark colored texture and rich medicinal value. of plant biotechnology which are being used extensively to achieve
The increasing global demand and limitations in the availability of the different objectives of crop improvement in terms of yield, disease re-
gum has also resulted in its increasing costs. sistance and enhanced nutritional value.

6
S. Gupta et al. Journal of Herbal Medicine 11 (2018) 1–11

Plant tissue culture via in vitro propagation helps in the conservation plant growth regulator (PGR) for shoot induction and multiplication.
of endangered, medicinally and economically important plant species. They also recorded 100% rooting in MS medium without any PGRs, but
It also provides scope for improvisation by generation of /virus free maximum number of roots (2.2–3.7 and 2.8) was reported on MS
plants, maintaining supply of seasonal crops throughout the year and medium containing Indole-3-acetic acid (IAA) at 0.05, 1 & 2 mg/L and
rapid bulk propagation. Plants possess a variety of phytochemicals Indole-3-butyric acid (IBA) at 2 mg/L, respectively (Islam et al., 2005).
which exhibit numerous medicinal and non-medicinal properties, but Studies based on establishment of an in vitro propagation method
lack of knowledge about the phytocomposition and pathways involved using seeds are also reported. In a study carried out by Marfori (2010),
makes it difficult to exploit their properties at commercial scale. nodes from seedlings in a sterile environment were subjected to MS
Metabolite profiling aids in identification of key enzymes involved in medium supplemented with varying concentration regimes of different
biosynthesis of commercially important phytochemicals. Genetic en- growth hormones and the highest number of axillary shoots per ex-
gineering leads to generation of new varieties possessing the desired plants (4.6) was obtained in medium containing 2.5 μM BAP, 5 μM Ki-
characteristics and are commonly referred to as genetically modified netin (Kn) and 2.5 μM Thidiazuron (TDZ). Other cytokinins, i.e. Kn
(GM) plants. (10 μM) and TDZ (5–10 μM) showed inhibitory effect. Saini et al. (2012)
In addition, the analysis of morphological, biochemical and genetic also reported that nodal segments obtained from aseptically grown
variability using suitable molecular markers not only helps in the se- seedlings, when inoculated on MS medium containing 4.44 μM BAP
lection of superior varieties of interest for future breeding programs and showed best response (9 ± 1.0) and highest number of roots (8 ± 0.8)
mass multiplication for commercial purposes, but also provides insights were observed on IAA (2.85 μM) and IBA (4.29 μM).
into the effects of different biotic and abiotic factors on the morpho- Devendra et al. (2012) carried out tissue culture study using zygotic
logical, phytochemical and genetic characteristics (Goyal et al., 2015). embryos as explants. Zygotic embryos grown on MS medium containing
Such analysis facilitates the selection and identification of superior 10.75 μM NAA resulted in white organogenic callus, which was further
varieties of plants with particular desired characteristics, which can be subjected to induction of somatic embryos using MS media supple-
further subjected to mass multiplication and breeding programs. mented with 4.52 μM 2,4-Dichlorophenoxyacetic acid (2,4 D) and
11.09 μM BAP. They concluded that MS medium with 13.31 μM BAP
5.2.1. Micropropagation showed highest somatic embryo induction frequencies with germina-
In recent years, in vitro micropropagation based techniques have tion efficiency of 77.2%. Further, maximum rooting efficiency (78.4%)
proved a fruitful approach for mass multiplication and germplasm was reported in MS medium supplemented with 10.75 μM NAA.
conservation of endangered as well as economically and medicinally Similar studies of callus cultures were also carried out using leaves
important plants (Goyal et al., 2015; Jain et al., 2009; Steinitz et al., and cotyledons as explants.
2009; Stephenson and Fahey, 2004). Highest callus growth was obtained using leaf explants in MS
Plant tissue culture technologies serve as an excellent alternative to medium supplemented with 0.1 mg/L 2,4-D (Khalafalla et al., 2011).
conventional propagation methods, resulting in year round availability Nieves and Aspuria (2011) established callus culture for cotyledons and
of the tree which involves rapid clonal mass multiplication in a stipu- observed increased callus formation on MS medium containing 2.5 mg/
lated time and space. This approach enables meeting the huge in- L BAP. Also, enrichment of media with TDZ results in compact, nodular
dustrial demands for drumstick tree as raw materials in variety of drug and loose calli (Nieves and Aspuria, 2011).
and dietary formulations. Riyathong et al. (2010) used stem explants obtained from in vitro
According to Jain et al., (2011), in vitro regeneration and multi- raised seedlings and 100% shoot formation was obtained on MS
plication techniques are not only being used as a one-stop solution to all medium supplemented with BAP (1–4 mg/L), and the highest number
the major concerns such as overexploitation, enhancement of nutri- of shoots per explant (10.2) was achieved with BAP (2 mg/L).
tional value and disease resistance but can also be used for commercial In a recent report by Avila-Treviño et al. (2017), successful micro-
production of metabolites. propagation protocol was established using buds and cotyledonary
Therefore, current biotechnology research is focused on optimiza- nodes of the tree without using any plant growth regulators and they
tion of in vitro propagation methods using different parts of M. oleifera further illustrated the genetic similarity among the regenerated plant-
in order to meet the commercial demand and overcome the formerly lets using RAMP markers. The tissue culture studies are summarized in
discussed issues. Table 1.
In vitro propagation of M. oleifera was reported by Stephenson and Thus, it can be concluded that the in vitro propagation studies car-
Fahey (2004), in which they used meristematic stem nodes and im- ried out so far provide a platform for further/future interventions in
mature seeds as explants and reported that the explants showed best order to obtain superior varieties with enhanced medicinal and nutri-
response in full and half strength Murashige and Skoog (MS) medium. tional value. In one such attempt, Mathur and Kamal (2012) studied the
Multiplication of shoots was dramatically increased when the nodal biosynthesis of “trigonelline – a metabolically active pyridine alkaloid”
segments were cultured on membrane raft. The use of isothiazolones for in different components of M. oleifera including the callus cultures and
improved survival rate of the rooted plantlets was also reported in the reported that the trigonelline production in in vitro grown cultures can
study. be significantly enhanced using nicotinic acid as precursor in the
In a similar report by Förster et al. (2013), nodes taken from the medium.
sterile seedlings of two different ecotypes were cultured on MS medium Additionally, use of plant cell suspension cultures and hairy root
supplemented with different concentrations of 6-benzylaminopurine cultures for large scale production of commercially important sec-
(BAP). Results revealed that MS medium without BAP and with 0.5 mg/ ondary metabolites have been suggested by Rao and Ravishankar
L BAP showed best response in terms of shoot length (3.81–4.10 cm) (2002), whereas Förster et al. (2013) concluded that in vitro cultures are
and number of offshoots (4.34–4.40), respectively. In experiments successfully being used in medicine and food industries as a source for
conducted using nodal segments as an explant, Islam et al. (2005) re- important metabolites.
ported that at BAP (1 mg/L and 1.5 mg/L) all the explants produced The importance of comprehensive metabolite profiling and en-
shoots and maximum number of shoots (4 ± 0.29) were obtained with gineering was highlighted by Zhou and Wu (2006). They illustrated the
1 mg/L BAP. On the other hand, Mathur and Kamal (2012) reported the role of plant tissue culture techniques along with plant genomics,
best response on MS medium supplemented with BAP:1-Naphthalene transcriptomics, proteomics and metabolomics for production of im-
acetic acid (NAA) at 3:3 mg/L. Further, Islam et al. (2005) found that portant plant pharmaceuticals and neutraceuticals on a commercial
repeated subcultures on same BAP concentrations resulted in increased scale.
number of shoots (25) and concluded that BAP is the most effective Therefore, it is inferred that comprehensive knowledge of the

7
S. Gupta et al. Journal of Herbal Medicine 11 (2018) 1–11

Table 1
In vitro propagation studies in M. oleifera.

Explant Used PGRs Used Method of Propagation Reference(s)

Meristematic Nodes, Immature Seeds BAP, Ancymidol, NAA, Kinetin Direct Shoot Induction Stephenson and Fahey (2004)
Nodes BAP, IBA, IAA Direct Shoot Induction Islam et al. (2005)
Seeds BAP, Kinetin, TDZ, NAA Direct Shoot Induction Marfori (2010)
Leaves 2,4-D Callus Khalafalla et al. (2011)
Cotyledons BAP, 2,4-D, TDZ Callus Nieves and Aspuria (2011)
Leaves, Zygotic embryos 2,4-D, NAA, Kinetin, BAP Somatic embryogenesis Devendra et al. (2012)
Nodes IBA, BAP Direct Shoot Induction Mathur and Kamal (2012)
Nodes Triacontanol (TRIA), BAP, IAA, IBA Direct Shoot Induction Saini et al. (2012)
Nodes BAP, IAA Direct Shoot Induction Förster et al. (2013)
Nodes, Leaf No PGR(s) Indirect regeneration/callus induction Avila-Treviño et al. (2017)

metabolites present and their biosynthesis pathway is the key behind References
converting plant cells and tissues into large bioreactors which are often
referred to as “plant cell factories” for commercial production of phy- Abalaka, M.E., Daniyan, S.Y., Oyeleke, S.B., Adeyemo, S.O., 2012. The antibacterial
tomolecules/phytochemicals. evaluation of Moringa oleifera leaf extracts on selected bacterial pathogens. J.
Microbiol. Res. 2, 1–4.
Abdulazeez, A.M., Ajiboye, O.S., Wudil, A.M., Abubakar, H., 2016. Partial purification
and characterization of angiotensin converting enzyme inhibitory alkaloids and fla-
6. Conclusion and future prospects vonoids from the leaves and seeds of Moringa oleifera. J. Adv. Biol. Biotechnol. 5,
1–11.
Agrawal, B., Mehta, A., 2008. Antiasthmatic activity of Moringa oleifera Lam: a clinical
In recent years, great emphasis has been laid on the utilization of study. Ind. J. Pharmacol. 40, 28–31.
natural resources as a source of food, medicine and waste management Ahmed, A.E.T.A.M., 2016. Purification and Characterization of Milk-Clotting Enzymes
from Moringa oleifera Lam. Seeds. University of Khartoum.
due to the harmful effects posed by the synthetic resources, in addition Akhtar, M., Hasany, S.M., Bhanger, M.I., Iqbal, S., 2007. Sorption potential of Moringa
to their non-renewable nature. Moringa oleifera Lam. is a tree with oleifera pods for the removal of organic pollutants from aqueous solutions. J. Hazard.
multidimensional utilities which serves as an excellent reservoir for Mater. 141, 546–556.
Ali, G.H., El-Taweel, G.E., Ali, M.A., 2004. The cytotoxicity and antimicrobial efficiency
pharmaceuticals and neutraceuticals. The rich phytochemical profile
of Moringa oleifera seeds extracts. Int. J. Environ. Stud. 61, 699–708.
and advances in biotechnological techniques has led to generation of Anwar, F., Latif, S., Ashraf, M., Gilani, A.H., 2007. Moringa oleifera: a food plant with
new avenues aimed towards enhancement of overall commercial value multiple medicinal uses. Phytother. Res. 21, 17–25.
of the tree. Araújo, L.C.C., Aguiar, J.S., Napoleão, T.H., Mota, F.V.B., Barros, A.L.S., Moura, M.C.,
Coriolano, M.C., Coelho, L.C.B.B., Silva, T.G., Paiva, P.M.G., 2013. Evaluation of
In vitro propagation techniques have emerged as a life-line for ex situ cytotoxic and anti-inflammatory activities of extracts and lectins from Moringa olei-
conservation of endangered and elite germplasm and also lay the fera seeds. PLoS One 8, e81973.
foundation for the concept of “plant cell factories” i.e. the natural Aregheore, E.M., 2002. Intake and digestibility of Moringa oleifera–batiki grass mixtures
by growing goats. Small Rumin. Res. 46, 23–28.
bioreactors for production of plant products. This concept provides new Arora, D.S., Onsare, J.G., 2014. In vitro antimicrobial evaluation and phytoconstituents of
insights into development of more effective, eco-friendly and biode- Moringa oleifera pod husks. Ind. Crops Prod. 52, 125–135.
gradable products using mass multiplication and production techni- Arora, D.S., Onsare, J.G., Kaur, H., 2013. Bioprospecting of Moringa (Moringaceae): mi-
crobiological perspective. J. Pharmacogn. Phytochem. 1, 193–215.
ques. Establishment of plant cell factories demands thorough knowl- Atawodi, S.E., Atawodi, J.C., Idakwo, G.A., Pfundstein, B., Haubner, R., Wurtele, G.,
edge of the metabolomics of the plant as it facilitates in identification Bartsch, H., Owen, R.W., 2010. Evaluation of the polyphenol content and antioxidant
and alteration/modulation of the key components for enhanced pro- properties of methanol extracts of the leaves, stem, and root barks of Moringa oleifera
Lam. J. Med. Food 13, 710–716.
duction of targeted plant product(s). Auwal, M.S., Tijjani, A.N., Sadiq, M.A., Saka, S., Mairiga, I.A., Shuaibu, A., Adawaren, E.,
Although efficient in vitro propagation protocols for M. oleifera have Gulani, I.A., 2013. Antibacterial and haematological activity of Moringa oleifera
been established, comprehensive studies involving the use of metabolite aqueous seed extract in Wistar albino rats. Sokoto J. Vet. Sci. 11, 28–37.
Avila-Treviño, J.A., Muñoz-Alemán, J.M., Pérez-Molphe-Balch, E., Rodríguez-Sahagún,
profiling and engineering for enhanced metabolite production in in vitro
A., Morales-Domínguez, J.F., 2017. In vitro propagation from bud and apex explants
cultures are still lacking. Knowledge of biosynthetic pathways of the of Moringa oleifera and evaluation of the genetic stability with RAMP marker. S. Afr.
desired compounds in plants and in culture systems will facilitate the J. Bot. 108, 149–156.
modulation and fabrication of such compounds in in vitro cultures. Babiker, E.E., Juhaimi, F.A.L., Ghafoor, K., Abdoun, K.A., 2016. Comparative study on
feeding value of Moringa leaves as a partial replacement for alfalfa hay in ewes and
Therefore, a comprehensive metabolite profiling followed by biosyn- goats. Livest. Sci. 195, 21–26.
thetic pathway identification using high throughput techniques such as Bajpai, M., Pande, A., Tewari, S.K., Prakash, D., 2005. Phenolic contents and antioxidant
GC–MS and NMR is essential for identification and characterization of activity of some food and medicinal plants. Int. J. Food Sci. Nutr. 56, 287–291.
Bakre, A.G., Aderibigbe, A.O., Ademowo, O.G., 2013. Studies on neuropharmacological
novel neutraceuticals and pharmaceuticals. profile of ethanol extract of Moringa oleifera leaves in mice. J. Ethnopharmacol. 149,
The use of biotechnological approaches thereby not only facilitates 783–789.
selection and conservation of superior varieties but is also a stepping Basawaraj, S.P., Srinivas, R.S., Upendra, K., Prakassh, G.K., 2010. Evaluation of Moringa
oleifera gum as a binder in tablet formulation. Int. J. Res. Ayurveda Pharm. 1,
stone to commercialization of important/targeted plant products. 590–596.
There is no doubt that the shortcomings in M. oleifera commercia- Beltrán-Heredia, J., Sánchez Martín, J., 2008. Azo dye removal by Moringa oleifera seed
lization can be overcome by the use of biotechnological tools, and it is extract coagulation. Color. Technol. 124, 310–317.
Berkovich, L., Earon, G., Ron, I., Rimmon, A., Vexler, A., Lev-Ari, S., 2013. Moringa
likely that the future research in this context will make M. oleifera an
oleifera aqueous leaf extract down-regulates nuclear factor-kappaB and increases
important solution for the various health and environmental issues of cytotoxic effect of chemotherapy in pancreatic cancer cells. BMC. Complement.
the present era/generation. Altern. Med. 13, 212–218.
Bhandari, M.M., 1995. Flora of the Indian Desert. MPS Repros, Jodhpur, India.
Bhatia, A., Bharti, S.K., Tewari, S.K., Sidhu, O.P., Roy, R., 2013. Metabolic profiling for
studying chemotype variations in Withania somnifera (L.) Dunal fruits using GC–MS
Conflicts of interest and NMR spectroscopy. Phytochemistry 93, 105–115.
Bhatnagar, M., Parwani, L., Sharma, V., Ganguli, J., Bhatnagar, A., 2013. Hemostatic:
antibacterial biopolymers from Acacia arabica (Lam.) Willd. and Moringa oleifera
The authors have no conflict of interest. (Lam.) as potential wound dressing materials. Indian J. Exp. Biol. 51, 804–810.
Bhende, S., Jadhav, N., 2012. Moringa coagulant as a stabilizer for amorphous solids: part
I. AAPS PharmSciTech 13, 400–410.

8
S. Gupta et al. Journal of Herbal Medicine 11 (2018) 1–11

Bose, C.K., 2007. Possible role of Moringa oleifera Lam. root in epithelial ovarian cancer. oleifera on arsenic-induced toxicity in rats. Environ. Toxicol. Pharmacol. 20,
Med. Gen. Med. 9, 26–34. 456–464.
Broin, M., Santaella, C., Cuine, S., Kokou, K., Peltier, G., Joet, T., 2002. Flocculent activity Gupta, A., Gautam, M.K., Singh, R.K., Kumar, M.V., Rao, C.V., Goel, R.K., Anupurba, S.,
of a recombinant protein from Moringa oleifera Lam. seeds. Appl. Microbiol. 2010. Immunomodulatory effect of Moringa oleifera Lam. extract on cyclopho-
Biotechnol. 60, 114–119. sphamide induced toxicity in mice. Indian J. Exp. Biol. 48, 1157–1160.
Charoensin, S., 2014. Antioxidant and anticancer activities of Moringa oleifera leaves. J. Gupta, R., Mathur, M., Bajaj, V.K., Katariya, P., Yadav, S., Kamal, R., Gupta, R.S., 2012.
Med. Plants Res. 8, 318–325. Evaluation of antidiabetic and antioxidant activity of Moringa oleifera in experimental
Chatterjee, S., Srivastava, S., Khalid, A., Singh, N., Sangwan, R.S., Sidhu, O.P., Roy, R., diabetes. J. Diabates 4, 164–171.
Khetrapal, C.L., Tuli, R., 2010. Comprehensive metabolic fingerprinting of Withania Hamza, A.A., 2010. Ameliorative effects of Moringa oleifera Lam. seed extract on liver
somnifera leaf and root extracts. Phytochemistry 71, 1085–1094. fibrosis in rats. Food Chem. Toxicol. 48, 345–355.
Cheenpracha, S., Park, E.J., Yoshida, W.Y., Barit, C., Wall, M., Pezzuto, J.M., Chang, L.C., Hannan, M.A., Kang, J.Y., Mohibbullah, M., Hong, Y.K., Lee, H., Choi, J.S., Choi, I.S.,
2010. Potential anti-inflammatory phenolic glycosides from the medicinal plant Moon, I.S., 2014. Moringa oleifera with promising neuronal survival and neurite
Moringa oleifera fruits. Biorg. Med. Chem. 18, 6598–6602. outgrowth promoting potentials. J. Ethnopharmacol. 152, 142–150.
Chuang, P.H., Lee, C.W., Chou, J.Y., Murugan, M., Shieh, B.J., Chen, H.M., 2007. Anti- Hukkeri, V.I., Nagathan, C.V., Karadi, R.V., Patil, B.S., 2006. Antipyretic and wound
fungal activity of crude extracts and essential oil of Moringa oleifera Lam. Bioresour. healing activities of Moringa oleifera Lam. in rats. Ind. J. Pharm. Sci. 68, 124-126.
Technol. 98, 232–236. Inbathamizh, L., Padmini, E., 2012. Gas Chromatography-Mass Spectrometric analyses of
Chumark, P., Khunawat, P., Sanvarinda, Y., Phornchirasilp, S., Morales, N.P., Phivthong- methanol extract of Moringa oleifera flowers. Int. J. Chem. Anal. Sci. 2, 1394–1397.
ngam, L., Ratanachamnong, P., Srisawat, S., Klai-upsorn, S.P., 2008. The in vitro and Islam, S., Jahan, M.A.A., Khatun, R., 2005. In vitro regeneration and multiplication of
ex vivo antioxidant properties: hypolipidaemic and antiatherosclerotic activities of year-round fruit bearing Moringa oleifera L. J. Biol. Sci. 5, 145–148.
water extract of Moringa oleifera Lam. leaves. J. Ethnopharmacol. 116, 439–446. Jabeen, R., Shahid, M., Jamil, A., Ashraf, M., 2008. Microscopic evaluation of the anti-
Coppin, J.P., Xu, Y., Chen, H., Pan, M.H., Ho, C.T., Juliani, R., Simon, J.E., Wu, Q., 2013. microbial activity of seed extracts of Moringa oleifera. Pak. J. Bot. 40, 1349–1358.
Determination of flavonoids by LC/MS and anti-inflammatory activity in Moringa Jain, R., Sinha, A., Kachhwaha, S., Kothari, S.L., 2009. Micropropagation of Withania
oleifera. J. Funct. Foods 5, 1892–1899. coagulans (Stocks) Dunal: a critically endangered medicinal herb. J. Plant Biochem.
Dahiru, D., Onubiyi, J.A., Umaru, H.A., 2006. Phytochemical screening and anti- Biotechnol. 18, 249–252.
ulcerogenic effect of Moringa oleifera aqueous leaf extract. Afr. J. Tradit. Complement. Jain, P.G., Patil, S.D., Haswani, N.G., Girase, M.V., Surana, S.J., 2010. Hypolipidemic
Altern. Med. 3, 70–75. activity of Moringa oleifera Lam. Moringaceae, on high fat diet induced hyperlipi-
Dangi, S.Y., Jolly, C.I., Narayanan, S., 2002. Antihypertensive activity of the total alka- demia in albino rats. Rev. Bras. Framacogn. 20, 969–973.
loids from the leaves of Moringa oleifera. Pharm. Biol. 40, 144–148. Jain, R., Sinha, A., Jain, D., Kachhwaha, S., Kothari, S.L., 2011. Adventitious shoot re-
De Oliveira, C.F.R., Luz, L.A., Paiva, P.M.G., Coelho, L.C.B.B., Marangoni, S., Macedo, generation and in vitro biosynthesis of steroidal lactones in Withania coagulans
M.L.R., 2011. Evaluation of seed coagulant Moringa oleifera lectin (cMoL) as a (Stocks) Dunal. Plant Cell Tissue Organ Cult. 105, 135–140.
bioinsecticidal tool with potential for the control of insects. Process Biochem. 46, Jaiswal, D., Rai, P.K., Kumar, A., Mehta, S., Watal, G., 2009. Effect of Moringa oleifera
498–504. Lam. leaves aqueous extract therapy on hyperglycemic rats. J. Ethnopharmacol. 123,
Devendra, B.N., Talluri, V.S.S.L.P., Srinivas, N., 2012. Callus induction and somatic em- 392–396.
bryogenesis of Moringa oleifera Lam. an anti-radiation plant. J. Agr. Technol. 8, Jarald, E.E., Sharma, S., Sheeja, E., Ahmad, S., Patni, S., Daud, A., 2012. Characterization
1953–1963. of Moringa oleifera Lam. gum to establish it as a pharmaceutical excipient. Ind. J.
Divi, S.M., Bellamkonda, R., Dasireddy, S.K., 2012. Evaluation of antidiabetic and anti- Pharm. Educ. Res. 46, 211–216.
hyperlipedemic potential of aqueous extract of Moringa oleifera in fructose fed insulin Jung, I.L., 2014. Soluble extract from Moringa oleifera leaves with a new anticancer ac-
resistant and STZ induced diabetic wistar rats: a comparative study. Asian J. Pharm. tivity. PLoS One 9, e95492.
Clin. Res. 5, 67–72. Kafuku, G., Mbarawa, M., 2010. Alkaline catalyzed biodiesel production from Moringa
Doughari, J.H., Pukuma, M.S., De, N., 2007. Antibacterial effects of Balanites aegyptiaca L. oleifera oil with optimized production parameters. Appl. Energy 87, 2561–2565.
Drel. and Moringa oleifera Lam. on Salmonella typhi. Afr. J. Biotechnol. 6, 2212–2215. Kar, A., Choudhary, B.K., Bandyopadhyay, N.G., 1999. Preliminary studies on the in-
Ezeamuzie, I.C., Ambakederemo, A.W., Shode, F.O., Ekwebelem, S.C., 1996. organic constituents of some indigenous hypoglycaemic herbs on oral glucose tol-
Antiinflammatory effects of Moringa oleifera root extract. Int. J. Pharm. 34, 207–212. erance test. J. Ethnopharmacol. 64, 179–184.
Förster, N., Mewis, I., Ulrichs, C., 2013. Moringa oleifera—establishment and multi- Karadi, R.V., Gadge, N.B., Alagawadi, K.R., Savadi, R.V., 2006. Effect of Moringa oleifera
plication of different ecotypes in vitro. Gesunde Pflanz. 65, 21–31. Lam. root-wood on ethylene glycol induced urolithiasis in rats. J. Ethnopharmacol.
Fahey, J.W., 2005. Moringa oleifera: a review of the medical evidence for its nutritional, 105, 306–311.
therapeutic, and prophylactic properties. Part 1. Trees Life J. 1, 1–15. Kekuda, T.R.P., Mallikarjun, N., Swathi, D., Nayana, K.V., Aiyar, M.B., Rohini, T.R., 2010.
Faizi, S., Siddiqui, S.B., Saleem, R., Siddiqui, S., Aftab, K., Gilani, A.H., 1994a. Novel Antibacterial and antifungal efficacy of steam distillate of Moringa oleifera Lam. J.
hypotensive agents, niazimin A, niazimin B, niazicin A and niazicin B from Moringa Pharm. Sci. Res. 2, 34–37.
oleifera: isolation of first naturally occurring carbamates. J. Chem. Soc. Perkin Trans. Khalafalla, M.M., Abdellatef, E., Dafalla, H.M., Nassrallah, A.A., Aboul-Enein, K.M.,
1, 3035–3040. Lightfoot, D.A., El-Deeb, F.E., El-Shemy, H.A., 2010. Active principle from Moringa
Faizi, S., Siddiqui, B.S., Saleem, R., Siddiqui, S., Aftab, K., Gilani, A.H., 1994b. Isolation oleifera Lam leaves effective against two leukemias and a hepatocarcinoma. Afr. J.
and structure elucidation of new nitrile and mustard oil glycosides from Moringa Biotechnol. 9, 8467–8471.
oleifera and their effect on blood pressure. J. Nat. Prod. 57, 1256–1261. Khalafalla, M.M., Nassrallah, H.M.D.A., Aboul-Enein, K.M., El-Shemy, H.A., Abdellatef,
Faizi, S., Siddiqui, B.S., Saleem, R., Aftab, K., Shaheen, F., Gilani, A.H., 1998. Hypotensive E., 2011. Dedifferentiation of leaf explants and antileukemia activity of an ethanolic
constituents from the pods of Moringa oleifera. Planta Med. 64, 225–228. extract of cell cultures of Moringa oleifera. Afr. J. Biotechnol. 10, 2746–2750.
Farooq, F., Rai, M., Tiwari, A., Khan, A.A., Farooq, S., 2012. Medicinal properties of Khanuja, S.P.S., 2012. Functional diversity of plant metabolome and microbiome in
Moringa oleifera: an overview of promising healer. J. Med. Plants Res. 6, 4368–4374. health services to the human life. Proc. Natl. Acad. Sci. India Sect. B: Biol. Sci. 82,
Fatima, T., Sajid, M.S., Jawad-ul-Hassan, M., Siddique, R.M., Iqbal, Z., 2014. 291–294.
Phytomedicinal value of Moringa oleifera with special reference to antiparasitics. Pak. Kibazohi, O., Sangwan, R.S., 2011. Vegetable oil production potential from Jatropha
J. Agr. Sci. 51, 251–262. curcas, Croton megalocarpus, Aleurites moluccana, Moringa oleifera and Pachira glabra:
Ferreira, P.M.P., Farias, D.F., Oliviera, J.T.A., Carvalho, A.F.U., 2008. Moringa oleifera: assessment of renewable energy resources for bio-energy production in Africa.
bioactive compounds and nutritional potential. Rev. Nutr. 21, 431–437. Biomass Bioenergy 35, 1352–1356.
Ganguly, R., Guha, D., 2008. Alteration of brain monoamines & EEG wave pattern in rat Krishnamurthy, P.T., Vardarajalu, A., Wadhwani, A., Patel, V., 2015. Identification and
model of Alzheimer’s disease & protection by Moringa oleifera. Ind. J. Med. Res. 128, characterization of a potent anticancer fraction from the leaf extracts of Moringa
744–751. oleifera L. Indian J. Exp. Biol. 53, 98–103.
Gassenschmidt, U., Jany, K.D., Bernhard, T., Niebergall, H., 1995. Isolation and char- Kumar, A.N., Pari, L., 2003. Antioxidant action of Moringa oleifera Lam. (drumstick)
acterization of a flocculating protein from Moringa oleifera Lam. BBA-Gen. Subj. 1243, against antitubercular drugs induced lipid peroxidation in rats. J. Med. Food 6,
477–481. 255–259.
Ghasi, S., Nwobodo, E., Ofili, J., 2000. Hypocholesterolemic effects of crude extract of Kumar, V., Pandey, N., Mohan, N., Singh, R.P., 2012. Antibacterial & antioxidant activity
leaf of Moringa oleifera Lam. in high-fat diet fed Wistar rats. J. Ethnopharmacol. 69, of different extract of Moringa oleifera leaves–an in-vitro study. Int. J. Pharm. Sci. Rev.
21–25. Res. 12, 89–94.
Gholap, P.A., Nirmal, S.A., Pattan, S.R., Pal, S.C., Mandal, S.C., 2012. Potential of Moringa Kumbhare, M.R., Guleha, V., Sivakumar, T., 2012. Estimation of total phenolic content,
oleifera root and Citrus sinensis fruit rind extracts in the treatment of ulcerative colitis cytotoxicity and in–vitro antioxidant activity of stem bark of Moringa oleifera. Asian
in mice. Pharm. Biol. 50, 1297–1302. Pac. J. Trop. Dis. 2, 144–150.
Goyal, B.R., Agrawal, B.B., Goyal, R.K., Mehta, A.A., 2007. Phyto-pharmacology of Kushwaha, V., Saxena, K., Verma, S.K., Lakshmi, V., Sharma, R.K., Murthy, P.K., 2011.
Moringa oleifera Lam.: an overview. Nat. Prod. Radiance 6, 347–353. Antifilarial activity of gum from Moringa oleifera Lam. on human lymphatic filaria
Goyal, P., Jain, R., Kachhwaha, S., Kothari, S.L., 2015. Assessment of genetic diversity in Brugia malayi. Chron. Young Sci. 2, 201–206.
Pithecellobium dulce (Roxb.) Benth. germplasm using RAPD and ISSR markers. Trees- Lalas, S., Tsaknis, J., 2002. Extraction and identification of natural antioxidant from the
Struct Funct 29, 637–653. seeds of the Moringa oleifera tree variety of Malawi. J. Am. Oil Chem. Soc. 79,
Guevara, A.P., Vargas, C., Sakurai, H., Fujiwara, Y., Hashimoto, K., Maoka, T., Kozuka, 677–683.
M., Ito, Y., Tokuda, H., Nishino, H., 1999. An antitumor promoter from Moringa Lambole, V., Kumar, U., 2012. Effect of Moringa oleifera Lam. on normal and dex-
oleifera lam. Mutat. Res./Genet. Toxicol. Environ. Mutagen. 440, 181–188. amethasone suppressed wound healing. Asian Pac. J. Trop. Biomed. 2, S219–S223.
Gupta, M., Mazumder, U.K., Chakrabarti, S., 1999. CNS activities of methanolic extract of Lee, H.J., Jeong, Y.J., Lee, T.S., Park, Y.Y., Chae, W.G., Chung, I.K., Chang, H.W., Kim,
Moringa oleifera root in mice. Fitoterapia 70, 244–250. C.H., Choi, Y.H., Kim, W.J., 2013. Moringa fruit inhibits LPS-induced NO/iNOS ex-
Gupta, R., Kannan, G.M., Sharma, M., Flora, S.J.S., 2005. Therapeutic effects of Moringa pression through suppressing the NF-κ B activation in RAW264. 7 cells. Am. J. Chin.

9
S. Gupta et al. Journal of Herbal Medicine 11 (2018) 1–11

Med. 41, 1109–1123. dispersions. AAPS PharmSciTech 14, 569–577.


Lee, K.Y., Yang, H.J., Song, K.B., 2016. Application of a puffer fish skin gelatin film Nworu, C.S., Okoye, E.L., Ezeifeka, G.O., Esimone, C.O., 2013. Extracts of Moringa oleifera
containing Moringa oleifera Lam. leaf extract to the packaging of Gouda cheese. J. Lam. showing inhibitory activity against early steps in the infectivity of HIV-1 len-
Food Sci. Technol. 1–8. tiviral particles in a viral vector-based screening. Afr. J. Biotechnol. 12, 4866–4873.
Leone, A., Spada, A., Battezzati, A., Schiraldi, A., Aristil, J., Bertoli, S., 2015. Cultivation, Ogbunugafor, H., Igwo-Ezikpe, M., Igwilo, I., Ozumba, N., Adenekan, S., Ugochukw, C.,
genetic, ethnopharmacology, phytochemistry and pharmacology of Moringa oleifera Onyekwelu, O., Ekechi, A., 2012. In vitro and in vivo evaluation of antioxidant
leaves: an overview. Int. J. Mol. Sci. 16, 12791–12835. properties of Moringa oleifera ethanolic leaves extract and effect on serum lipid in-
Lipipun, V., Kurokawa, M., Suttisri, R., Taweechotipatr, P., Pramyothin, P., Hattori, M., dices in rat. Maced. J. Med. Sci. 5, 397–403.
Shiraki, K., 2003. Efficacy of Thai medicinal plant extracts against herpes simplex Okuda, T., Baes, A.U., Nishijima, W., Okada, M., 2001. Isolation and characterization of
virus type 1 infection in vitro and in vivo. Antiviral Res. 60, 175–180. coagulant extracted from Moringa oleifera seed by salt solution. Water Res. 35,
Mahajan, S.G., Mehta, A.A., 2007. Inhibitory action of ethanolic extract of seeds of 405–410.
Moringa oleifera Lam. on systemic and local anaphylaxis. J. Immunotoxicol. 4, Oluduro, O.A., Aderiye, B.I., Connolly, J.D., Akintayo, E.T., Famurewa, O., 2010.
287–294. Characterization and antimicrobial activity of 4-(β-D-glucopyranosyl-1→ 4-(-L-
Mahajan, S.G., Mehta, A.A., 2008. Effect of Moringa oleifera Lam. seed extract on oval- rhamnopyranosyloxy)-benzyl thiocarboxamide; a novel bioactive compound from
bumin-induced airway inflammation in guinea pigs. Inhal. Toxicol. 20, 897–909. Moringa oleifera seed extract. Folia Microbiol. 55, 422–426.
Mahajan, S.G., Mali, R.G., Mehta, A.A., 2007. Protective effect of ethanolic extract of Oluduro, A.O., 2012. Evaluation of antimicrobial properties and nutritional potentials of
seeds of Moringa oleifera Lam. against inflammation associated with development of Moringa oleifera Lam. leaf in South-Western Nigeria. Malays. J. Microbiol. 8, 59–67.
arthritis in rats. J. Immunotoxicol. 4, 39–47. Onsare, J.G., Arora, D.S., 2015. Antibiofilm potential of flavonoids extracted from
Maheshwari, K., Yadav, R.K., Malhotra, J., Dhawan, N.G., Mohan, L., 2014. Fascinating Moringa oleifera seed coat against Staphylococcus aureus, Pseudomonas aeruginosa and
nutritional, prophylactic, therapeutic and socio-economic reconcile attributable to Candida albicans. J. Appl. Microbiol. 118, 313–325.
drum stick tree (Moringa oleifera Lam.). Glob. J. Med. Res.-B: Pharm. Drug Discov. Pal, S.K., Mukherjee, P.K., Saha, B.P., 1995. Studies on the antiulcer activity of Moringa
Toxicol. Med. 14, 11–22. oleifera leaf extract on gastric ulcer models in rats. Phytother. Res. 9, 463–465.
Maldini, M., Maksoud, S.A., Natella, F., Montoro, P., Petretto, G.L., Foddai, M., De Nicola, Pamok, S., Vinitketkumnuen, S.S.U., Saenphet, K., 2012. Antiproliferative effect of
G.R., Chessa, M., Pintore, G., 2014. Moringa oleifera: study of phenolics and gluco- Moringa oleifera Lam. and Pseuderanthemum palatiferum(Nees) Radlk extracts on the
sinolates by mass spectrometry. J. Mass Spectrom. 49, 900–910. colon cancer cells. J. Med. Plants Res. 6, 139–145.
Mandal, B., Bhattacharjee, I., Kundu, J.K., Chandra, G., 2014. Efficacy of extracts of two Panda, D.S., Ansari, S.A., 2013. Preformulation study on the gum of Moringa oleifera.
medicinal plants of India against some pathogenic bacteria. Indian J. Biol. Sci. 20, Malays. J. Pharm. Sci. 11, 41–47.
49–55. Panda, D., Si, S., Swain, S., Kanungo, S.K., Gupta, R., 2006. Preparation and evaluation of
Marfori, E.C., 2010. Clonal micropropagation of Moringa oleifera L. Philipp. Agric. Sci. 93, gels from gum of Moringa oleifera. Ind. J. Pharm. Sci. 68, 777–780.
454–457. Panda, D.S., Choudhury, N.S.K., Yedukondalu, M., Si, S., Gupta, R., 2008. Evaluation of
Mathur, M., Kamal, R., 2012. Studies on trigonelline from Moringa oleifera and its in vitro gum of Moringa oleifera as a binder and release retardant in tablet formulation. Ind. J.
regulation by feeding precursor in cell cultures. Rev. Bras. Framacogn. 22, 994–1001. Pharm. Sci. 70, 614–618.
Mazumder, U.K., Gupta, M., Chakrabarti, S., Pal, D., 1999. Evaluation of hematological Pandey, A., Pandey, R.D., Tripathi, P., Gupta, P.P., Haider, J., Bhatt, S., Singh, A.V., 2012.
and hepatorenal functions of methanolic extract of Moringa oleifera Lam. root treated Moringa ioleifera Lam. (Sahijan)-a plant with a plethora of diverse therapeutic ben-
mice. Indian J. Exp. Biol. 37, 612–614. efits: an updated retrospection. Med. Aromat. Plants 1, 101–108.
Mbikay, M., 2012. Therapeutic potential of Moringa oleifera leaves in chronic hypergly- Pari, L., Kumar, N.A., 2002. Hepatoprotective activity of Moringa oleifera on anti-
cemia and dyslipidemia: a review. Front. Pharmacol. 3, 1–12. tubercular drug-induced liver damage in rats. J. Med. Food 5, 171–177.
Mehta, K., Balaraman, R., Amin, A.H., Bafna, P.A., Gulati, O.D., 2003. Effect of fruits of Parvathy, M.V.S., Umamaheshwari, A., 2007. Cytotoxic effect of Moringa oleifera leaf
Moringa oleifera on the lipid profile of normal and hypercholesterolaemic rabbits. J. extracts on human multiple myeloma cell lines. Trends Med. Res. 2, 44–50.
Ethnopharmacol. 86, 191–195. Parwani, L., Bhatnagar, M., Bhatnagar, A., Sharma, V., Sharma, V., 2016. Evaluation of
Mehta, J., Shukla, A., Bukhariya, V., Charde, R., 2011. The magic remedy of Moringa Moringa oleifera seed biopolymer-PVA composite hydrogel in wound healing dressing.
oliferia: an overview. Int. J. Biomed. Adv. Res. 2, 215–227. Iran. Polym. J. 1–13.
Mendieta-Araica, B., Spörndly, E., Reyes-Sánchez, N., Spörndly, R., 2011. Feeding Patel, J.P., Gami, B., Patel, K., 2010a. Evaluation of in vitro schizonticidal properties of
Moringa oleifera fresh or ensiled to dairy cows—effects on milk yield and milk flavor. acetone extract of some indian medicinal plants. Adv. Biol. Res. 4, 253–258.
Trop. Anim. Health Prod. 43, 1039–1047. Patel, S., Thakur, A.S., Chandy, A., Manigauha, A., 2010b. Moringa oleifera: a review of
Morton, J.F., 1991. The Horseradish tree, Moringa pterygosperma (Moringaceae)—a boon their medicinal and economical importance to the health and nation. Drug Invent.
to arid lands? Econ. Bot. 45, 318–333. Today 2, 339–342.
Moura, M.C., Pontual, E.V., Gomes, F.S., Napoleão, T.H., Xavier, H.S., Paiva, P.M.G., Patel, M.T., Patel, J.K., Upadhyay, U.M., 2012. Assessment of various pharmaceutical
Coelho, L.C.B.B., 2011. Preparations of Moringa oleifera flowers to treat contaminated excipient properties of natural Moringa oleifera gum [Mucoadhesion, disintegration,
water. In: Daniels, J.A. (Ed.), Advances in Envinronmental Research. Nova Science binder]. Int. J. Pharm. Life Sci. 3, 1833–1847.
Publishers, Inc., pp. 269–275. Paula, P.C., Oliveira, J.T.A., Sousa, D.O.B., Alves, B.G.T., Carvalho, A.F.U., Franco, O.L.,
Moyo, B., Masika, P.J., Muchenje, V., 2012a. Antimicrobial activities of Moringa oleifera Vasconcelos, I.M., 2016. Insulin-like plant proteins as potential innovative drugs to
Lam. leaf extracts. Afr. J. Biotechnol. 11, 2797–2802. treat diabetes—the Moringa oleifera case study. New Biotechnol.. http://dx.doi.org/
Moyo, B., Oyedemi, S., Masika, P.J., Muchenje, V., 2012b. Polyphenolic content and 10.1016/j.nbt.2016.10.005. (in press).
antioxidant properties of Moringa oleifera leaf extracts and enzymatic activity of liver Pontual, E.V., Carvalho, B.E., Bezerra, R.S., Coelho, L.C.B.B., Napoleão, T.H., Paiva,
from goats supplemented with Moringa oleifera leaves/sunflower seed cake. Meat Sci. P.M.G., 2012. Caseinolytic and milk-clotting activities from Moringa oleifera flowers.
91, 441–447. Food Chem. 135, 1848–1854.
Muangnoi, C., Chingsuwanrote, P., Praengamthanachoti, P., Svasti, S., Tuntipopipat, S., Prabhu, K., Murugan, K., Nareshkumar, A., Ramasubramanian, N., Bragadeeswaran, S.,
2012. Moringa oleifera pod inhibits inflammatory mediator production by lipopoly- 2011. Larvicidal and repellent potential of Moringa oleifera against malarial vector,
saccharide-stimulated RAW 264.7 murine macrophage cell lines. Inflammation 35, Anopheles stephensi Liston (Insecta: Diptera: Culicidae). Asian Pac. J. Trop. Biomed. 1,
445–455. 124–129.
Muhammad, A.A., Pauzi, N.A.S., Arulselvan, P., Abas, F., Fakurazi, S., 2013. In vitro Pritchard, M., Craven, T., Mkandawire, T., Edmondson, A.S., O’neill, J.G., 2010. A
wound healing potential and identification of bioactive compounds from Moringa comparison between Moringa oleifera and chemical coagulants in the purification of
oleifera Lam. BioMed Res. Int. 2013. drinking water–an alternative sustainable solution for developing countries. Phys.
Murakami, A., Kitazono, Y., Jiwajinda, S., Koshimizu, K., Ohigashi, H., 1998. Niaziminin, Chem. Earth 35, 798–805.
a thiocarbamate from the leaves of Moringa oleifera, holds a strict structural re- Rachmawati, I., Rifa’i, M., 2014. In vitro immunomodulatory activity of aqueous extract
quirement for inhibition of tumor-promoter-induced Epstein-Barr virus activation. of Moringa oleifera Lam. leaf to the CD4+, CD8+ and B220+ cells in Mus musculus. J.
Planta Med. 64, 319–323. Exp. Life Sci. 4, 15–20.
Nair, S., Varalakshmi, K.N., 2011. Anticancer, cytotoxic potential of Moringa oleifera ex- Rahman, M.M., Sheikh, M.M.I., Sharmin, S.A., Islam, M.S., Rahman, M.A., Rahman, M.M.,
tracts on HeLa cell line. J. Nat. Pharm. 2, 138–142. Alam, M.F., 2009. Antibacterial activity of leaf juice and extracts of Moringa oleifera
Nandave, M., Ojha, S.K., Joshi, S., Kumari, S., Arya, D.S., 2009. Moringa oleifera leaf Lam. against some human pathogenic bacteria. CMU J. Nat. Sci. 8, 219–227.
extract prevents isoproterenol-induced myocardial damage in rats: evidence for an Raj, A.J., Gopalakrishnan, V.K., Yadav, S.A., Dorairaj, S., 2011. Antimicrobial activity of
antioxidant, antiperoxidative, and cardioprotective intervention. J. Med. Food 12, Moringa oleifera (Lam.) root extract. J. Pharm. Res. 4, 1426–1430.
47–55. Ramabulana, T., Mavunda, R.D., Steenkamp, P.A., Piater, L.A., Dubery, I.A., Madala, N.E.,
Ndabigengesere, A., Narasiah, K.S., Talbot, B.G., 1995. Active agents and mechanism of 2016. Perturbation of pharmacologically relevant polyphenolic compounds in
coagulation of turbid waters using Moringa oleifera. Water Res. 29, 703–710. Moringa oleifera against photo-oxidative damages imposed by gamma radiation. J.
Ndong, M., Uehara, M., Katsumata, S., Sato, S., Suzuki, K., 2007. Preventive effects of Photochem. Photobiol. B: Biol. 156, 79–86.
Moringa oleifera (Lam.) on hyperlipidemia and hepatocyte ultrastructural changes in Ramachandran, C., Peter, K.V., Gopalakrishnan, P.K., 1980. Drumstick (Moringa oleifera):
iron deficient rats. Biosci. Biotechnol. Biochem. 71, 1826–1833. a multipurpose Indian vegetable. Econ. Bot. 34, 276–283.
Nieves, M.C., Aspuria, E.T., 2011. Callus induction in cotyledons of Moringa oleifera Lam. Ranjan, R., Swarup, D., Patra, R.C., Chandra, V., 2009. Tamarindus indica L. and Moringa
Philipp. Agric. Sci. 94, 239–247. oleifera M. extract administration ameliorates fluoride toxicity in rabbits. Indian J.
Nikkon, F., Saud, Z.A., Rahman, M.H., Haque, M.E., 2003. In vitro antimicrobial activity Exp. Biol. 47, 900–905.
of the compound isolated from chloroform extract of Moringa oleifera Lam. Pak. J. Rao, S.R., Ravishankar, G.A., 2002. Plant cell cultures: chemical factories of secondary
Biol. Sci. 6, 1888–1890. metabolites. Biotechnol. Adv. 20, 101–153.
Noolkar, S.B., Jadhav, N.R., Bhende, S.A., Killedar, S.G., 2013. Solid-state characteriza- Rao, A.V., Devi, P.U., Kamath, R., 2001. In vivo radioprotective effect of Moringa oleifera
tion and dissolution properties of Meloxicam–Moringa Coagulant–PVP ternary solid leaves. Indian J. Exp. Biol. 39, 858–863.

10
S. Gupta et al. Journal of Herbal Medicine 11 (2018) 1–11

Rashid, U., Anwar, F., Moser, B.R., Knothe, G., 2008. Moringa oleifera oil: a possible source apoptosis by Moringa oleifera leaf extract on human cancer cells. Food Chem. Toxicol.
of biodiesel. Bioresour. Technol. 99, 8175–8179. 49, 1270–1275.
Ray, K., Hazra, R., Guha, D., 2003. Central inhibitory effect of Moringa oleifera root ex- Steinitz, B., Tabib, Y., Gaba, V., Gefen, T., Vaknin, Y., 2009. Vegetative micro-cloning to
tract: possible role of neurotransmitters. Indian J. Exp. Biol. 41, 1279–1284. sustain biodiversity of threatened Moringa species. In Vitro Cell. Dev. Biol.-Plant 45,
Razis, A.F.A., Ibrahim, M.D., Kntayya, S.B., 2014. Health benefits of Moringa oleifera. 65–71.
Asian Pac. J. Can. Prev. 15, 8571–8576. Stephenson, K.K., Fahey, J.W., 2004. Development of tissue culture methods for the
Reddy, D.H.K., Harinath, Y., Seshaiah, K., Reddy, A.V.R., 2010a. Biosorption of Pb (II) rescue and propagation of endangered Moringa spp. germplasm. Econ. Bot. 58,
from aqueous solutions using chemically modified Moringa oleifera tree leaves. Chem. S116–S124.
Eng. J. 162, 626–634. Stevens, G.C., Baiyeri, K.P., Akinnnagbe, O., 2013. Ethno-medicinal and culinary uses of
Reddy, D.H.K., Seshaiah, K., Reddy, A.V.R., Rao, M.M., Wang, M.C., 2010b. Biosorption Moringa oleifera Lam. in Nigeria. J. Med. Plants Res. 7, 799–804.
of Pb2+ from aqueous solutions by Moringa oleifera bark: equilibrium and kinetic Stohs, S.J., Hartman, M.J., 2015. Review of the safety and efficacy of Moringa oleifera.
studies. J. Hazard. Mater. 174, 831–838. Phytother. Res. 29, 796–804.
Reddy, D.H.K., Seshaiah, K., Reddy, A.V.R., Lee, S.M., 2012. Optimization of Cd (II), Cu Subrahmanyam, S.V., Shah, J.J., 1988. The metabolic status of traumatic gum ducts in
(II) and Ni (II) biosorption by chemically modified Moringa oleifera leaves powder. Moringa oleifera Lam. IAWA Bull. 9, 187–195.
Carbohydr. Polym. 88, 1077–1086. Sudha, P., Asdaq, S.M., Dhamingi, S.S., Chandrakala, G.K., 2010. Immunomodulatory
Riyathong, T., Dheeranupattana, S., Palee, J., Shank, L., 2010. Shoot multiplication and activity of methanolic leaf extract of Moringa oleifera in animals. Ind. J. Physiol.
plant regeneration from in vitro cultures of Drumstick tree (Moringa oleifera Lam.). Pharmacol. 54, 133–140.
The 8th International Symposium on Biocontrol and Biotechnology 154–159. Sulaiman, M.R., Zakaria, Z.A., Bujarimin, A.S., Somchit, M.N., Israf, D.A., Moin, S., 2008.
Roy, B.K., Bashar, M.K., Hossain, S.M.J., Huque, K.S., Makkar, H.P.S., 2016. Performance Evaluation of Moringa oleifera aqueous extract for antinociceptive and anti-in-
evaluation of Moringa oleifera and available Roughages (Maize and Australian Sweet flammatory activities in animal models. Pharm. Biol. 46, 838–845.
Jumbo) on feeding values of growing BLRI cattle breed-1 (BCB-1) bulls. Am. J. Exp. Sutherland, J.P., Folkard, G.K., Mtawali, M.A., Grant, W.D., 1994. Moringa oleifera as a
Agr. 14, 1–9. natural coagulant. In: 20th WEDC Conference, Affordable Water Supply and
Saini, R.K., Shetty, N.P., Giridhar, P., Ravishankar, G.A., 2012. Rapid in vitro regeneration Sanitation. Colombo, Sri Lanka. pp. 297–299.
method for Moringa oleifera and performance evaluation of field grown nutritionally Tahiliani, P., Kar, A., 2000. Role of Moringa oleifera leaf extract in the regulation of
enriched tissue cultured plants. 3 Biotech 2, 187–192. thyroid hormone status in adult male and female rats. Pharmacol. Res. 41, 319–323.
Santos, A.F.S., Argolo, A.C.C., Coelho, L.C.B.B., Paiva, P.M.G., 2005. Detection of water Thilza, I.B., Sanni, S., Isah, Z., Sanni, F., Talle, M., Joseph, M., 2010. In vitro antimicrobial
soluble lectin and antioxidant component from Moringa oleifera seeds. Water Res. 39, activity of water extract of Moringa oleifera leaf stalk on bacteria normally implicated
975–980. in eye diseases. Acad. Arena 2, 80–82.
Santos, A.F.S., Luz, L.A., Argolo, A.C.C., Teixeira, J.A., Paiva, P.M.G., Coelho, L.C.B.B., Thurber, M.D., Fahey, J.W., 2009. Adoption of Moringa oleifera to combat under-nutrition
2009. Isolation of a seed coagulant Moringa oleifera lectin. Process Biochem. 44, viewed through the lens of the Diffusion of Innovations theory. Ecol. Food Nutr. 48,
504–508. 212–225.
Sarwatt, S.V., Kapange, S.S., Kakengi, A.M.V., 2002. Substituting sunflower seed-cake Tiloke, C., Phulukdaree, A., Chuturgoon, A.A., 2013. The antiproliferative effect of
with Moringa oleifera leaves as a supplemental goat feed in Tanzania. Agroforest. Syst. Moringa oleifera crude aqueous leaf extract on cancerous human alveolar epithelial
56, 241–247. cells. BMC. Complement. Altern. Med. 13, 226–233.
Sashidhara, K.V., Rosaiah, J.N., Tyagi, E., Shukla, R., Raghubir, R., Rajendran, S.M., Tumer, T.B., Rojas-Silva, P., Poulev, A., Raskin, I., Waterman, C., 2015. Direct and in-
2009. Rare dipeptide and urea derivatives from roots of Moringa oleifera as potential direct antioxidant activity of polyphenol-and isothiocyanate-enriched fractions from
anti-inflammatory and antinociceptive agents. Eur. J. Med. Chem. 44, 432–436. Moringa oleifera. J. Agr. Food Chem. 63, 1505–1513.
Satish, A., Kumar, R.P., Rakshith, D., Satish, S., Ahmed, F., 2013. Antimutagenic and Valarmathy, K., Gokulakrishnan, M., Kausar, M.S., Paul, K., 2010. A study of anti-
antioxidant activity of Ficus benghalensisstem bark and Moringa oleiferaroot extract. microbial activity of ethanolic extracts of various plant leaves against selected mi-
Int. J. Chem. Anal. Sci. 4, 45–48. crobial species. Int. J. Pharm. Sci. Res. 1, 293–295.
Seshadri, S., Nambiar, V.S., 2003. Kanjero (Digera arvensis) and Drumstick leaves (Moringa Varma, J.N.R., Kumar, C.P., Reddy, A.K., Raju, P.P., 2014. Evaluation of Moringa oleifera
oleifera): nutrient profile and potential for human consumption. In: Simopoulos, A.P., gum as sustained release polymer in Diclofenac Sodium tablet formulation. Int. J.
Gopalan, C. (Eds.), Plants in Human Health and Nutrition Policy. Karger Medical and Res. Pharm. Chem. 4, 687–693.
Scientific Publishers, Basel, pp. 41–59. Viera, G.H.F., Mourão, J.A., Ângelo, Â.M., Costa, R.A., Vieira, R.H.S.F., 2010.
Shaban, A., Mishra, G.M., Nautiyal, R., Srivastava, S., Tripathi, K., Chaudhary, P., Verma, Antibacterial effect (in vitro) of Moringa oleifera and Annona muricata against Gram
S.K., 2012. In vitro cytotoxicity of Moringa oleifera against different human cancer cell positive and Gram negative bacteria. Rev. Inst. Med. Trop. S. P. 52, 129–132.
lines. Asian J. Pharm. Clin. Res. 5, 271–272. Villasenor, I.M., Lim-Sylianco, C.Y., Dayrit, F., 1989. Mutagens from roasted seeds of
Shah, D.P., Jain, V.C., Dalvadi, H.P., Ramani, V.D., Patel, K.G., Saralai, M.G., Jani, G.K., Moringa oleifera. Mutat. Res. 224, 209–212.
2011. A preliminary investigation of Moringa oleifera Lam. gum as a pharmaceutical Vongsak, B., Sithisarn, P., Mangmool, S., Thongpraditchote, S., Wongkrajang, Y.,
excipient. Int. J. Pharm. Res. Technol. 1, 12–16. Gritsanapan, W., 2013. Maximizing total phenolics, total flavonoids contents and
Shukla, S., Mathur, R., Prakash, A.O., 1988. Antifertility profile of the aqueous extract of antioxidant activity of Moringa oleifera leaf extract by the appropriate extraction
Moringa oleifera roots. J. Ethnopharmacol. 22, 51–62. method. Ind. Crop. Prod. 44, 566–571.
Siddhuraju, P., Becker, K., 2003. Antioxidant properties of various solvent extracts of total Waterman, C., Cheng, D.M., Rojas-Silva, P., Poulev, A., Dreifus, J., Lila, M.A., Raskin, I.,
phenolic constituents from three different agroclimatic origins of drumstick tree 2014. Stable, water extractable isothiocyanates from Moringa oleifera leaves attenuate
(Moringa oleifera Lam.) leaves. J. Agr. Food Chem. 51, 2144–2155. inflammation in vitro. Phytochemistry 103, 114–122.
Singh, R.S.G., Negi, P.S., Radha, C., 2013. Phenolic composition, antioxidant and anti- Zade, V.S., Dabhadkar, D.K., Thakare, V.G., Pare, S.R., 2013. Effect of aqueous extract of
microbial activities of free and bound phenolic extracts of Moringa oleifera seed flour. Moringa oleifera seed on sexual activity of male albino rats. Biol. Forum–Int. J. 5,
J. Funct. Foods 5, 1883–1891. 129–140.
Sreelatha, S., Padma, P.R., 2010. Modulatory effects of Moringa oleifera extracts against Zhou, L.G., Wu, J.Y., 2006. Development and application of medicinal plant tissue cul-
hydrogen peroxide-induced cytotoxicity and oxidative damage. Hum. Exp. Toxicol. tures for production of drugs and herbal medicinals in China. Nat. Prod. Rep. 23,
30, 1359–1368. 789–810.
Sreelatha, S., Jeyachitra, A., Padma, P.R., 2011. Antiproliferation and induction of

11

You might also like