Download as pdf or txt
Download as pdf or txt
You are on page 1of 26

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/364239970

Evaluation of Secondary Metabolites Profiling of Ginger (Zingiber officinale


Roscoe) Rhizome using GC-MS and Its Antibacterial Potential on
Staphylococcus aureus and Escherichia co...

Article  in  Microbiology Research Journal International · October 2022


DOI: 10.9734/MRJI/2022/v32i730397

CITATIONS READS

0 63

2 authors:

Johnson Momoh Oluremi Nurudeen Olaleye


Lagos State University of Science and Technology Lagos State Polytechnic
46 PUBLICATIONS   209 CITATIONS    32 PUBLICATIONS   116 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

It's TETfund grant and T.T Oshin is the lead author. View project

-250G>A and -514 C>T polymorphism in the human hepatic lipase gene promoter influence plasma lipid profiles and lipoprotein ratios in Lagos University Teaching
Hospital patients with ischemic stroke. View project

All content following this page was uploaded by Johnson Momoh on 07 October 2022.

The user has requested enhancement of the downloaded file.


Microbiology Research Journal International

32(7): 7-31, 2022; Article no.MRJI.92461


ISSN: 2456-7043
(Past name: British Microbiology Research Journal, Past ISSN: 2231-0886, NLM ID: 101608140)

Evaluation of Secondary Metabolites Profiling of


Ginger (Zingiber officinale Roscoe) Rhizome
using GC-MS and Its Antibacterial Potential on
Staphylococcus aureus and Escherichia coli
Johnson Oshiobugie Momoh a* and Oluremi Nurudeen Olaleye b

a
Biochemistry Unit, Department of Chemical Sciences, College of Basic Sciences, Lagos State
University of Science and Technology (LASUSTECH), Ikorodu, Lagos State, Nigeria.
b
Microbiology Unit, Department of Biological Sciences, College of Basic Sciences, Lagos State
University of Science and Technology (LASUSTECH), Ikorodu, Lagos State, Nigeria.

Authors’ contributions

This work was carried out in collaboration between both authors. Both authors read and approved the
final manuscript.

Article Information
DOI: 10.9734/MRJI/2022/v32i730397

Open Peer Review History:


This journal follows the Advanced Open Peer Review policy. Identity of the Reviewers, Editor(s) and additional Reviewers, peer
review comments, different versions of the manuscript, comments of the editors, etc are available here:
https://www.sdiarticle5.com/review-history/92461

Received 24 July 2022


Original Research Article Accepted 30 September 2022
Published 07 October 2022

ABSTRACT

Objective: Ginger (Zingiber officinale Roscoe) rhizome is a well-known food spice and flavoring
ingredient with wide range of medicinal properties. The rhizome of ginger consists of unique
secondary metabolites compounds. The study evaluates the secondary metabolites profiling of
ginger (Zingiber officinale Roscoe) rhizome using Gas Chromatography-Mass Spectrometry (GC-
MS) and its antibacterial potential on Staphylococcus aureus and Escherichia coli.
Methodology: The GC-MS and phytochemical screening of the aqueous ginger (Zingiber officinale
Roscoe) rhizome extract were determined using standard procedures. Antibacterial activities were
determined by agar well diffusion methods. The minimum inhibitory concentrations (MIC) were
determined using standard procedure.
Results: The result of the GC-MS analysis shows that thirty six compounds were identified in the
ginger (Zingiber officinale Roscoe) rhizome using GC-MS analysis with tridecane with molecular
formula of C13H28 being the most abundant with peak area of 16.94% and retention time of 12.849.
The phytochemical screening shows that the plant contains saponins, alkaloids, glycoside, simple
_____________________________________________________________________________________________________

*Corresponding author: E-mail: [email protected], [email protected];


Momoh and Olaleye; MRJI, 32(7): 7-31, 2022; Article no.MRJI.92461

phenolics, tannins, flavonoids carbohydrates and reducing sugar. The study shows that at 250
mg/ml, the aqueous ginger extract exhibited little or no response with zone of inhibition of 9.85±0.39
and 8.19±1.33 mg/ml against Staphylococcus aureus and Escherichia coli respectively. The extract
exhibited weak response antibacterial activity against E. coli and moderate response against S.
aureus with zone of inhibition of 13.62±2.03 and 16.73±1.83 at 500 mg/dl respectively. Augmentin
showed moderate and strong response with zone of inhibition of 17.23±1.67 and 21.13±1.34 mg/ml
against E. coli and S. aureus at concentration of 7.50 mg/ml respectively. At 15 mg/ml, augmentin
showed strong response with zone of inhibition of 23.00±2.88 mg/ml against E. coli and potent
response with zone of inhibition of 30.50±2.64 mg/ml against S. aureus. The Minimum inhibitory
concentration (MIC) values for the aqueous ginger are 125 and 250 mg/ml for S. aureus and E. coli
and 7.81 and 15.63 for augmentin solution for the sane organisms respectively.
Conclusions: The aqueous ginger (Zingiber officinale Roscoe) rhizome contains secondary
metabolites and possesses poor antibacterial activity against Staphylococcus aureus and
Escherichia coli and may prevent pathogenic diseases caused by these organisms.

Keywords: Antimicrobial activity; Escherichia coli; GC-MS; ginger; Staphylococcus aureus.

1. INTRODUCTION To date, many techniques, such as liquid


chromatography-mass spectrometry (LC-MS),
gas chromatography-mass spectrometry (GC-
“Ginger (Zingiber officinale Roscoe), rhizome
MS), and nuclear magnetic resonance (NMR)
belongs to the Zingiberaceae family and the plant
have been widely employed for metabolite
has been consumed as a spice and an herbal
profiling” [7]. “Among these techniques, GC-MS
medicine used for the treatment of various
has the advantages of low cost compared to the
diseases for a very long period of time” [1].
other analytical methods, high reproducibility,
“Studies have shown the different bioactive
high resolution, highly repeatable mass spectral
compounds in ginger and the main compounds
fragmentation, and few matrix effects” [8].
are terpene and phenolic compounds. The
phenolic compounds are mainly gingerols,
shogaols, and paradols, which account for the Metabolite profiles have been obtained from
various bioactivities of ginger” [2]. “The pungency various medicinal plants, including: Curucuma
of fresh ginger is mainly due to the gingerols, species [9], Carica papaya leaf [10], Hunteria
whereas the pungency of dried ginger is primarily umbellata seed extract [11], hexane leaf, stem
due to the presence of shogaols, mainly 6- and root extracts of Azadirachta indica A. Juss
shogaol, which are dehydrated forms of [12], methanolic root, stem and leaf extracts of
gingerols” [3]. “6-gingerol and 6-shogaol are the Vernonia amygdalina [13], Carica papaya seed
two active components found in ginger which oil [14], Rehmannia glutinosa using GC-MS
produce a depressor response on blood pressure combined with multivariate statistical analysis
at lower doses in cardiovascular system” [4], [15] and Azadirachta indica root [16].
because of these “health-promoting properties of
the plant, ginger can be considered as an active Medicinal herbs rely on secondary plant
ingredient that can be added to food substances metabolites for their metabolism and actions.
for reducing the risk of cardiovascular disease” Ginger has been found to possess various
[5,6]. biological activities, such as anti-inflammatory
[17], antioxidant [18], anticancer [19] and
“Secondary metabolites are substances antimicrobial [20] activities. Furthermore,
manufactured by plants that exert a wide range numerous studies have shown that ginger
of effects on the plant itself and on other living possesses the potential to prevent and
organisms. They act as antimicrobials, maintain ameliorate the effect of several diseases, such
perennial growth, and induce flowering, fruit set as obesity [21], diabetes mellitus [22],
and abscission. Over 50,000 secondary neurodegenerative diseases [23], respiratory
metabolites have been discovered in the plant disorders [24] and cardiovascular diseases [25].
kingdom. Metabolite profiling requires an
analytical system that can generate useful “Escherichia coli is a gram negative, rod-shaped
datasets and identify the compounds of interest. bacterium that is common inhabitant of the

8
Momoh and Olaleye; MRJI, 32(7): 7-31, 2022; Article no.MRJI.92461

animal and human gut, and may also be found in spectrum of the unidentified compound was
vegetation, soil and water. It is the leading compared with the spectrum of the identified
pathogen causing urinary tract infections” [26-29] compounds stored in the National Institute
and “is among the most common pathogens Standard and Technique library. The names,
causing blood stream infections” [30], otitis molecular weight, structure of the compounds in
media, diarrhea, meningitis, wounds and other the test material were determined.
complications in humans [26,31,32]. “Escherichia
coli is also the most common cause of food and 2.3 Preparation of Aqueous Garlic Extract
water-borne human diarrhea worldwide and in
developing countries, causing many deaths in Aqueous Zingiber officinale extract was prepared
children under the age of five years” [33]. according to the method described by Momoh et
al. [36]. The ginger was cleaned with water to
remove any adhering soil on their surfaces. 100g
“Staphylococcus aureus is a gram positive
of garlic was taken after the removal of the outer
bacterium and they cause wide range of
skin surfaces and cut into small pieces by sterile
infections in human and animals” [34]. “They are
scalpel. The small pieces were blended with 200
found on human skin and mucous membranes.
ml sterile distilled water using sterile blender for 5
However, it can also be found in other areas of
minutes. The homogenized mixture was filtered
human contact including water, soil and food
using white cloth, centrifuged at 2000 × g for 10
products” [34]. They causes serious infections
minutes and the clear supernatant was used for
like bacteremia, septic arthritis, pneumonia,
the experiment. The filtered extract was used for
wound sepsis, septicemia, osteomyelitis,
the study within 4 hours of preparation.
endocarditis, food poisoning, bone and joint
infections and toxic shock syndrome [34]. The
2.4 Preliminary Phytochemical Analysis
study evaluates the secondary metabolites
profiling of ginger (Zingiber officinale Roscoe) The presence of glycosides, tannins, saponin,
rhizome using GC-MS and its antibacterial reducing sugars, alkaloids, flavonoids were
potential on Staphylococcus aureus and determined by qualitative procedures [37,38].
Escherichia coli.
2.5 Test Organisms
2. MATERIALS AND METHODS
To study the antibacterial activity of aqueous
2.1 Collection of Ginger Plant Zingiber officinale extract against two bacterial
strains (Staphylococcus aureus a gram-positive
bacterium with ATCC #6538 and Escherichia coli
The ginger (Zingiber officinale Roscoe), rhizome a gram negative bacterium with ATCC # 25922)
was purchased from Ikorodu market and stored were used for the study. The two microorganisms
in a refrigerator in the Department of Chemical were maintained at 4°C on Nutrient Agar slant in
Sciences (Biochemistry unit), Lagos State the Department of Chemical Sciences and fresh
University of Science and Technology. subcultures were made before use.
2.2 Gas Chromatography - Mass 2.5.1 Inoculum preparation
Spectrometry (GC-MS) Analysis of
Ginger A loopful of isolated colonies of the two
organisms (Staphylococcus aureus and
GC-MS analysis of the Zingiber officinale Escherichia coli) were inoculated separately into
rhizome was carried out on an Agilent technology 4 ml of peptone water, incubated at 37°C for 4
7890 GC system equipped with a mass hours. These actively growing bacterial
spectrometric detector (MSD) as described by suspensions were then adjusted with peptone
Momoh et al. [35]. water to obtain turbidity visually comparable to
that of 0.5 McFarland standards using standard
2.2.1 Detection of components procedure [34]. “The 0.5 McFarland standard
was prepared by mixing 0.5ml of 1.75% (w/v)
Analysis of mass spectrum GC-MS was barium chloride dehydrate (BaCl2. 2H2O) with
conducted by the database of the National 99.5 ml of 1% (v/v) H2SO4. This turbidity was
8
Institute Standard and Technique (NIST) which equivalent to approximately 1 x 10 colony
contained more than 62,000 patterns. The forming units per ml (CFU/ml)” [34].

9
Momoh and Olaleye; MRJI, 32(7): 7-31, 2022; Article no.MRJI.92461

2.5.2 Determination of diameter of zone of ml of Mueller-Hinton broth was poured to a set of


inhibition using agar well diffusion different test tubes and autoclaved.
method Subsequently, 1 ml of 100% aqueous ginger
extract (2g/ml) was poured to the first separate
Agar well-diffusion method was employed for the test tube to make a concentration of 50%, and
determination of the antibacterial activity of two-fold serial dilutions were made by
aqueous ginger extract. Twenty four hours of transferring 1 ml from one tube to another. Then,
broth culture of the two microorganisms an overnight broth culture of the different test
(Staphylococcus aureus and Escherichia coli) organisms were adjusted to McFarland turbidity
were suspended into sterile nutrient broth. It was standard and 100 μl of the different cell
standardized by gradually adding 9% normal suspensions were added to each of the separate
saline to compare its turbidity to McFarland tubes. The tubes were incubated aerobically at
8
standard of 0.5 which is approximately 1 x 10 37°C for 18 hours. Negative control tube was
colony forming units per ml. Petri-dishes were made by pouring 1ml of normal saline instead of
prepared by loading about 25 ml of an the aqueous ginger extract. The lowest
autoclaved nutrient agar on sterile plates and left concentration of the dilution without bacterial
to solidify. 100 μl of a standardized culture growth was considered as the minimum inhibition
(adjusted to 0.5 McFarland) of the two different concentration.
organisms were added onto the different agar
plates. The surface of each plate was drilled
using a sterile cork borer (6 mm) and 3 wells 2.6 Statistical Analysis
were punched out on each plate followed by
loading of 100 μl of the aqueous ginger extract of All analyses were carried out in triplicate
different concentration in the wells and allowed to determination and results were expressed as
diffuse at room temperature for 2 hours. The mean S . Students -test was used for
plates were incubated at 37ºC for 18-24 hours for comparison. The data analysis was done using
bacterial pathogens. The diameters of the one way analysis of variance (ANOVA) Post Hoc
inhibition zone (mm) were measured. The Turkey Graph Pad prism computer software
susceptibility of the two different organisms to version 5.01. P-value < 0.05 was considered
different concentration of aqueous ginger significant.
extracts were assayed using standard
procedures [11,34]. The experiment was
repeated thrice, for each replicate, the readings
3. RESULTS
were taken in three different fixed directions and
the average values were recorded [11, 34]. “The 3.1 Result for Gas-Chromatography–
inhibitory responses were classified as potent
Mass Spectrometry of ginger
response, ++++, zone diameter >30 mm; strong
response, +++, zone diameter between 21-30 (Zingiber officinale) rhizome
mm; moderate response, ++, zone diameter
between 16-20 mm; weak response, +, zone The Gas-Chromatography–Mass Spectrometry
diameter between 10-15 mm; and little or no chromatogram and the compounds found in
response, zone diameter <10 mm” [34,39]. ginger rhizome are shown in Fig. 1 and
Table 1.
2.5.3 Minimum Inhibitory Concentration (MIC)
of aqueous Zingiber officinale extract 3.2 Minimum Inhibitory Concentration
(MIC) for aqueous ginger and
“Minimum inhibition concentration is the lowest Augmentin antibiotic against
concentration of ginger extract that inhibited the Staphylococcus aureus and
growth of the test organisms as indicated by the Escherichia coli
absence of visible turbidity in the tube compared
with the control tubes” [16,34]. The MIC of the
The values for the MICs of aqueous ginger
aqueous ginger rhizome extract was determined
(Zingiber officinale) and Augmentin antibiotic
according to standard method [16,34]. The MIC
against Staphylococcus aureus and Escherichia
of the aqueous ginger extract was assayed using
coli are shown in Table 4.
serial dilution method. In this method, a total of 1

10
Momoh and Olaleye; MRJI, 32(7): 7-31, 2022; Article no.MRJI.92461

Fig. 1. Gas-Chromatography–Mass Spectrometry chromatogram of ginger (Zingiber officinale) rhizome

11
Momoh and Olaleye; MRJI, 32(7): 7-31, 2022; Article no.MRJI.92461

Table 1. Compounds found in the ginger (Zingiber officinale) rhizome analyzed using Gas-Chromatography–Mass Spectrometry

PK# RT Peak Name of the compound Molecular Molecular Ref# CAS# Qual.
Area Formulae Weight
(%) (g/mol)
1 4.295 1.00 Hexanal C6H12O 100.1589 3833 000066-25-1 90
2 8.203 0.76 Octanal C8H16O 128.2120 12694 000124-13-0 90
3 11.384 6.58 Dodecane C12H26 170.3348 39972 000112-40-3 97
4 11.504 2.19 Decanal C10H20O 156.2652 29133 000112-31-2 98
5 12.849 16.94 Tridecane C13H28 184.3614 51394 000629-50-5 96
6 15.315 3.58 Benzene, 1-(1,5-dimethyl-4- C15H22 202.3352 66866 000644-30-4 99
hexenyl)-4-methyl-
7 15.464 5.85 1,3-Cyclohexadiene, 5-(1,5- C15H24 204.3511 68761 000495-60-3 94
dimethyl-4-hexenyl)-2-methyl-,
[S-(R*,S*)]-
8 15.579 2.31 alpha.-Farnesene C15H24 204.3511 68573 000502-61-4 81
9 15.624 1.51 beta.-Bisabolene C15H24 204.3511 68571 000495-61-4 98
10 15.830 3.89 Cyclohexene, 3-(1,5-dimethyl- C15H24 204.3511 68741 020307-83-9 93
4-hexenyl)-6-methylene-, [S-
(R*,S*)]-
11 16.208 0.91 Cyclohexanemethanol, 4- C15H26O 222.3663 85863 000639-99-6 83
ethenyl-.alpha.,.alpha.,4-
trimethyl-3-(1-methylethenyl)-,
[1R-(1.alpha.,3.alpha.,
4.beta.)]-
12 16.294 0.82 1,6,10-Dodecatrien-3-ol, C15H26O 222.3663 85747 007212-44-4 91
3,7,11-trimethyl-
13 16.540 1.00 4-(1-Hydroxyallyl)-2- C10H12O 180.2005 47432 112465-50-6 96
methoxyphenol
14 16.660 0.87 trans-Sesquisabinene hydrate C15H26O 222.3663 85739 145512-84-1 95
15 16.935 1.33 (1S,2R,5R)-2-Methyl-5-((R)-6- C15H26O 222.3663 85811 058319-05-4 87

12
Momoh and Olaleye; MRJI, 32(7): 7-31, 2022; Article no.MRJI.92461

PK# RT Peak Name of the compound Molecular Molecular Ref# CAS# Qual.
Area Formulae Weight
(%) (g/mol)
methylhept-5-en-2-yl)
bicyclo[3.1.0]hexan-2-ol
16 17.410 15.95 Butan-2-one, 4-(3-hydroxy-2- C11H14O3 194.23 59345 303187-89-5 98
methoxyphenyl)-
17 17.787 1.65 3-Cyclohexene-1-methanol, C15H26O 222.3663 85832 023178-88-3 56
.alpha., 4-dimethyl-.alpha.-(4-
methyl-3-pentenyl)-, [R-
(R*,R*)]-
18 17.890 1.04 2,6,10-Dodecatrien-1-ol, C15H26O 222.3663 85748 004602-84-0 55
3,7,11-trimethyl-
19 18.422 0.63 1,2-Cyclohexanediol, cyclic C6H10O3S 162.21 33637 019456-19-0 41
sulfite, trans-
20 19.138 1.72 2-Butanone, 4-(2,6,6-trimethyl- C13H22O 194.3132 59811 039721-65-8 38
2-cyclohexen-1-yl)-, (R)-
21 19.773 0.53 Cyclopropane carboxamide, 2- C13H21NO 207.31 71615 331416-19-4 51
cyclopropyl-2-methyl-N-(1-
cyclopropylethyl)-
22 21.272 0.62 1,6,10,14-Hexadecatetraen-3- C20H34O 290.4834 150239 001113-21-9 59
ol, 3, 7,11,15-tetramethyl-,
(E,E)-
23 22.388 0.74 1,6,10,14,18,22- C30H50O 426.7174 249597 097232-74-1 80
Tetracosahexaen-3-ol,
2,6,10,15,19,23-hexamethyl-,
(all-E)-(.+/-.)-
24 23.080 2.23 (E)-1-(4-Hydroxy-3- C17H24O3 276.3707 136509 863913-65-9 95
methoxyphenyl) dec-3-en-5-
one
25 23.166 7.39 3-Decanone, 1-(4-hydroxy-3- C17H26O3 278.3865 138319 027113-22-0 98
methoxyphenyl)-

13
Momoh and Olaleye; MRJI, 32(7): 7-31, 2022; Article no.MRJI.92461

PK# RT Peak Name of the compound Molecular Molecular Ref# CAS# Qual.
Area Formulae Weight
(%) (g/mol)
26 23.692 6.36 1-(4-Hydroxy-3- C17H24O3 276.3707 136506 000555-66-8 98
methoxyphenyl) dec-4-en-3-
one
27 24.019 2.00 1-(4-Hydroxy-3- C17H24O4 292.4 151730 061871-71-4 96
methoxyphenyl) decane-3,5-
dione
28 24.356 0.61 Vanillin C8H8O3 152.15 26591 000121-33-5 45
29 24.522 0.75 5-Hydroxy-1-(4-hydroxy-3- C17H26O4 294.391 153613 039886-76-5 97
methoxyphenyl) decan-3-one
30 24.911 0.59 1-(4-Hydroxy-3- C19H30O3 306.4397 165516 027113-23-1 93
methoxyphenyl) dodecan-3-
one
31 25.392 1.25 1-(4-Hydroxy-3- C15H20O3 248.32 109653 211176-76-0 74
methoxyphenyl) oct-4-en-3-one
32 25.512 0.79 (3R,5S)-1-(4-Hydroxy-3- C21H32O6 380.4752 227162 143615-75-2 99
methoxyphenyl) decane-3,5-
diyl diacetate
33 25.684 1.19 1-(3,4-Dimethoxyphenyl) C22H34O6 394.5018 235343 053254-52-7 42
decane-3,5-diyl diacetate
34 26.433 0.93 (E)-1-(4-Hydroxy-3- C21H32O3 332.4770 190321 1278586-98- 95
methoxyphenyl) tetradec-3-en- 3
5-one
35 27.114 2.27 1-(4-Hydroxy-3- C21H32O3 332.5 190316 036752-54-2 97
methoxyphenyl) tetradec-4-en-
3-one
36 27.526 1.21 1-(4-Hydroxy-3- C21H32O4 348.5 204403 079067-90-6 97
methoxyphenyl) tetradecane-
3,5-dione

14
Momoh and Olaleye; MRJI, 32(7): 7-31, 2022; Article no.MRJI.92461

100 44

56

41
O

43
50

57
27 29

39 45
55 72
42 82
28 58 67 71
15 18 26 31 38 40 46 51 53
60
62 65 69 73 77 79 81 83 85 87 99
0
10 20 30 40 50 60 70 80 90 100 110
(mainlib) Hexanal

Structure of Hexanal Structure of Octanal


100 57 43
100

41
O
43

57
71
50
29
50 70
41
85
27 68 82
55
29 112
27 98
39 95
112 170
53 127
0 15 63 67 77 91 141
10 20 30 40 50 60 70 80 90 100 110 120 130 140 150 160 170 180
(mainlib) Dodecane 15 31 60 128 138
37 79 91 123 155
0
10 20 30 40 50 60 70 80 90 100 110 120 130 140 150 160 170
Structure of Dodecane (mainlib) Decanal

Structure of Decanal
57 100 119
100
43
132

41
50 105

71
50 55
91 145 202
39 69 83
41 27 77
65 95
53 79
29 85 31 58 63 110 128 152 159 187
0
20 30 40 50 60 70 80 90 100 110 120 130 140 150 160 170 180 190 200 210
(mainlib) Benzene, 1-(1,5-dimethyl-4-hexenyl)-4-methyl-
27
39 99 113 127
53 141 155 184
0
20 30
(mainlib) Tridecane
40 50 60 70 80 90 100 110 120 130 140 150 160 170 180 190
Structure of Benzene, 1-(1,5-dimethyl-4-hexenyl)-4-methyl-

Structure of Tridecane

15
Momoh and Olaleye; MRJI, 32(7): 7-31, 2022; Article no.MRJI.92461

100 93 100 41

93
119

55 69

50 50 107
41 79
69
91
43 81 119
77 91 77
29 39 53
56 123 135
105 27
39 67
27 53 204
79 161 51 57
43 51 65 109 133 147 161 204
63 189
0 0
20 30 40 50 60 70 80 90 100 110 120 130 140 150 160 170 180 190 200 210 20 30 40 50 60 70 80 90 100 110 120 130 140 150 160 170 180 190 200 210
(mainlib) 1,3-Cyclohexadiene, 5-(1,5-dimethyl-4-hexenyl)-2-methyl-, [S-(R*,S*)]- (mainlib) α-Farnesene

Structure of 1,3-Cyclohexadiene, 5-(1,5-dimethyl-4-hexenyl)-2- Structure of alpha.-Farnesene


methyl-, [S-(R*,S*)]-
100 69 100 69

41 93 41

50 50 93

55 91
67 79 109 204 57 161
77
91 119 161 109 120 133
55 39 71 79
39 77 81 135 27 67 105 204
27 43 43 53 112
65 147 189 51 147
51 97 175 29 45 89 175 189
0 0
20 30 40 50 60 70 80 90 100 110 120 130 140 150 160 170 180 190 200 210 220 20 30 40 50 60 70 80 90 100 110 120 130 140 150 160 170 180 190 200 210
(mainlib) β-Bisabolene (mainlib) Cyclohexene, 3-(1,5-dimethyl-4-hexenyl)-6-methylene-, [S-(R*,S*)]-

Structure of beta.-Bisabolene Structure of Cyclohexene, 3-(1,5-dimethyl-4-hexenyl)-6-methylene-, [S-


(R*,S*)]-

Structure of Cyclohexanemethanol, 4-ethenyl-.alpha.,.alpha.,4-


Structure of 1,6,10-Dodecatrien-3-ol, 3,7,11-trimethyl-
trimethyl-3-(1-methylethenyl)-, [1R-(1.alpha.,3.alpha., 4.beta.)]-

16
Momoh and Olaleye; MRJI, 32(7): 7-31, 2022; Article no.MRJI.92461

100 137

OH

50
124
HO
180

77 91 109
51 55 65 79 94 119
39 152
29 43 74 81 89 163
0
20 30 40 50 60 70 80 90 100 110 120 130 140 150 160 170 180 190
(mainlib) 4-(1-Hydroxyallyl)-2-methoxyphenol

Structure of 4-(1-Hydroxyallyl)-2-methoxyphenol Structure of trans-Sesquisabinene hydrate

100 69 100 137

82

H O

119
43 194
50 93 HO 50 O
55 H
67 OH
109
71 79 95

137 119
53 43 91 151
39 77 207
57 97 161 77 122
29 151 94 107
45 179 189 39 45 51 55 65 179
165 222 69 74 89 115 161
0 0
20 30 40 50 60 70 80 90 100 110 120 130 140 150 160 170 180 190 200 210 220 230 30 40 50 60 70 80 90 100 110 120 130 140 150 160 170 180 190 200 210
(mainlib) (1S,2R,5R)-2-Methyl-5-((R)-6-methylhept-5-en-2-yl)bicyclo[3.1.0]hexan-2-ol (mainlib) Butan-2-one, 4-(3-hydroxy-2-methoxyphenyl)-

Structure of (1S,2R,5R)-2-Methyl-5-((R)-6-methylhept-5-en-2- Structure of Butan-2-one, 4-(3-hydroxy-2-methoxyphenyl)-


yl)bicyclo[3.1.0]hexan-2-ol

17
Momoh and Olaleye; MRJI, 32(7): 7-31, 2022; Article no.MRJI.92461

100 69 69
100

OH

50 OH
43 50 41
41
109 119
81 93
67 95 81
71 79 91 135
55 77 161 204
93
53 67
189 222 55 95 107 121 136
0 29 39 57 79 84 91
40 50 60 70 80 90 100 110 120 130 140 150 160 170 180 190 200 210 220 230
(mainlib) 3-Cyclohexene-1-methanol, α,4-dimethyl-α-(4-methyl-3-pentenyl)-, [R-(R*,R*)]- 15 31 51 59 97 127 147 153 161 179 191 204 222
0
10 20 30 40 50 60 70 80 90 100 110 120 130 140 150 160 170 180 190 200 210 220 230
(mainlib) 2,6,10-Dodecatrien-1-ol, 3,7,11-trimethyl-
Structure of 3-Cyclohexene-1-methanol, .alpha., 4-dimethyl-.alpha.-
(4-methyl-3-pentenyl)-, [R-(R*,R*)]- Structure of 2,6,10-Dodecatrien-1-ol, 3,7,11-trimethyl-
100 43

41

50 95 109

55
69
81
29 136
39 67 123 151
53 71 93
79 84 194
57 97 161 176
45 51 73 147
0
20 30 40 50 60 70 80 90 100 110 120 130 140 150 160 170 180 190 200
(mainlib) 2-Butanone, 4-(2,6,6-trimethyl-2-cyclohexen-1-yl)-, (R)-

Structure of 1,2-Cyclohexanediol, cyclic sulfite, trans-


Structure of 2-Butanone, 4-(2,6,6-trimethyl-2-cyclohexen-1-yl)-, (R)-

Structure of Cyclopropane carboxamide, 2-cyclopropyl-2-methyl-N- Structure of 1,6,10,14-Hexadecatetraen-3-ol, 3, 7,11,15-tetramethyl-,


(1-cyclopropylethyl)- (E,E)-

18
Momoh and Olaleye; MRJI, 32(7): 7-31, 2022; Article no.MRJI.92461

100 137

50
HO

276
41 55 65 77 91 122 151 160
0
27 97 107 179 217 243
20 30 40 50 60 70 80 90 100 110 120 130 140 150 160 170 180 190 200 210 220 230 240 250 260 270 280 290
(mainlib) (E)-1-(4-Hydroxy-3-methoxyphenyl)dec-3-en-5-one
Structure of 1,6,10,14,18,22-Tetracosahexaen-3-ol, 2,6,10,15,19,23-
hexamethyl-, (all-E)-(.+/-.)- Structure of (E)-1-(4-Hydroxy-3 methoxyphenyl) dec-3-en-5-one
100 137 100 137

O O

HO HO

50 205
50 O
276
278
55
151 179 151
119
119 91
57 77 125
91 41 94
65 107 145
43 65 77 107 163 173 187 219 227 245 261
161 194 247 0
0 30 40 50 60 70 80 90 100 110 120 130 140 150 160 170 180 190 200 210 220 230 240 250 260 270 280 290
30 40 50 60 70 80 90 100 110 120 130 140 150 160 170 180 190 200 210 220 230 240 250 260 270 280 290 (mainlib) 1-(4-Hydroxy-3-methoxyphenyl)dec-4-en-3-one
(mainlib) 3-Decanone, 1-(4-hydroxy-3-methoxyphenyl)-

Structure of 3-Decanone, 1-(4-hydroxy-3-methoxyphenyl)- Structure of 1-(4-Hydroxy-3-methoxyphenyl) dec-4-en-3-one

19
Momoh and Olaleye; MRJI, 32(7): 7-31, 2022; Article no.MRJI.92461

100 137

O O

50
HO

292
150

43 55 122 179 193


65 71 77 84 91 99 107 161 203 221 231
0
30 40 50 60 70 80 90 100 110 120 130 140 150 160 170 180 190 200 210 220 230 240 250 260 270 280 290 300
(mainlib) 1-(4-Hydroxy-3-methoxyphenyl)decane-3,5-dione

Structure of Vanillin
Structure of 1-(4-Hydroxy-3-methoxyphenyl) decane-3,5-dione
100 137

HO
205
50

304

55
119 151
41 69 91 124
29 65 77 94 107 163 173 187 215 261 273 287
0
20 30 40 50 60 70 80 90 100 110 120 130 140 150 160 170 180 190 200 210 220 230 240 250 260 270 280 290 300 310 320
(mainlib) 1-(4-Hydroxy-3-methoxyphenyl)dodec-4-en-3-one
Structure of 5-Hydroxy-1-(4-hydroxy-3-methoxyphenyl) decan-3-one
Structure of 1-(4-Hydroxy-3-methoxyphenyl) dodecan-3-one

Structure of 1-(4-Hydroxy-3-methoxyphenyl) oct-4-en-3-one Structure of (3R,5S)-1-(4-Hydroxy-3-methoxyphenyl)decane-3,5-diyl


diacetate

20
Momoh and Olaleye; MRJI, 32(7): 7-31, 2022; Article no.MRJI.92461

100 43
151
O

O O O
O

50
O

203
164
177 189
394
107 138
55 243
29 67 77 91 121
217 231 259
274
334
280 292 303
0
20 40 60 80 100 120 140 160 180 200 220 240 260 280 300 320 340 360 380 400
(mainlib) 1-(3,4-Dimethoxyphenyl)decane-3,5-diyl diacetate

Structure of (E)-1-(4-Hydroxy-3-methoxyphenyl) tetradec-4-en-3-one


Structure of 1-(3,4-Dimethoxyphenyl)decane-3,5-diyl diacetate
137 100 137
100
O
O
O O

HO O
205
50
50 HO

332
348
150

55 151 43 55 179
119 77 91 107 122 161
197
221 236 290 320
43 91 0
69 81 94 107 145 163 177 187 30 40 50 60 70 80 90 100 110 120 130 140 150 160 170 180 190 200 210 220 230 240 250 260 270 280 290 300 310 320 330 340 350 360
0 219 289 301 315 (mainlib) 1-(4-Hydroxy-3-methoxyphenyl)tetradecane-3,5-dione
30 40 50 60 70 80 90 100 110 120 130 140 150 160 170 180 190 200 210 220 230 240 250 260 270 280 290 300 310 320 330 340
(mainlib) 1-(4-Hydroxy-3-methoxyphenyl)tetradec-4-en-3-one

Structure of 1-(4-Hydroxy-3-methoxyphenyl) tetradecane-3,5-dione


Structure of 1-(4-Hydroxy-3-methoxyphenyl) tetradec-4-en-3-one

Fig. 2. Mass spectrum and structure of 36 different compounds obtained during GC-MS analysis of ginger rhizome

21
Momoh and Olaleye; MRJI, 32(7): 7-31, 2022; Article no.MRJI.92461

Table 2. The qualitative phytochemical constituents of aqueous ginger extract

Phytochemical constituent Inference


Saponins Present
Tannins Present
Simple phenolics Present
Flavonoids Present
Glycosides Present
Alkaloids Present
Carbohydrate Present
Reducing sugar Present

Fig. 3a. Zone of inhibition at 250 mg/ml of the Fig. 3b. Zone of inhibition at 250 mg/ml of
aqueous extract of ginger against the aqueous extract of ginger against
Staphylococcus aureus Staphylococcus aureus

22
Momoh and Olaleye; MRJI, 32(7): 7-31, 2022; Article no.MRJI.92461

Fig. 3d. Zone of inhibition at 500 mg/ml of


Fig. 3c. Zone of inhibition at 500 mg/ml of the the aqueous extract of ginger against
aqueous extract of ginger against Staphylococcus aureus
Staphylococcus aureus

Fig. 3e. Zone of inhibition at 250 mg/ml of the Fig. 3f. Zone of inhibition at 500 mg/ml of the
aqueous extract of ginger against Escherichia aqueous extract of ginger against
coli Escherichia coli

Fig. 3. Zone of inhibition of, aqueous extracts of ginger (Zingiber officinale) rhizome against Staphylococcus aureus and Escherichia coli at 250
and 500 mg/ml

23
Momoh and Olaleye; MRJI, 32(7): 7-31, 2022; Article no.MRJI.92461

Table 3. Zone of inhibition of aqueous extract of ginger (Zingiber officinale) against Escherichia coli and Staphylococcus aureus

Test organisms Aqueous extract of Zone of Interpretation Concentration of Zone of Interpretation


ginger inhibition for Augmentin solution inhibition of
concentration aqueous used (mg/ml) Augmentin
(mg/ml) extract of solution (mm)
ginger
(mm)
c d
Escherichia coli 250 8.19±1.33 little or no response 7.50 17.23±1.67 ++
c c
Staphylococcus 250 9.85±0.39 little or no response 7.50 21.13±1.34 +++
aureus
b bc
Escherichia coli 500 13.62±2.03 + 15 23.00±2.88 +++
a a
Staphylococcus 500 16.73±1.83 ++ 15 30.50±2.64 ++++
aureus
Values are represented as mean ± SD. A comparison across the column was done using One way ANOVA Post Hoc Turkey test. The superscript a has the highest value
followed by b, c and d has the lowest value. A. P<0.05 was considered statistically significant. The inhibitory responses were classified as potent response, ++++, zone
diameter >30 mm; strong response, +++, zone diameter between 21-30 mm; moderate response, ++, zone diameter between 16-20 mm; weak response, +, zone diameter between
10-15 mm; and little or no response, zone diameter <10 mm

Table 4. Minimum Inhibitory Concentration (MIC) of aqueous extract of Zingiber officinale rhizome and Augmentin antibiotic against
Staphylococcus aureus and Escherichia coli

Organisms Staphylococcus aureus Escherichia coli


MIC for aqueous extract of ginger (mg/ml) 125.00 250.00
MIC for Augmentin solution (mg/ml) 7.81 15.63

24
Momoh and Olaleye; MRJI, 32(7): 7-31, 2022; Article no.MRJI.92461

4. DISCUSSION Hydroxy-1-(4-hydroxy-3-methoxyphenyl) decan-


3-one found in the rhizome of ginger is also
Ginger rhizomes and its products have been called 6-gingerol. 6-Gingerol is one of the
used widely as a food spice as well as in herbal primary bioactive phenylpropanoid of the rhizome
medicine for the protection and treatment of of ginger and has been reported to have a
diseases. Fig. 1 shows the Gas-Chromatography pharmacological activities including; antioxidant
– Mass Spectrometry chromatogram of ginger effect, anti-cancer, anti-inflammation, anti-
rhizome. A total of 36 compounds were identified oxidation and possess cytotoxic activity and
consisting of two prominent compounds and 34 Inhibit of angiogenesis, [44]. 1-(4-Hydroxy-3-
minor compounds (Table 1). The two major methoxyphenyl) oct-4-en-3-one is also called 4-
compounds and their percentage abundance are: Shogaol and possesses medicinal uses. 1,3-
Tridecane with molecular formula of C13H28 Cyclohexadiene, 5-(1,5-dimethyl-4-hexenyl)-2-
(RT=12.849 and peak area=16.94%) and Butan- methyl-, [S-(R*,S*)]- is also called alpha-
2-one, 4-(3-hydroxy-2-methoxyphenyl)- with Zingiberene. 1,3-Cyclohexadiene, 5-(1,5-
molecular formula of C11H14O3 (RT=17.410 and dimethyl-4-hexenyl)-2-methyl-, [S-(R*,S*)]- was
peak area=15.95%). the most abundant compound found in the
aqueous ginger extract according to the GC-MS
In one of our study, it was observed that the GC- analysis carried out by Momoh et al. [40]
MS analysis of aqueous ginger extract contains Zingiberene is the primary terpenoid in ginger.
11 different compounds: Propane, 1-chloro-2- (E)-1-(4-Hydroxy-3-methoxyphenyl) tetradec-4-
nitro-, Cyclopropene, 1-methyl-3-(2-methyl en-3-one compound found in the plant that is
cyclopropyl)-, 1,3-Cyclohexadiene, 5-(1,5- used in this study is also called 10-Shogaol. A
dimethy 4-hexenyl)-2-methyl-, [S-(R*,S*) ]- , study shows that “10-Shogaol possesses
Preg-4-en-3-one, 17.alpha.-hydroxy-17.beta.- antioxidant activity, promoted human normal
cyano, (E)-.beta.-Famesene, 3-Bromo-N epidermal keratinocytes and dermal fibroblasts
(3,5dichlorophenyl) -benzamide, TMS derivative, cell growths. It enhances growth factor
Trisiloxane, 1,1,1,5,5,5-hexamethyl-3,3-bis- production in transforming growth factor-β (TGF-
[(trimethylsilyl)oxy]-, 2,5-Dihydroxybenzoic acid, β), platelet derived growth factor-αβ (P GF-αβ)
3TMS derivative, Cyclodecasiloxane, and vascular endothelial growth factors (VEGF)
eicosamethyl-, 3-Isopropoxy-1,1,1,5,5,5- of both cells. In the in vitro wound healing assay
hexamethyl-3-(trimethylsiloxy)-trisiloxane and for 12 or 24 h, with 10-shogaol, the fibroblasts
1,1,3,3,5,5,7,7,9,9,11,11,13,13- and keratinocytes migrated more rapidly than the
tetradecamethylheptasiloxane. vehicle control group” [45].

1,3-Cyclohexadiene, 5-(1,5-dimethy 4-hexenyl)- The preliminary qualitative analysis of the


2-methyl-, [S-(R*,S*) ]- was the most abundant different secondary metabolites present in the
compound in the aqueous ginger extract with aqueous extracts of ginger was investigated. The
peak area of 33.98 % and retention time of aqueous ginger showed that they contain simple
14.554 [40]. “Two highly alkylated gingerols, 10- phenolics, alkaloids, glycosides, saponins,
gingerol and 12-gingerol, seem to be effective in flavonoids, tannins,, carbohydrate and reducing
inhibiting the growth of oral pathogens at a MIC sugar. “Tannins which are phenolic compounds
range of 6–30 μg/mL and killing these oral tend to dissolve in water and tend to be polar.
pathogens at a minimum bactericidal Terpenoids are fat soluble. One of the terpenoids
concentration range of 4–20 μg/mL” [41]. “In which has the potential as an antimicrobial is
another study, it was observed that Four ginger triterpenoid. Flavonoids are generally more
components namely, 6-dehydrogingerdione, 10- soluble in water or polar solvents because they
gingerol, 6-shogaol, and 6-gingerol have shown bonds with hydroxyl groups. Glycosides are
antibacterial effects against extensively drug- compounds that contain non-sugar and sugar
resistant Acinetobacter baumannii” [42]. Keith components. Saponins are generally in the form
and Singletary [43] study enumerates the of glycosides so they tend to be polar. Saponins
biological importance of vanillin to include the are surface active compounds that produce foam
following: pain relief, antidepressant, antisickling, if shaken in water. This happens because
antianxiety, protect against nerve damage and saponins have polar and non-polar groups that
neurodegeneration, correct blood glucose and will form micelles. When the micelle is formed the
lipid dysregulation. 1-(4-Hydroxy-3- polar group will face out while the non-polar
methoxyphenyl) decane-3,5-dione is also called groups face inside so it looks like foams. The
gingerdione and possess vital medicinal uses. 5- antimicrobial activity of plants is due to saponins,

25
Momoh and Olaleye; MRJI, 32(7): 7-31, 2022; Article no.MRJI.92461

essential oils, tannins, phenolic compounds and Pseudomonas aeruginosa PA14” [52].
flavonoids” [46]. It is interesting to note that even “Methanolic fraction and crude extract of ginger
crude extracts of these plants showed good inhibited biofilm formation, glucan synthesis, and
activity against multidrug resistant strains where the adherence of Streptococcus mutans by
modern antibiotic drug has limited effect. downregulating virulence genes. Consistent with
the in-vitro study, a reduction in caries
Escherichia coli and Staphylococcus aureus development caused by Streptococcus mutans
were selected for the study and tested against was found in a treated group of rats” [53].
aqueous ginger and augmentin. In our study, the Furthermore, “an in vitro study revealed that
aqueous ginger extract exhibited moderate gingerenone-A and 6-shogaol found in ginger
response antimicrobial activity against exhibited an inhibitory effect on Staphylococcus
Staphylococcus aureus and weak response aureus by inhibiting the activity of 6-
against Escherichia coli with zone of inhibition of hydroxymethyl-7, 8-dihydropterin
16.73±1.83 and 13.62±2.03 at 500 mg/dl pyrophosphokinase (HPPK) in pathogen” [54].
respectively. The study shows that at 250 mg/ml, “6-Hydroxymethyl-7,8-dihydropterin
the aqueous ginger extracts exhibited little or no pyrophosphokinase (HPPK) is an enzyme of the
response with zone of inhibition of 9.85±0.39 and folic acid biosynthetic pathway that catalyzes the
8.19±1.33 against S. aureus and E. coli magnesium-dependent pyrophosphorylation of 6-
respectively. The poor activity of the aqueous hydroxymethyl-7,8-dihydropterin, utilising ATP to
ginger used in this study may be due to the low form 6-hydroxymethyl-7,8-dihydropterin
concentration of the active ingredients (5- pyrophosphate. The product formed can be used
Hydroxy-1-(4-hydroxy-3-methoxyphenyl) decan- for design of inhibitors with a potential
3-one also called 6-gingerol has 0.75%, 1-(4- therapeutic value. Zingiber officinale essential oil
Hydroxy-3-methoxyphenyl) dec-4-en-3-one (6- exhibited inhibitory activity against Fusarium
Shogaol) has 6.36%, (E)-1-(4-Hydroxy-3- verticillioides with an MIC of 2500 µg/mL” [50].
methoxyphenyl) tetradec-3-en-5-one (10- Ginger has been shown to be effective against
Isoshogaol ) has 0.93%, 1-(4-Hydroxy-3- the growth of both gram-negative and gram-
methoxyphenyl) decane-3,5-dione (Gingerdione) positive bacteria like: Escherichia coli,
has 2.00%, 1-(4-Hydroxy-3-methoxyphenyl) Salmonella typhi and Staphylococcus aureus
tetradecane-3,5-dione (10-Gingerdione) has [55].
1.21%, 1,3-Cyclohexadiene, 5-(1,5-dimethyl-4-
hexenyl)-2-methyl-, [S-(R*,S*)]- ( Zingiberene) In a study carried out by Samuel-Penu and
has 5.85% etc ) found in the ginger as obtained Baridakara [56], it was observed that the
during GC-MS analysis (Table 1). aqueous extract of ginger (Zingiber officinale) did
not inhibit any growth of bacteria both at 100 and
Several natural spices and herbs have been 50% concentrations while the ethanolic extract of
developed into natural effective antimicrobial ginger inhibited and a zone diameter of 11 mm
agents against many pathogenic microorganisms was recorded both for Staphylococcus
[47]. Zingiber officinale has been reported to epidermidis and Bacillus sp. at the 100%
have antifungal, antibacterial and antiviral concentration. In another study, it was observed
activities [48,49]. Different research works have that the zone diameter for ethanolic extract of
shown the antimicrobial activities of ginger. A ginger rhizomes against E.coli varies from
study has shown the antifungal activity of 8.50±0.12 to 15.50±0.30 at concentration
Zingiber officinale essential oil on Fusarium between 25 to 200 µg/ml and 9.30±0.32 to
verticillioides and it reduces the biosynthesis of 13.55±0.20 for S. aureus at the same
ergosterol; affecting membrane integrity; concentration [57]. In a research work carried out
decreasing the production of fumonisin B1 and by Njobdi1 et al. [58], it was observed that “at a
fumonisin B2. [50]. “The formation of biofilm is a concentrations of 10, 20, 30 , and 40 mg/ml, the
factor that can cause infection and antimicrobial zones of inhibition of dried Z. officinale extracts
resistance. A study found that Zingiber officinale on S. aureus were 11.00 ±1.41 mm, 13.5 ± 0.71
inhibited the growth of a multidrug-resistant strain mm, 14.00±2.66 mm and 17.5 ± 0.87 mm
of Pseudomonas aeruginosa by affecting respectively and on E. coli were 6.00 ± 2.83 mm,
membrane integrity and inhibiting biofilm 7.5 ± 2.12 mm, 8.00 ± 2.83 mm and 14.5± 6.08
formation” [51]. Furthermore, “treatment with mm respectively. Fresh ginger showed
ginger extract blocked biofilm formation via a 15.00±1.40 mm and 12.00±2.83 mm at 100%
reduction in the level of bis-(3´-3´)-cyclic dimeric and 50% concentrations respectively on S.
guanosine monophosphate (c-di-GMP) in aureus and 15.00±3.54 mm and 13.00±2.66 mm

26
Momoh and Olaleye; MRJI, 32(7): 7-31, 2022; Article no.MRJI.92461

on E. coli respectively but has no effect at 25% bacteria have an additional outer membrane on
and 12.5% concentration on both organisms”. their cell wall, the entry of secondary metabolites
Gull et al. [59] study shows that the aqueous from ginger and the augmentin solution may be
extract of ginger inhibits the growth of different interrupted and its effects are lesser on the cell.
pathogenic bacteria like: E.coli, S. aureus, However, gram positive bacteria lack the outer
S.typhi, S. epidermidis, K. pneumonia, Shigella, membrane and therefore they are more
B. subtilis, and P. aeruginos with zone of susceptible to, easily entering of these
inhibition ranging from 11 ± 0 to 13 ± 0.47 mm compounds. The study shows that the minimum
with concentration ranging from 25 to 200. inhibitory concentration (MIC) values for the
aqueous ginger for Staphylococcus aureus and
Amoxicillin/clavulanic acid, also known as co- Escherichia coli are 125 and 250 mg/ml and 7.81
amoxiclav or amox-clav, sold under the brand and 15.63 mg/ml for augmentin solution for the
name augmentin, is an antibiotic medication sane organisms respectively.
used for the treatment of a number of bacterial
infections. It is a combination consisting of MIC’s of ginger for E. coli (175 mg/ml),
amoxicillin, a β-lactam antibiotic, and potassium Staphylococcus aureus (125 mg/ml) and
clavulanate, a β-lactamase inhibitor. It is Salmonella (150 mg/ml) was observed in a study
specifically used for otitis media, urinary tract carried out by Virenda et al. [55]. Ponmurugan
infections, streptococcal pharyngitis, pneumonia, and Shyamkumar, [57] study shows that
cellulitis, and animal bites. Augmentin showed ethanolic extract of ginger rhizomes has MICs of
moderate response with zone of inhibition of 75.60 and 68.45 against E. coli and S. aureus
17.23±1.67 against Escherichia coli and strong respectively. Gull et al. [59], study indicate that
response with zone of inhibition of 21.13±1.34 aqueous extract of ginger has an MICs of 0.1
against Staphylococcus aureus at concentration and 0.6 mg/ml for E.coli and S. aureus
of 7.50 mg/ml respectively. At 15 mg/ml, the drug respectively. MIC of dried and fresh Z. officinale
(augmentin) showed strong response with zone extracts on E. coli and S. aureus isolates are
of inhibition of 23.00±2.88 against E. coli and both 2.5 mg/ml respectively [58].
potent response with zone of inhibition of 5. CONCLUSION
30.50±2.64 against S. aureus. In a study
consisting of 973 bacteria isolates, 823 were E. This study shows that thirty six compounds were
coli and 150 were Klebsiella spp. More of the identified in the ginger (Zingiber officinale
organisms were found to be susceptible to Roscoe) rhizome using GC-MS analysis with
amoxicillin-clavulanic acid than Ampicillin- tridecane with molecular formula of C13H28 being
sulbactam, regardless of the susceptibility testing the most abundant with peak area of 16.94% and
methodology used in their study [60]. Njobdi1 et retention time of 12.849. The preliminary
al. [58] study shows that Augmentin has a zone phytochemical analysis of the extract of Zingiber
of inhibition of 21.00±1.41 mm against E. coli. officinale shows the presence of secondary
Gram negative bacteria (Escherichia coli ) was metabolites like saponins, alkaloids, glycoside,
more resistant than gram positive bacteria Simple phenolics, tannins, flavonoids,
(Staphylococcus aureus), since they have lower carbohydrates and reducing sugar. The aqueous
zone of inhibition for both the aqueous ginger ginger extract has poor antibacterial activity
and augmentin solution as shown in Table 3. against Escherichia coli and Staphylococcus
These variations in inhibition may be because of aureus due to the low concentration of the active
differences in the composition and structure compounds obtained during GC-MS analysis.
surface between gram positive and gram This is an indication that the ginger extract has
negative bacteria. In addition to the cell wall and antibacterial potential.
cell membrane, gram negative bacteria have an
outer membrane composed of a phospholipid COMPETING INTERESTS
bilayer, which may be protective barrier against
Authors have declared that no competing
the ginger extract and augmentin solution used.
interests exist.
Furthermore, the cell walls of gram positive
bacteria have a large amount of peptidoglycan REFERENCES
and a small amount of lipid, while in the case of
gram negative bacteria, due to the presence of 1. Han YA, Song CW, Koh WS, Yon GH, Kim
an outer membrane, a large amount of lipid and YS, Ryu SY et al. Anti-inflammatory effects
a small amount of peptidoglycan is found in the of the Zingiber officinale Roscoe
cell wall. In addition to that, gram negative constituent 12-dehydrogingerdione in

27
Momoh and Olaleye; MRJI, 32(7): 7-31, 2022; Article no.MRJI.92461

lipopolysaccharide-stimulated raw 264.7 of methanolic leaf extract of Carica papaya


cells. Phytother Res. 2013;27(8):1200-5. against Plasmodium berghei NK65
DOI: 10.1002/ptr.4847, PMID 23027684. infection in Swiss mice. ARRB.
2. Stoner GD. Ginger: is it ready for prime 2020;35(12):183-97:Article
time? Cancer Prev Res (Phila). no.ARRB.60062.
2013;6(4):257-62. DOI: 10.9734/ARRB/2020/v35i1230323.
DOI: 10.1158/1940-6207.CAPR-13-0055, 11. Aderele OR, Rasaq AK, Momoh JO.
PMID 23559451. Phytochemical screening, mathematical
3. Ali BH, Blunden G, Tanira MO, Nemmar A. analysis and antimicrobial activity of
Some physico-chemical, pharmacological methanolic seed extract of Hunteria
and toxicological properties of ginger umbellata. Eur J Med Plants: 101583475.
(Zingerber officinale) – a review. Food 2020;31(16):1-17:Article no. EJMP.61248
Chem Toxicol. 2008;46(2):409-20. ISSN: 2231-0894.
DOI: 10.1016/j.fct.2007.09.085, PMID DOI: 10.9734/EJMP/2020/v31i1630325.
17950516. 12. Momoh JO, Adeniyi MO, Aderele OR. AAS
4. Suekawa M, Ishige A, Yuasa K, Sudo K, and GC-MS analysis of phytocomponents
Aburada M, Hosoya E. Pharmacological in the leaf, stem and root of Azadirachta
studies on ginger. I. Pharmacological indica A. Juss (Dongoyaro). BJPR.
actions of pungent constituents, (6). J 2017;15(4):1-12.
Pharmacobio Dyn. 1984;7(11):836-48. Article no.BJPR.30611.
DOI: 10.1248/bpb1978.7.836, PMID DOI: 10.9734/BJPR/2017/30611.
6335723. 13. Longe AO, Momoh JO, Asoro II. Gas
5. Gunathilake KDPP, Rupasinghe HPV, Pitts chromatography-mass spectrometry (GC-
NL. Formulation and characterization of a MS) analysis of phytocomponents in the
bioactives-enriched fruit beverage root, stem bark and leaf of Vernonia
designed for cardioprotection. Food Res amygdalin. World J Pharm Res.
Int. 2013;52(2):535-41. 2017;6(2):35-49.
DOI: 10.1016/j.foodres.2013.02.051. DOI: 10.20959/wjpr20172-7701.
6. Gunathilake KDPP, Wang Y, Rupasinghe 14. Oshin TT, Rasaq AK, Oluwafemi F,
HPV. Hypocholesterolemic and Momoh JO. Histological and cytotoxic
hypotensive effects of a fruit-based evaluation of Carica papaya seed oil and
functional beverage in spontane-ously its potential biodiesel feedstock. J Ecol
hypertensive rats fed with cholesterol-rich Eng. 2021;13(2):43-52.
diet. J Funct Foods. 2013;5(3):1392-401. DOI: 10.5897/JECE2020.0468.
DOI: 10.1016/j.jff.2013.05.007. 15. Chang WT, Thissen U, Ehlert KA, Koek
7. Wolfender JL, Marti G, Thomas A, MM, Jellema RH, Hankemeier T et al.
Bertrand S. Current approaches and Effects of growth conditions and
challenges for the metabolite profiling of processing on Rehmannia glutinosa using
complex natural extracts. J Chromatogr A. fingerprint strategy. Planta Med.
2015;1382:136-64. DOI: 2006;72(5):458-67.
10.1016/j.chroma.2014.10.091, PMID DOI: 10.1055/s-2005-916241, PMID
25464997. 16557461.
8. Kopka J. Current challenges and 16. Momoh JO, Olaleye ON. Biochemical
developments in GC–MS based metabolite Characterization and Molecular
profiling technology. J Biotechnol. Identification of Escherichia coli Isolate
2006;124(1):312-22. from Abattoir Wastes and Its Susceptibility
DOI: 10.1016/j.jbiotec.2005.12.012, PMID to ethanolic Root Extract of Azadirachta
16434119. indica (neem). JAMB. 2022;22(10):31-50.
9. Xiang Z, Wang XQ, Cai XJ, Zeng S. Article no.JAMB .91100.
Metabolomics study on quality control and DOI: 10.9734/JAMB/2022/v22i1030502.
discrimination of three Curcuma species 17. Zhang M, Viennois E, Prasad M, Zhang Y,
based on gas chromatograph-mass Wang L, Zhang Z, et al. Edible ginger-
spectrometry. Phytochem Anal. derived nanoparticles: A novel therapeutic
2011;22(5):411-8. approach for the prevention and treatment
DOI: 10.1002/pca.1296, PMID 21433157. of inflammatory bowel disease and colitis-
10. Momoh JO, Damazio OA, Oyegbami OM. associated cancer. Biomaterials. 2016;101:
GC–MS analysis and antimalarial activity 321-40.

28
Momoh and Olaleye; MRJI, 32(7): 7-31, 2022; Article no.MRJI.92461

DOI: 10.1016/j.biomaterials.2016.06.018, al. Effect of dietary supplementation of


PMID 27318094. ginger and turmeric rhizomes on
18. Nile SH, Park SW. Chromatographic angiotensin-1 converting enzyme (ACE)
analysis, antioxidant, anti-inflammatory, and arginase activities in L-NAME induced
and xanthine oxidase inhibitory activities of hypertensive rats. J Funct Foods.
ginger extracts and its reference 2015;17:792-801.
compounds. Ind Crops Prod. 2015;70: DOI: 10.1016/j.jff.2015.06.011.
238-44. 26. Abdurasid AA, Momoh JO, Aderele OR.
DOI: 10.1016/j.indcrop.2015.03.033. Experimental and Mathematical Model for
19. Citronberg J, Bostick R, Ahearn T, the Study of the in-vitro Susceptibility of
Turgeon DK, Ruffin MT, Djuric Z et al. antimicrobial Agents against Escherichi
Effects of Ginger Supplementation on Cell- coli Isolates Obtained From Abattoir
Cycle Biomarkers in the Normal-Appearing Centres in Ikorodu, Lagos, Nigeria. Curr J
Colonic Mucosa of Patients at Increased Appl Sci Technol. 2018;28(2):1-15:Article
Risk for Colorectal Cancer: Results from a no.CJAST.37290.
Pilot, Randomized, and Controlled Trial. DOI: 10.9734/CJAST/2018/37290.
Cancer Prev Res. 2013;6(4): 27. Wagenlehner FM, Naber KG, Weidner W.
271-81. Rational antibiotic therapy of urinary tract
DOI: 10.1158/1940-6207.CAPR-12-0327. infections. Med Monatsschr Pharm.
20. Kumar NV, Murthy PS, Manjunatha JR, 2008;31(10):385-90; quiz 391. PMID
Bettadaiah BK. Synthesis and quorum 18972869.
sensing inhibitory activity of key phenolic 28. De Francesco MA, Giuseppe R, Laura P,
compounds of ginger and their derivatives. Riccardo N, Nin M. Urinary tract infections
Food Chem. 2014;159:451-7. in Brescia, Italy: etiology of uropathogens
DOI: 10.1016/j.foodchem.2014.03.039, and antimicrobial resistance of common
PMID 24767081. uropathogens Med Sci Moni.
21. Suk S, Kwon GT, Lee E, Jang WJ, Yang 2007;13(6):136-44.
H, Kim JH, et al. Gingerenone A, a 29. Kashef N, Djavid GE, Shahbazi S.
polyphenol present in ginger, suppresses Antimicrobial susceptibility patterns of
obesity and adipose tissue inflammation in community-acquired uropathogens in
high-fat diet-fed mice. Mol Nutr Food Res. Tehran, Iran. J Infect Dev Ctries.
2017;61(10):1700139. 2010;4(4):202-6.
DOI: 10.1002/mnfr.201700139, PMID DOI: 10.3855/jidc.540, PMID 20440056.
28556482. 30. Biedenbach DJ, Moet GJ, Jones RN.
22. Wei CK, Tsai YH, Korinek M, Hung PH, El- Occurrence and antimicrobial resistance
Shazly M, Cheng YB et al. 6-Paradol and pattern comparisons among bloodstream
6-shogaol, the pungent compounds of infection isolates from the SENTRY
ginger, promote glucose utilization in Antimicrobial Surveillance Program (1997-
adipocytes and myotubes, and 6-paradol 2002). Diagn Microbiol Infect Dis.
reduces blood glucose in high-fat diet-fed 2004;50(1):59-69. DOI:
mice. Int J Mol Sci. 2017;18(1):168. 10.1016/j.diagmicrobio.2004.05.003, PMID
DOI: 10.3390/ijms18010168, PMID 15380279.
28106738. 31. Gebre-Sellassie S. Antimicrobial
23. Ho SC, Chang KS, Lin CC. Anti- resistance patterns of clinical bacterial
neuroinflammatory capacity of fresh ginger isolates in southern Ethiopia. Ethiop Med
is attributed mainly to 10-gingerol. Food J. 2007;45(4):363-70.
Chem. 2013;141(3):3183-91. 32. Khan NA, Saba N, Abdus S, Ali AQ.
DOI: 10.1016/j.foodchem.2013.06.010, Incidence and antibiogram patterns of E.
PMID 23871076. coli isolates from various clinical samples
24. Townsend EA, Siviski ME, Zhang Y, Xu C, from patients at NIH Islamabad. Pak J Biol
Hoonjan B, Emala CWE. _effect of ginger Sci. 2002;1:111-3.
and its constituents on airway smooth 33. Turner SM, Scott-Tucker A, Cooper LM,
muscle relaxation and calcium regulation. Henderson IR. Weapons of mass
Am J Resp. Mol: Cell Press. 2013;48:157- destruction: virulence factors of the global
63. killer enterotoxigenic Escherichia coli.
25. Akinyemi AJ, Thome GR, Morsch VM, FEMS Microbiol Lett. 2006;263(1):
Stefanello N, Goularte JF, Belló-Klein A et 10-20.

29
Momoh and Olaleye; MRJI, 32(7): 7-31, 2022; Article no.MRJI.92461

DOI: 10.1111/j.1574-6968.2006.00401.x, Acinetobacter baumannii. Phytother Res.


PMID 16958845. 2010;24(12):1825-30.
34. Momoh JO, Manuwa AA, Bankole YO. DOI: 10.1002/ptr.3201, PMID 20564496.
Phytochemical screening, atomic 43. Singletary KW. Vanilla: potential health
absorption spectroscopy, GC-MS and benefits. Nutr Today. 2020;55(4):
antibacterial activities of turmeric 186-96.
(Curcuma longa L.) rhizome extracts. Adv DOI: 10.1097/NT.0000000000000412.
Microbiol. 2022;22(9):116-31. Article no. 44. Belachew GT, Tekelemariam BA,
JAMB.88973. Hanumanthaiah P, Namo FM. Therapeutic
DOI: 10.9734/jamb/2022/v22i930498. Value of 6-gingerol (1-[4′-hydroxy-3′-
35. Momoh JO, Damazio OA, Ajetunmobi AO, methoxyphenyl]-5-hydroxy-3-decanone): A
Babalola AO, Adekunle OM, Busari NO, review. JPRI. 2021;33(9):63-75:Article
Musa AA. Phytochemical analysis and no.JPRI.66011.
antiplasmodial (curative) activities of DOI: 10.9734/jpri/2021/v33i931225.
methanolic leaf extract of Morinda lucida 45. Chen CY, Cheng KC, Chang AY, Lin YT,
(Ewe Oruwo) in male Swiss mice infected Hseu YC, Wang HM. 10-Shogaol, an
with Plasmodium berghei NK65. IJTDH. antioxidant from Zingiber officinale for
2019;37(1):1-13:Article no.IJTDH.47956. SkinCell proliferation and migration
DOI: 10.9734/IJTDH/2019/v37i130156. enhancer. Int J Mol Sci. 2012;13(2):1762-
36. Momoh J, Oluremi NO, Odetunde SK. 77.
Antimicrobial and antioxidant properties of DOI: 10.3390/ijms13021762, PMID
aqueous garlic (Allium sativum) extract 22408422.
against Staphylococcus aureus and 46. Aboaba O, Efuwape BM. Antibacterial
Pseudomonas aeruginosa. Br Microbiol properties of some Nigerian species. Bio
Res J: 101608140. 2016;14(1):1-11:Article Res Comm. 2001;13:183-8.
no.BMRJ.24095 ISSN: 2231-0886. 47. Awan UA, Ali S, Shahnawaz AM, Shafique
37. Trease GE, Evans WC. Pharmacognsy. I, Zafar A, Khan MAR et al. Biological
th
11 Ed. London: Brailliar Tiridel Can activities of Allium sativum and Zingiber
Macmillian Publishers. 1986;60-75. officinale extracts on clinically important
38. Momoh JO, Aderele OR, Rasaq AK. Sub- bacterial pathogens, their phytochemical
acute and protective effect of Cymbopogon and FT-IR spectroscopic analysis. Pak J
citratus against carbon tetrachloride- Pharm Sci. 2017;30(3):729-45. PMID
induced liver damage. Afr J Biochem Res. 28653916.
2020;14(4):112-24. 48. Moon Y, Lee H, Lee S. Inhibitory effects of
DOI: 10.5897/AJBR2019.1064. three monoterpenes from ginger essential
39. Moo-Key K, Young-Mi K, Hoi-Seon K. oil on growth and aflatoxin production of
Growth-inhibiting effects of Juniperus Aspergillus flavus and their gene regulation
virginiana leaf-extracted components in aflatoxin biosynthesis. Appl Biol Chem.
towards human intestinal bacteria. Food 2018;61(2):243-50.
Sci Biotechnol. 2005;14(1):164-7. DOI: 10.1007/s13765-018-0352-x.
40. Momoh JO, Manuwa AA, Oshin TT. 49. Nassan MA, Mohamed EH.
Phytochemical screening, gas Immunopathological and antimicrobial
chromatography: mass spectrometry and effect of black pepper, ginger and thyme
antidiabetic properties of aqueous extract extracts on experimental model of acute
of ginger (Zingiber officinale) in alloxan: hematogenous pyelonephritis in albino
induced diabetic Wistar rats. J rats. Int J Immunopathol Pharmacol.
Pharmacogn Phytochem. 2022;11(5):11-9. 2014;27(4):531-41.
DOI: 10.22271/phyto.2022.v11.i5a.14488. DOI: 10.1177/039463201402700409,
41. Park M, Bae J, Lee DS. Antibacterial PMID 25572733.
activity of (10). Phytother Res. 50. Yamamoto-Ribeiro MM, Grespan R,
2008;22(11):1446-9. Kohiyama CY, Ferreira FD, Mossini SA,
DOI: 10.1002/ptr.2473, PMID 18814211. Silva EL et al.. Effect of Zingiber officinale
42. Wang HM, Chen CY, Chen HA, Huang essential oil on Fusarium verticillioides and
WC, Lin WR, Chen TC, et al. Zingiber fumonisin production. Food Chem.
officinale (ginger) com-pounds have 2013;141(3):3147-52.
tetracycline-resistance modifying effects DOI: 10.1016/j.foodchem.2013.05.144,
against clinical extensively drug-resistant PMID 23871071.

30
Momoh and Olaleye; MRJI, 32(7): 7-31, 2022; Article no.MRJI.92461

51. Chakotiya AS, Tanwar A, Narula A, garlic. J Pharmacogn Phytochem.


Sharma RK. Zingiber officinale: its 2013;2(3):143-8.
antibacterial activity on Pseudomonas 56. Samuel-Penu B, Baridakara SC. Anti-
aeruginosa and mode of action evaluated microbial activities of turmeric and ginger
by flow cytometry. Microb Pathog. on bacterial isolates of normal skin flora.
2017;107:254-60. JAMB. 2021;21(3):59-62. Article no.
DOI: 10.1016/j.micpath.2017.03.029, PMID JAMB.67256.
28389345. DOI: 10.9734/JAMB/2021/v21i330336.
52. Kim HS, Park HD. Ginger extract inhibits 57. Karuppiah P, Rajaram S. Antibacterial
biofilm formation by Pseudomonas effect of Allium sativum cloves and
aeruginosa PA14. PLOS ONE. Zingiber officinale rhizomes against
2013;8(9):e76106. multiple-drug resistant clinical pathogens.
DOI: 10.1371/journal.pone.0076106, PMID Asian Pac J Trop Biomed. 2012;2(8):597-
24086697. 601.
53. Hasan S, Danishuddin M, Khan AU. DOI: 10.1016/S2221-1691(12)60104-X,
Inhibitory effect of Zingiber officinale PMID 23569978.
towards Streptococcus mutans virulence 58. Njobdi S, Gambo M, Ishaku GA.
and caries development: In vitro and in Antibacterial activity of Zingiber officinale
vivo studies. BMC Microbiol. 2015; on Escherichia coli and Staphylococcus
15:1. aureus. JABB. 2018;19(1):1-8:Article
DOI: 10.1186/s12866-014-0320-5, PMID no.JABB.43534.
25591663. DOI: 10.9734/JABB/2018/43534.
54. Rampogu S, Baek A, Gajula RG, Zeb A, 59. Gull I, Saeed M, Shaukat H, Aslam SM,
Bavi RS, Kumar R et al. Ginger (Zingiber Samra ZQ, Athar AM. Inhibitory effect of
officinale) phytochemicals-gingerenone-A Allium sativum and Zingiber officinale
and shogaol inhibit SaHPPK: molecular extracts on clinically important drug
docking, molecular dynamics simulations resistant pathogenic bacteria. Ann Clin
and in vitro approaches. Ann Clin Microbiol Microbiol Antimicrob. 2012;11:8.
Antimicrob. 2018;17(1):16. DOI: 10.1186/1476-0711-11-8, PMID
DOI: 10.1186/s12941-018-0266-9, PMID 22540232.
29609660. 60. Kacmaz B, Sultan N. In vitro
55. Virenda VP, Shalini T, Nirmala K, Chetan susceptibilities of Escherichia coli and
N, Kalpagam P. in vitro evaluation on Klebsiella spp. to ampicillin-sulbactam and
antioxidant and antimicrobial activity of amoxicillin-clavulanic acid. Jpn J Infect Dis.
spice extracts of ginger, turmeric and 2007;60(4):227-9. PMID 17642541.
_________________________________________________________________________________
© 2022 Momoh and Olaleye; This is an Open Access article distributed under the terms of the Creative Commons Attribution
License (http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any
medium, provided the original work is properly cited.

Peer-review history:
The peer review history for this paper can be accessed here:
https://www.sdiarticle5.com/review-history/92461

31

View publication stats

You might also like