Download as pdf or txt
Download as pdf or txt
You are on page 1of 33

EFFECT OF ETHANOLIC SEED EXTRACT OF CAESALPINIA

BONDUCELLA ON MIFEPRISTONE INDUCED PCOS RATS


PROJECT REPORT

Submitted to RIMT University in partial fulfilment of reward of degree for

BACHELOR OF PHARMACY

NAME OF THE SUPERVISIOR NAME OF CO-SUPERVISOR


MS. HURMANDEEP KAUR MR. RAJAT
ASSISTANT PROFESSOR ASSOCIATE PROFESSOR

ISHFAQ NAZIR
ROLL NO: 20-B-PHAR-135

COLLEGE OF PHARMACY
RIMT UNIVERSITY, MANDI GOBINDGARH, PUNJAB-
147301 (2020-2024)
CERTIFICATE

The project report entitled “Effect of ethanolic seed extract of Caesalpinia bonducella
on Mifepristone induced PCOS rats” submitted by Ishfaq Nazir (20- B- PHAR-135)
of B-Pharmacy, 8th Semester as a requirement for award of Bachelor of Pharmacy
degree is in accordance to the instructions laid down in course Regulations, 2014 framed
under section 6,7,8 of the Bachelor of Pharmacy as per Pharmacy Council of India, New
Delhi.

Prof. (Dr.) Mohit Mehta Prof. (Dr.) Parminder Nain

Head of Department Dean & Principal

College of Pharmacy College of Pharmacy

RIMT University, RIMT University,

Mandi-Gobindgarh (Punjab) Mandi-Gobindgarh (Punjab)


CERTIFICATE

The research work in project report entitled “Effect of ethanolic seed extract of

Caesalpinia bonducella on Mifepristone induced PCOS rats” submitted by “Ishfaq

Nazir (20-B-PHAR-135)” of B- Pharmacy 8th Semester as a requirement for award of

Bachelor of Pharmacy degree is conducted under my supervision at College of

Pharmacy, RIMT University, Mandi – Gobindgarh (Punjab) To best of our

knowledge, the work embodied in this project report is an original piece of work and is

in accordance to instructions laid down in course Regulation, 2014 framed under section

6, 7 & 8 of the Bachelor of Pharmacy as per Pharmacy Council of India, New Delhi

Supervisor Co-Supervisor

Ms. Hurmandeep Kaur Mr. Rajat

Assistant Professor Associate Professor


College of pharmacy College of pharmacy
RIMT University RIMT University
Mandi Gobindgarh (Punjab) Mandi Gobindgarh (Punjab)
DECLARATION BY CANDIDATE

Ishfaq Nazir (20-B-PHAR-135) of B-Pharmacy 8th semester of College of Pharmacy


hereby declare that the work presented in project report entitled “Effect of ethanolic
seed extract of Caesalpinia bonducella on Mifepristone induced PCOS rats” is
original work conducted by him/her at College of Pharmacy, RIMT University,
Mandi – Gobindgarh (Punjab) as partial fulfillment for award of Bachelor of
Pharmacy. This work has not been submitted earlier in part or full for the award of any
degree.

Ishfaq Nazir
(20-B-PHAR-135)
Dedicated to God & everyone

who supported me during this

journey.
ACKNOWLEDGEMENT

I am extremely grateful to The Almighty God, the most gracious, the most
merciful for giving me the strength, knowledge and opportunity to complete this
study with zeal and enthusiasm. I am indebted to you, The Almighty God.

I would like to express my deepest gratitude and immeasurable appreciation to my


guide Ms. Hurmandeep Kaur for her patience, immense knowledge, worthy
guidance, professional attitude and appreciable criticism that improved this study.
This endeavour would not have been possible without you.

All that I am, or hope to be, I owe it to my parents at times, our own light
goes out and is rekindled by the spark from someone's continuous support. These
words are not enough to express my gratitude to my respected father Mr. Nazir
Ahmad and loving mother Ms. Halima. Thank you for showering me with your
love and blessing. I will forever be grateful. I could not have taken this journey
without the immense support from all the professor of department Bachelor
of Pharmacy, (COP) RIMT University. I was able to accomplish my career goal,
all thanks to you.
I would like to express my sincere gratitude to my fellow friend and everyone who
volunteered to participate in this study. Thank you so much for keeping me in high
spirit, motivated, helping to overcome every barrier. Word cannot express my
gratitude to each one of you.

Hereby, I idolize and acclaim The Almighty God who paved my path toward
success with enormous blessing.

Ishfaq Nazir

(20-B-PHAR-135)
INDEX

S.NO. TITLE PAGE NO.

1. Abstract 1

2. Aims and Objective 2

3. Introduction 3-4

4. Review of Literature 5-12

5. Discussion 13-15

6. Conclusion 16

7. Future Directions 16

8. Bibliography 17-23
LIST OF TABLES

S.NO. TITLE PAGE NO.

1. Function of microRNAs observed in polycystic ovaries 8


LIST OF FIGURES

S.NO. TITLE PAGE NO.

1 Etiology of PCOS 5

2 Effect of ESECB on LH, FSH, insulin, and PRL levels in female 11


rats
3 Effect of ESECB on testosterone and progesterone levels in 12
female rats
4 Effect of ESECB on estrone and estradiol levels in female rats 12
ABSTRACT

1. ABSTRACT:

Polycystic ovarian syndrome is a major concern for today’s young generation because
of unhealthy lifestyle. PCOS is hormone related chronic condition which causes
anovulation leading to infertility. It is said to affect the physical and emotional
wellbeing of the person who is suffering from it. The approximate percentage of
patients suffering from pcos is around 9-14% many of which are undiagnosed. As such
the cause of PCOS is unknown but it is said that women with family history and type
2 diabetes associated with hormonal imbalance, irregular periods, excess androgen
levels and cysts in ovaries. The current study aims to assess the impact of Caesalpinia
bonduella (ESECB) ethanolic seed extract on altered hormone levels in female rats
with PCOS caused by mifepristone. Mifepristone (4 mg/kg b.w.) was administered
daily to female rats for 8 days, causing PCOS, which was verified by the continuous
estrous cycle. After the rats were given ESECB 200 and 400 mg/kg b.w. p.o. for 28
days after PCOS was developed, the hormonal parameters were examined. One
common medication utilized was metformin. In PCOS rats, there was a notable rise (p
< 0.01) in the levels of luteinizing hormone, prolactin, insulin, testosterone, estrone, and
estradiol, and a fall in the levels of follicle stimulating hormone and progesterone
hormone, which returned to normal in the animals treated with ESECB medication.
Out of the two dosages, 400 mg/kg b.w. of ESECB medication therapy had a notable
impact in reversing this hormonal imbalance. Its impact was similar to that of the
study's positive control.

Keywords: Caesalpinia bonducella; ESECB; PCOS; Mifepristone

COLLEGE OF PHARMACY, RIMT UNIVERSITY, MANDI GOBINDGARH; PUNJAB-147301


Page 1
AIM & OBJECTIVES

2. AIM & OBJECTIVES:

Aim: The aim of this study is to investigate the effect of ethanolic seed extract of
Caesalpinia bonducella on mifepristone-induced polycystic ovary syndrome
(PCOS) in rats.

Objectives:
➢ To induce PCOS in experimental rats using mifepristone.
➢ To administer ethanolic seed extract of Caesalpinia bonducella to PCOS-
induced rats.
➢ To evaluate the impact of Caesalpinia bonducella extract on hormonal
imbalances associated with PCOS, including alterations in estrous cycle and
serum hormone levels (such as testosterone, luteinizing hormone, follicle-
stimulating hormone, and insulin).
➢ To assess the histological changes in ovarian morphology and follicular
development following treatment with Caesalpinia bonducella extract.
➢ To investigate the potential mechanisms underlying the therapeutic effects of
Caesalpinia bonducella extract on PCOS, including antioxidant and anti-
inflammatory properties.
➢ To compare the observed effects of Caesalpinia bonducella extract with standard
treatments for PCOS, such as metformin or other herbal remedies.
➢ To draw conclusions regarding the efficacy of Caesalpinia bonducella extract as
a potential therapeutic agent for PCOS and provide insights for future research
directions

COLLEGE OF PHARMACY, RIMT UNIVERSITY, MANDI GOBINDGARH; PUNJAB- 147301


Page 2
INTRODUCTION

3. INTRODUCTION:

PCOS, or polycystic ovarian syndrome, is regarded as a complex illness that affects women
in reproductive age groups and is primarily responsible for decreased fertility due to
hormonal and metabolic issues [Teede H et al., 2010]. Anovulation, multiple antral follicles,
hyperandrogenemia, hirsutism, oligomenorrhea, and amenorrhea are among the
pathophysiological symptoms [Demirel M et al., 2016]. Metabolic problems include
obesity, dyslipidemia, and insulin resistance [Li et al.,2013]. Despite its unclear origin,
PCOS may be treated in a variety of ways, making it a significant study issue for the
scientific community. The primary areas of interest for this research include lipid
imbalance, oxidative stress, insulin resistance, and genetics. The primary focus of PCOS
treatment currently available is ovulation induction through fertility-enhancing measures.
An alternate treatment method is required since these medications will cause serious side
effects such psychological disorders, joint or muscle discomfort, and arthritis [Bency et al.,
2016]. Based on a combination of scientific and clinical approaches, there is growing
evidence that genetic and environmental variables contribute to the etiology of PCOS, even
if the underlying causes of the illness remain unknown [Liu J et al, 2021]. Continuous
exposure to environmental contaminants has been extensively studied in relation to the
pathophysiology of PCOS, both in terms of its causation and aggravation [Teede HJ et al.,
2018]. The role that antioxidants play in PCOS treatment has been related to oxidative
stress, which is related to the pathogenesis of the illness [Trent M et al., 2020]. In PCOS-
affected women, antioxidants may improve glycemic control and ovulation rate. Because
natural antioxidants and phytochemicals may counteract the effects of free radicals, they are
becoming more and more well-liked for the treatment of oxidative disorders linked to stress
[Bannigida DM et al., 2020; Ogunlade B et al., 2021]. Aerobic metabolism inside cells and
the presence of contaminants continuously produce reactive oxygen species (ROS)
[Adelakun et al., 2021; Oyeniram et al., 2021; Ukwenya VO et al., 2021].
A multi-therapeutic strategy is needed to treat PCOS, and phytotherapy—which contains a
large number of phytochemicals and has the benefit of little to no adverse effects—is
currently receiving attention on a global scale.

COLLEGE OF PHARMACY, RIMT UNIVERSITY, MANDI GOBINDGARH; PUNJAB-147301


PAGE 3
INTRODUCTION

The plant Caesalpinia bonducella is a member of the Caesalpiniaceae family, which is also
known as "Kazharchikai" in Tamil. It is widespread across India, particularly in the
country's tropical areas [Asolkar et al., 1992].
The firm, glossy seed is greenish to ash gray in color and is referred known as a "bonduc
nut" or "fever nut" [Ca-Ci et al., 1992]. The seeds have a variety of medicinal uses, including
immunomodulatory, antioxidant, anti-inflammatory, antipyretic, analgesic, hypoglycemic,
and hypo-lipidemic [Shukla et al., 2009; Mehta et al., 2010]. The powder made from leaves
is utilized to treat gynecological issues such as leucorrhoea and menorrhagia. Despite
having a number of medical benefits, the seed's ability to cure PCOS has not been clinically
proven. In order to determine if ethanolic seed extract of Caesalpinia bonducella (ESECB)
may alleviate PCOS in female rats that has been produced by mifepristone, the current study
was conducted [Mantzors C et al., 1995].

COLLEGE OF PHARMACY, RIMT UNIVERSITY, MANDI GOBINDGARH; PUNJAB-147301


PAGE 4
REVIEW OF LITERATURE

4. REVIEW OF LITERATURE

Etiology and pathophysiology of PCOS

The complex multifactorial condition known as PCOS is impacted by a person's lifestyle


choices as well as environmental and hereditary factors. Owing to its varied clinical
presentations, the cause of this illness remains unclear. Because of this, the disease's many
side effects continue to provide therapeutic challenges [Xu Y et al., 2018]. Intrinsic ovarian
dysfunction, which is significantly influenced by exogenous variables such
hyperinsulinemia and abnormalities of the hypothalamic-pituitary-ovarian axis, is part of
the pathogenesis of this illness. Wherein the emergence of the syndrome's features appears
to be caused by many genetic defects [Archard et al., 1921]. Generally speaking, the
development of polycystic ovaries occurs when they are driven to generate excessive levels
of androgens [Li M et al., 2022]. Androgen overproduction is linked to inflammation and
genes that are resistant to insulin (Fig. 1).

Fig. 1. The three main etiology of PCOS are (A) insulin resistance and hyperinsulinemia,
(B) heredity and genetic factors, and (C) inflammation. CRP: C-reactive protein; FSH:
follicle-stimulating hormone; IL: interleukin; LH: luteinizing hormone; PCOS: polycystic
ovary syndrome; TNF: tumor necrosis factor.

COLLEGE OF PHARMACY, RIMT UNIVERSITY, MANDI GOBINDGARH; PUNJAB-147301


PAGE 5
REVIEW OF LITERATURE

Insulin resistance and hyperinsulinemia

A condition known as insulin resistance occurs when the body needs more insulin than usual
to produce a typical reaction [Shen HR et al., 2013]. Pancreatic B cells release insulin, which
after attaching to its receptor, activates the PI3K/PKB and MAPK/Ras signaling pathways,
hence controlling metabolism. Consequently, Peripheral tissues become less sensitive to
insulin due to interference with insulin receptors and signaling pathways, which disrupts how
these tissues utilize glucose. Consequently, blood insulin levels rise as a result of the body's
compensatory insulin production [Wang T et al., 2013]. Achard and Thiers published the
first study on the connection between insulin metabolism and hyperandrogenism in 1921.
Additional details on insulin metabolism in PCOS patients were revealed in a follow-up
study published in 1976 [De Leo V et al., 2003]. In between 70% and 50% of PCOS patients,
hyperandrogenism and insulin resistance are present [Dankner R et al., 2009]. In PCOS
patients, the control of insulin resistance is influenced by DNA methylation, histone status,
and microRNA (miRNA) expression. Between those who are ill and those who are not,
there are also notable differences in the expression levels of miRNAs. Follicle development,
ovulation, and the normal physiological function of the ovary are all impacted by insulin
resistance, which is followed by aberrant glucose metabolism in PCOS patients.
Furthermore, by the additive action of cytochrome P450C17a, elevated insulin levels
significantly contribute to increased androgen release in theca cells [Hiam D et al., 2019].
Increased secretion of male androgen hormones in women is linked to the clinical signs of
PCOS associated with hyperinsulinemia. By stimulating luteinizing hormone (LH)
secretion by ovarian theca cells and inhibiting the synthesis and secretion of insulin-like
growth factor-binding protein (IGFBP) and sex hormone-binding globulin (SHBG),
hyperinsulinemia may either directly or indirectly stimulate the ovaries, leading to an
increase in the production of androgens [Goldrat A et al., 2018] (Fig. 1A). Metabolic
syndrome, another name for hyperinsulinemia, is linked to the emergence of obesity,
dyslipidemia, and hypertension. Due to an increase in renal salt retention, hyperinsulinemia
has been linked recently to the development of hypertension. High plasma triglycerides, low-
density lipoprotein (LDL), and adapted low-density lipoproteins are indicative of
dyslipidemia [Balen A et al., 2004].

COLLEGE OF PHARMACY, RIMT UNIVERSITY, MANDI GOBINDGARH; PUNJAB-147301


PAGE 6
REVIEW OF LITERATURE

Heredity and genetic factors

It is established where the pathogenesis of PCOS originated and what aspects are inherited.
These results led to the identification of 19 PCOS risk gene loci in the neuroendocrine,
reproductive, and metabolic pathways [Strauss et al., 2003]. Future research is necessary to
confirm the high potential of a number of loci, such as DENN domain-containing 1A
(DENND1A), follicle- stimulating hormone receptor (FSHR), thyroid adenoma-associated
gene (THADA), and insulin variable number of tandem repeats (INS-VNTR) [Day F et al.,
2018]. Since less than 10% of the heritability of PCOS is accounted for by the discovered
genetic risk alleles, other etiological variables should undoubtedly be taken into account in
addition to inheritance [Hoeger et al., 2021]. Certain genes that are expressed more often in
PCOS-affected women produce more testosterone. Gonadotropin- releasing hormone
(GnRH) production rises in PCOS-affected women, which raises LH output relative to FSH.
Theca cells produce testosterone by gradually upregulating the expression of steroidogenic
acute regulatory protein (StAR), P450 side-chain cleavage (P450scc), 3β- hydroxysteroid
dehydrogenase (3β-HSD), and cytochrome P450c17 (CYP17) in response to regulating the
rise in LH production. Then, in the granulosa cells, androstenedione is transformed into
estrogen by aromatase while being influenced by FSH. But there is a relative lack of FSH
production, which frequently lowers the activity of aromatase and hinders the formation of
follicles (Fig. 1B). Consequently, excessive androgen buildup takes place, which causes
PCOS women to become hyperandrogenic [Saddick et al., 2020]. There is a lot of evidence
to imply that the development of PCOS involves the miRNA axis. miRNAs are single-
stranded, 20–24 nucleotide short non-coding RNAs (ncRNAs) that control other RNA.
They are present in PCOS patients' serum, plasma, and follicular fluid (FF). Diabetes
mellitus, insulin sensitivity, inflammation, and several malignancies have all been linked to
altered miRNA levels. According to recent research, ncRNAs have a role in the onset of
PCOS. Highly functional RNAs known as ncRNAs regulate a significant number of
biological processes and can be exploited as therapeutic markers or diagnostic indicators in
PCOS patients [Mu L et al., 2021]. Table 1 provides an overview of the miRNAs involved
in PCOS.

COLLEGE OF PHARMACY, RIMT UNIVERSITY, MANDI GOBINDGARH; PUNJAB-147301


PAGE 7
REVIEW OF LITERATURE
TABLE 1:

Function of microRNAs observed in polycystic ovaries

MicroRNA Reported Function(s) References

miR-9 Inhibits testosterone release and increases PCN [Roth et al.,


expression 2014]
miR-30c Increased expression after FSH exposure [Long et al.,
2014]
miR-93 Inhibits SIRT1 and GLUT4 [Chen YH et al.,
2013
miR-146a Reduces progesterone, estradiol, and testosterone [Long et al.,
release 2014]
miR-21 Reduced in obesity and type 2 diabetes mellitus, [Diez-Fraile et
increased expression after FSH exposure; al., 2014]
inconclusive testosterone response

Low-grade inflammation

Small proteins called cytokines are produced by specific immune cell types and released by
ovarian follicular cells, leukocytes, and oocytes [Sirotkin AV et al., 2011]. Consequently,
the ovary's synthesis of these markers shows that they can have varying ovarian activity and
function in an autocrine or paracrine way. These include processes including
steroidogenesis, ovarian cell proliferation, corpus luteum function, and hormone balance
management. According to recent research, proinflammatory conditions like PCOS are
linked to a little elevation in inflammatory markers such C-reactive protein (CRP). Adipose
tissue has the ability to directly create CRP, and under some circumstances, it may also be
stimulated by TNF-α and interleukin-6 (IL-6). Furthermore, based on the documented
evidence, the impact of inflammatory mediators or indicators like IL-8, IL-18, IL-33, and
IL-10 has been demonstrated in PCOS patients. Patients with PCOS showed a decline in
plasma IL-10 levels (Fig. 1C). Insulin resistance, obesity, diabetes mellitus, and
cardiovascular disease are metabolic features of polycystic ovary syndrome (PCOS) that
are linked to inflammation [Spritzer PM et al., 2022; Abraham S et al., 2021].

COLLEGE OF PHARMACY, RIMT UNIVERSITY, MANDI GOBINDGARH, PUNJAB-147301


PAGE 8
REVIEW OF LITERATURE

Induction of PCOS by Mifepristone in female rats

Antiprogesterone medication Mifepristone RU486 (4 mg/0.2 ml oil) was given to four- day
estrous cyclic rats over the course of eight consecutive days, beginning on the day of estrus
(day 1) [Sanchez JE et al., 1993]. A vaginal smear test and a Mifepristone RU486 injection
were administered after the erratic estrous cycle each day was used to watch and track the
development of PCOS in the rats. Animal grouping was done for subsequent treatment once
anovulatory cystic ovary condition, similar to those demonstrated in polycystic ovarian
disease, was proven by prolonged vaginal cornification stage.

Animal grouping and treatment pattern

For the analysis, thirty mature female Wistar rats were collected and divided into five
groups of six animals each [Meera B et al., 2020].

Group I: This group was given merely the vehicle (2% carboxymethyl cellulose (CMC)
suspension) as a standard control.

Group II was used as a negative control and was given just Mifepristone RU486. Group III:
For 28 days, PCOD-induced rats were given ESECB 200 mg/kg b.w.

Group IV: PCOD-induced rats received a 28-day treatment with ESECB 400 mg/kg b.w.
Group V: Metformin 20 mg/kg b.w. was administered for 28 days as a benchmark.

Blood sample

Following the administration of ESECB to every animal, they were all slaughtered while
under a light anesthetic. Blood samples were obtained using retro-orbital puncture and
placed in sterile eppendorff tubes. The serum was separated and utilized for the hormone
test by centrifugation at 3,000 rpm for 10 minutes [Meera B et al., 2020].

COLLEGE OF PHARMACY, RIMT UNIVERSITY, MANDI GOBINDGARH, PUNJAB-147301


PAGE 9
REVIEW OF LITERATURE

Estimation of hormonal assay

Enzyme-Linked Immuno Sorbent Assay (ELISA) technique was used to evaluate the levels
of hormones, including LH, FSH, prolactin (PRL), testosterone (T), estrone, estradiol,
progesterone, and insulin, using commercial diagnostic kits (TOSOH LILAC India Pvt
Ltd).

Statistical analysis

Mean ± SE data are presented. Dunnet's "t" test was used in conjunction with analysis of
variance (ANOVA) to conduct the statistical study. P values less than 0.05 were deemed
significant [Meera B et al., 2020].

RESULTS
Effect of ESECB on LH, FSH, insulin, and PRL levels

Figure 2 depicted the impact of the ESECB extract on many hormones, including insulin,
PRL, FSH, and LH. Comparable (p < 0.01) to the control rats, it is seen that the PCOS-
induced group II rats had significantly higher levels of LH and lower levels of FSH,
indicating the presence of an irregular ovulatory process. The ESECB medication treatment
for Group III and Group IV of mifepristone-induced PCOS rats resulted in a drop in LH and
an increase in FSH, indicating the restoration of ovulation.

Furthermore, compared to control rats, the PCOS-induced Group II rats exhibit a


statistically significant rise in insulin and PRL levels, suggesting the existence of
hyperinsulinemia, a key characteristic of PCOS. In the PCOS-induced and ESECB drug
treated group III and group IV mice, these levels were markedly restored. Insulin levels
significantly decreased in the rats given 400 mg/kg b.w. of ESECB extract; this decline was
more pronounced than it was in the positive control. The current study's usage of metformin
suggests that the medication extract may have the ability to increase insulin sensitivity
[Meera B et., al 2020]

COLLEGE OF PHARMACY, RIMT UNIVERSITY, MANDI GOBINDGARH, PUNJAB-147301


PAGE 10
REVIEW OF LITERATURE

Figure 2 :- Effect of ESECB on LH, FSH, insulin, and PRL levels in female rats. Values are
mean ± SEM of six animals in each group. Statistical significant test for comparison was
done by ANOVA followed by Dunnet’s “t” test. Comparison between a—Group I versus
Group II, b—Group II versus Group III, c— Group II versus Group IV, and d— Group II
versus Group V. p values—*p < 0.05, **p < 0.01, ***p < 0.001, NS–Not Significant.

Effect of ESECB on testosterone, estrone, estradiol, and progesterone levels

Figures 2 and 3 demonstrate the impact of ESECB extract on several hormones, including
progesterone, testosterone, estrone, and estradiol. Based on the figures, it can be noted that
the group II PCOS-induced rats had significantly higher levels of testosterone, estrone, and
estradiol (p < 0.01) than the normal control group. Additionally, it was noted that the group
III and group IV rats treated with ESECB had a considerable reversion of the values [Meera
B et al., 2020].

COLLEGE OF PHARMACY, RIMT UNIVERSITY, MANDI GOBINDGARH, PUNJAB-147301


PAGE 11
REVIEW OF LITERATURE

Figure 3:- Effect of ESECB on testosterone and progesterone levels in female rats. Values
are mean ± SEM of six animals in each group. Statistical significant test for comparison
was done by ANOVA followed by Dunnet’s “t” test. Comparison between a—Group I
versus Group II, b—Group II versus Group III, c—Group II versus Group IV, and d—
Group II versus Group V. p values—*p < 0.05, **p < 0.01,***P < 0.001, NS–NOT
SIGNIFICANT.

Figure 4:- Effect of ESECB on estrone and estradiol levels in female rats. Values are mean
± SEM of six animals in each group. Statistical significant test for comparison was done by
ANOVA followed by Dunnet’s “t” test. Comparison between a—Group I versus Group II,
b—Group II versus Group III, c—Group II versus Group IV, and d—Group II versus Group
V. p values—*p < 0.05, **P < 0.01, ***P < 0.001, NS–NOT SIGNIFICANT.

COLLEGE OF PHARMACY, RIMT UNIVERSITY, MANDI GOBINDGARH, PUNJAB-147301


PAGE 12
DISCUSSION

Based on the figure, it can be noted that the group II PCOS-induced rats had significantly
higher levels of testosterone, estrone, and estradiol (p < 0.01) than the normal control group.
The ESECB medication therapy considerably restored these reproductive hormone levels
in the rats in Group III and Group IV. In the current investigation, mifepristone therapy
resulted in a lowered progesterone level, which was then raised by ESECB medication
treatment, suggesting that drug extract may be able to reverse the antiprogesterone effects
of mifepristone [Meera B et al., 2020].

5. DISCUSSION

The hypothalamic, pituitary, and ovarian axis are all involved in the multiglandular
pathophysiology of PCOS, an endocrine disorder that causes disruptions in the cycle of
reproduction and metabolic complications like obesity, dyslipidemia, hyperinsulinemia, and
insulin resistance [Teede H et al., 2006]. An antagonistic synthetic steroid called RU486 has
a strong affinity for progesterone and glucocorticoid receptors, which inhibits progesterone
action and disrupts endocrine processes, as found in PCOS [Baulieu EE et al., 1991]. Adult
cycling female rats treated with RU486 for 4–9 days develop PCOS, anovulation, and
acclivity with high blood levels of T, E, and LH [Teede H et al., 2018]. The goal of the
current study is to examine how ESECB affects hormonal markers in PCOS- induced rats
using RU486 (mifepristone).

It is generally acknowledged that anovulation disrupts the feedback signaling from ovarian
sex hormones to the pituitary and brain, which influences the release of gonadotropin-
releasing hormone (GnRH) and, consequently, interferes with the normal production of LH
and FSH. The LH/FSH ratio increased in the current study, and a ratio larger than two is
regarded as the "gold standard for evaluating the gonadotropin status in PCOS’’ [Teede H
et al., 2010]. It is also revealed that elevated LH levels might be caused by a compromised
estrogenic feedback loop, which in turn drives the aberrant proliferation of theca cells and
ultimately results in PCOS. By greatly lowering LH levels, the ESECB extract effectively
corrected the LH/FSH ratio, shielding the ovaries from the theca cells' excessive growth and
aiding in the prevention of PCOS [Feng et al., 2013].

COLLEGE OF PHARMACY, RIMT UNIVERSITY, MANDI GOBINDGARH, PUNJAB-147301


PAGE 13
DISCUSSION

The PRL level in the PCOS rats increased noticeably in the current study, and ESECB
assisted in reducing the PRL levels. The observed hyperprolactinemia might be caused by
changes in the pulsatile release of GnRH, which alters the normal LH/FSH ratio, or by
increased levels of estrogen and testosterone. Anovulatory response and hypoestrogonism
are caused by PRL's inhibition of granulosa cell aromatase activity and blocking of
folliculogenesis in the ovary [Kulshreshtha B et al., 2017]. Many phytochemicals, including
bonducin, proteins, saponin, starch, sucrose, two phytosterols, namely sitosterol and
hepatsane, and fatty acids, including palmitic, stearic, lognoceric, oleic, and linolenic acid,
which are present in C. bonduella seeds, are used to correct the impaired metabolic functions
and reduce body weight and reproductive organs [Gurunath HM et al., 2010]. Of the several
androgens, testosterone is thought to be the most significant. In normal women, testosterone
is produced from both the ovaries and the adrenal glands. It is asserted that in the case of
PCOS-affected women, the ovaries are the source of all testosterone [Marshall K et al.,
2001]. Furthermore, reduced levels of sex hormone binding globulin (SHBG) have been
linked to an increase in the concentration of unbound androstenedione and testosterone,
which causes hyperandrogonism, Because hyperandrogenism modifies the metabolism of
glucose, it causes hyperinsulinemia [Cibula D et al., 2002]. There is a complicated
reciprocal relationship between hyperandrogenemia and insulin sensitivity. In the presence
of elevated androgens, the peripheral and hepatic insulin action is impeded, which in turn
leads to insulin resistance [Gambineri et al., 2002]. The rise in insulin levels, which is
another significant marker in the pathophysiology of PCOS because insulin inhibits the
production of SHBG in the hepatic cells, is the cause of the lower amounts of SHBG
[Wallace IR et al., 2013]. In the current study, we found that there were higher than of the
hormones insulin and testosterone in the mifepristone- induced.

PCOS rats that may have contributed to a substantial rise in the insulin resistance in contrast
to normal control rats. This may be explained by the possibility that either there is a flaw in
the insulin, attaching to the receptors, or it might be because to a drop in the quantity of
insulin receptors, which results from a reduction in the In PCOS women, insulin sensitivity
for glucose transport stimulation [Ciaraldi T et al., 1992]. Insulin exerts direct and indirect
functions in hyperandragonism in the pathophysiology of PCOS.

COLLEGE OF PHARMACY, RIMT UNIVERSITY, MANDI GOBINDGARH, PUNJAB-147301


PAGE 14
DISCUSSION

Ovarian tissues become hyperactive to insulin despite other tissues being resistant to it,
leading to enhanced steroidogenesis, particularly androgen production. Furthermore, insulin
and LH increase the production of androgens by theca cells, which results in persistent
hyperandrogenism and exacerbates PCOS problems [Baillargeon et al., 2006; Diamanti et
al., 2008]. Insulin and testosterone levels were significantly lowered by oral treatment of
ESECB extract, suggesting that insulin resistance has a suppressive impact. Previous
researches demonstrated that the hydro alcoholic extract has the ability to lower blood
glucose levels [Nadaf R et al., 2017; Gosh et al., 2012]. The phytoconstituents present in the
C. bonducella considerably reduces the insulin resistance [Shukla S et al., 2011]. The blood
progesterone level in the PCOS rats was found to be low, which is an indication of
anovulation, because the PCOS inducer utilized in this work is an antiprogestin [Srivastava
RK et al., 2006].

COLLEGE OF PHARMACY, RIMT UNIVERSITY, MANDI GOBINDGARH, PUNJAB-147301


PAGE 15
CONCLUSION

6. CONCLUSION

It is determined that anovulation, which is the primary cause of the pathophysiology in


PCOS instances, may be brought on by endocrine-based metabolic variables such as
hyperinsulinemia, insulin resistance, and hyperandrogonism. Because it contains a variety
of phytochemicals, ESECB may be able to help with a number of PCOS issues, including
hyperinsulinemia, insulin resistance, and hyperandrogenism. By encouraging ovulation, it
may also be a useful therapeutic option.

7. FUTURE DIRECTIONS

1. Clinical Translation: Transition findings to clinical trials in humans to validate


efficacy and safety.

2. Mechanistic Insights: Explore molecular mechanisms underlying the extract's


effects on PCOS.

3. Dose Optimization: Further optimize dosage and formulation for enhanced


therapeutic outcomes.

4. Long-term Effects: Investigate prolonged treatment effects and safety profile over
extended periods.

5. Combination Therapy: Assess synergistic effects with other PCOS treatments for
improved outcomes.

6. Secondary Metabolites: Identify specific compounds responsible for therapeutic


effects to refine treatments.

7. Clinical Biomarkers: Validate biomarkers for predicting treatment response and


personalized care.

COLLEGE OF PHARMACY, RIMT UNIVERSITY, MANDI GOBINDGARH, PUNJAB-147301


PAGE 16
BIBLIOGRAPHY

8. BIBLIOGRAPHY

➢ Abraham Gnanadass S, Divakar Prabhu Y, Valsala Gopalakrishnan A. Association of


metabolic and inflammatory markers with polycystic ovarian syndrome (PCOS): an
update. Arch GynecolObstet 2021;303(3):631–643. doi:10.1007/s00404-020-05951-2.

➢ Achard C, Thiers J. Le virilisme pilaire et son association a l’insuffisance glycolytique


(diabete des femmes a barbe). Bull Acad Natl Med 1921;86(29):51– 66.

➢ Adelakun SA, Ogunlade B, Iteire KA, Adedotun OA. Ameliorating potential and Cysteine
in cisplatin-induced oligo astheno teratozoospermia and seminiferous epithelium
degeneration in adult male Sprague-Dawley rats. Metabolism Open2021;12:100128.

➢ Asolkar, L. V., Kakkar, K. K. and Chakre, O. J., To glossary of Indian medicinal plants
with active principles, Second Suppl. Part 1. New Delhi: PID- CSIR; 1992.

➢ Baillargeon J-P, Nestler JE. Polycystic ovary syndrome: a syndrome of ovarian


hypersensitivity to insulin? J Clin Endocrinol Metab, 2006; 91:22–4.

➢ Balen A. The pathophysiology of polycystic ovary syndrome: trying to understand PCOS


and its endocrinology. Best Pract Res Clin Obstet Gynaecol 2004;18(5):685–706.
doi:10.1016/j.bpobgyn.2004.05.004.

➢ Bannigida DM, Nayak BS, Vijayaraghavan R. Insulin resistance and oxidative marker in
women with PCOS. Arch PhysiolBiochem 2020;126(2):183–6.

➢ Baulieu EE. The antisteroid RU486: its cellular and molecular mode of action. Trends
Endocrinol Metab, 1991; 2:233–9.

➢ Bency Baby, T., Smitha Rani, Remya, K., Shebina, P.R., and Azeem, A. K., Polycystic
ovarian syndrome: Therapeutic potential of herbal remedies-A review. Inter. J. herb .Med
.2016; 4: 91-96.

➢ Chen YH, Heneidi S, Lee JM, et al. miRNA-93 inhibits GLUT4 and is overexpressed in
adipose tissue of polycystic ovary syndrome patients and women with insulin resistance.

COLLEGE OF PHARMACY, RIMT UNIVERSITY, MANDI GOBINDGARH, PUNJAB-147301


PAGE 17
BIBLIOGRAPHY

Diabetes 2013;62(7):2278–2286. doi:10.2337/db12-0963.

➢ Ciaraldi T, El-Roeiy A, Madar, Z. Cellular mechanisms of insulin resistance in polycystic


ovarian syndrome. J. Clin. Endocrinol. Metab, 1992; 75:577–83.

➢ Cibula D, Skrha J, Hill L, Fanta M, Haakova A, Zivny J. Prediction of insulin sensitivity


in non obese women with polycystic ovary syndrome. J Clin Endocrinol Metab, 2002;
87:5821–5.

➢ Dankner R, Chetrit A, Shanik MH, et al. Basal-state hyperinsulinemia in healthy


normoglycemic adults is predictive of type 2 diabetes over a 24-year follow-up: a
preliminary report. Diabetes Care 2009;32(8):1464–1466. doi:10.2337/dc09-0153.

➢ Day F, Karaderi T, Jones MR, et al. Large-scale genome-wide meta-analysis of polycystic


ovary syndrome suggests shared genetic architecture for different diagnosis criteria. PLoS
Genet 2018;14(12):e1007813. doi:10.1371/journal.pgen.1007813.

➢ De Leo V, La Marca A, Petraglia F. Insulin-lowering agents in the management of


polycystic ovary syndrome. Endocr Rev 2003;24(5):633–667. doi:10.1210/er.2002-0015.

➢ Demirel MA, Ilhan M, Suntar I, Keles H, Akkol EK. Activity of Corylus avellana seed oil
in letrozole-induced polycystic ovary syndrome model in rats. Revista Brasileira de
Farmacognosia. 2016 Jan;26:83-8.

➢ Diamanti- Kandarakis E, Argyrakopoulou G, Economou F, Kandaraki E, Koutsilieris M.


Defects in insulin signaling pathways in ovarian steroidgenesis and other tissues in
polycystic ovary syndrome (PCOS). J Steroid Biochem Mol Biol, 2008; 109:242–6.

➢ Diez-Fraile A, Lammens T, Tilleman K, et al. Age-associated differential microRNA


levels in human follicular fluid reveal pathways potentially determining fertility and
success of in vitro fertilization. Hum Fertil 2014;17(2):90–98.
doi:10.3109/14647273.2014.897006.

➢ Feng, Y, Li X, Shao R. Genetic modeling of ovarian phenotypes in mice for the study of
human polycystic ovary syndrome. Am J Transl Res, 2013; 5(1):15– 20.

COLLEGE OF PHARMACY, RIMT UNIVERSITY, MANDI GOBINDGARH, PUNJAB-147301


PAGE 18
BIBLIOGRAPHY

➢ Gambineri A, Pelusi C, Vicennati V, Piggott U, Pasquali R. Obesity and the polycystic


ovary syndrome”. Int J Obesity Relat Metab Discord, 2002; 26(7):883– 96.

➢ Ghosh D, Chatterjee K, De D, Jana K, Ali K, Bera T. Antihyperglycemic and anti oxidative


effects of the hydro-methanolic extract of the seeds of Caesalpinia bonducella on
streptozotocin-induced diabetes in male albino rats. Pharmacognosy Res, 2012; 4:57.

➢ Goldrat O, Delbaere A. PCOS: update and diagnostic approach. ClinBiochem 2018;62:24–


31. doi:10.1016/j.clinbiochem.2018.09.001.

➢ Gurunath HM, Sidramayya MS, Patil KS, Wadkar GH. Effect of Caesalpinia bonducella
seed extracts on human neutrophil. J Pharmacy Res, 2010; 3(3):467–9.

➢ Hiam D, Moreno-Asso A, Teede HJ, et al. The genetics of polycystic ovary syndrome: an
overview of candidate gene systematic reviews and genome-wide association studies. J
Clin Med 2019;8(10):1606. doi:10.3390/jcm8101606.

➢ Hoeger KM, Dokras A, Piltonen T. Update on PCOS: consequences, challenges, and


guiding treatment. J Clin Endocrinol Metab 2021;106(3):e1071– e1083.
doi:10.1210/clinem/dgaa839.

➢ Kulshreshtha B, Pahuja I, Kothari D, Chawla I, Sharma N, Gupta S, Mittal A. Menstrual


cycle abnormalities in patients with pro lactinoma and drug-induced hyperpro lactinemia.
Indian J Endocrinol Metab, 2017; 21:545-50.

➢ Li M, Chi X, Wang Y, et al. Trends in insulin resistance: insights into mechanisms and
therapeutic strategy. Signal Transduct Target Ther 2022;7(1):216. doi:10.1038/s41392-
022-01073-0.

➢ Li, Y., Ma, H., Zhang, Y., Kuang, H., Hung, E., Ng, Y., et al., Effect of berberine on
insulin resistance in women with polycystic ovary syndrome: study protocol for a
randomized multicenter controlled trial. Trials. 2013; 14:226.

➢ Liu J, Qu T, Li Z, Yu L, Zhang S, Yuan D, et al. Serum kisspeptin levels in polycystic


ovary syndrome: A meta-analysis. J Obstet Gynaecol Res 2021;47:2157–65.

COLLEGE OF PHARMACY, RIMT UNIVERSITY, MANDI GOBINDGARH, PUNJAB-147301


PAGE 19
BIBLIOGRAPHY

➢ Long W, Zhao C, Ji C, et al. Characterization of serum microRNAs profile of PCOS and


identification of novel non-invasive biomarkers. Cell Physiol Biochem 2014;33(5):1304–
1315. doi:10.1159/000358698.

➢ Mantzoros C, Flier J. Insulin resistance: the clinical spectrum. Adv Endocrinol Metab
1995;6:193–232.

➢ Marshall K. Polycystic ovary syndrome: clinical considerations. Altern Med Rev, 2001;
6(3):272–92.

➢ Meera Murugesan B, Muralidharan P, Hari R. Effect of ethanolic seed extract of


Caesalpinia bonducella on hormones in mifepristone induced PCOS rats. J Appl Pharm
Sci, 2020; 10(02):072–076.

➢ Mu L, Sun X, Tu M, et al. Non-coding RNAs in polycystic ovary syndrome: a systematic


review and meta-analysis. Reprod Biol Endocrinol 2021;19(1):1–18. doi:10.1186/s12958-
020-00687-9.

➢ Nadaf R. A study of hypoglycemic effect of Caesalpinia bonducella extract on alloxan


induced diabetic albino rats. Int J Basic Clin Pharmacol, 2017; 6:2153.

➢ Ogunlade B, Adelakun SA, Akinola BK, Adedotun OA. Histomorphometric and collagen
fibers organization in annular punctured induced intervertebral disc degeneration in a
rabbit model: restorative and antioxidant role of Brassica. Advances in Traditional
Medicine 2021;21(2):341–50.

➢ Oyeniran DA, Ojewale AO, Jewo PI, Ashamu EA, Adeniyi, Adelakun SA. Infertility: A
product of smoke emanating from transfluthrin-coated insecticide paper
(TCIP).Toxicology Research and Application. 2021; 5: 23978473211025467.

➢ Roth LW, McCallie B, Alvero R, et al. Altered microRNA and gene expression in the
follicular fluid of women with polycystic ovary syndrome. J Assist Reprod Genet
2014;31(3):355–362. doi:10.1007/s10815-013-0161-4.

COLLEGE OF PHARMACY, RIMT UNIVERSITY, MANDI GOBINDGARH, PUNJAB-147301


PAGE 20
BIBLIOGRAPHY

➢ Saddick SY. Identifying genes associated with the development of human polycystic ovary
syndrome. Saudi J Biol Sci 2020;27(5):1271– 1279.doi:10.1016/j.sjbs.2020.01.012.

➢ Sanchez-Criado JE, Sanchez A, Ruiz A, Gaytan F. Endocrine and morphological features


of cystic ovarian condition in antiprogesterone RU486- treated rats. Acta Endocrinol
(Copenh), 1993; 129:237–45.

➢ Sharma, R. S., Dwivedi, S. K., and Swarup, D., Hypoglycaemic, anti- hyperglycemic,
hypolipidemic activities of Caesalpinia bonducella seeds in rats. J. Ethnopharmacol. 1997;
58: 39-44.

➢ Shen HR, Qiu LH, Zhang ZQ, et al. Genome-wide methylated DNA immune precipitation
analysis of patients with polycystic ovary syndrome. PLoS One 2013;8(5):e64801.
doi:10.1371/journal.pone.0064801.

➢ Shukla S, Mehta A, Mehta P, Bajpai VK. Evaluation of comparative antidiabetic effects


of ethanolic extracts of Caesalpinia bouncucella and Stevia rebaudianain normal and
alloxan-induced experimental rats. Romanian Biotechnol Lett, 2011; 16:6187–99.

➢ Shukla, S., Mehta, A., John, J., Singh, S., Mehta, P., and Vyas, S. P., Antioxidant activity
and total phenolic content of ethanolic extract of Caesalpinia bonducella seeds. Food.
Chem. Toxicol. 2009b; 47: 1848-51.

➢ Shukla, S., Mehta, A., Mehta, P., Vyas, S. P., Shukla, S., and Bajpai, V. K., Studies on
anti-inflammatory, antipyretic and analgesic properties of Caesalpinia bonducella F. seed
oil in experimental animal models. Food. Chem. Toxicol. 2010; 48: 61-64.

➢ Sirotkin AV. Cytokines: signalling molecules controlling ovarian functions. Int J Biochem
Cell Biol 2011;43(6):857–861. doi:10.1016/j. biocel.2011.03.001.

➢ Spritzer PM. Inflammatory mediators in polycystic ovary syndrome: the case of


interleukin-18. Arch Endocrinol Metab 2022;66(1):1–2. doi:10.20945/2359-
3997000000455.

COLLEGE OF PHARMACY, RIMT UNIVERSITY, MANDI GOBINDGARH, PUNJAB-147301


PAGE 21
BIBLIOGRAPHY

➢ Strauss JF III. Some new thoughts on the pathophysiology and genetics of polycystic ovary
syndrome. Ann N Y Acad Sci 2003;997(1):42–48. doi:10.1196/annals.1290.005.

➢ Teede H, Deeks A, Moran L. Polycystic ovary syndrome: a complex condition with


psychological, reproductive and metabolic manifestations that impacts on health across
the lifespan. BMC medicine. 2010 Dec;8:1-0.

➢ Teede H, Deeks A, Moran L. Polycystic ovary syndrome: a complex condition with


psychological, reproductive and metabolic manifestations that impacts on health across
the lifespan. BMC Med, 2010; 8:41.

➢ Teede H, Hutchison S, Zoungas S, Meyer C. Insulin resistance, the metabolic syndrome,


diabetes and cardiovascular disease risk in women with PCOS. Endocrine, 2006; 30:45–
53.

➢ Teede HJ, Misso ML, Costello MF, Dokras A, Laven J, Moran L, et al. Recommendations
from the international evidence-based guideline for theassessment and management of
polycystic ovary syndrome. Clin Endocrinol 2018;89:251–68.

➢ The Wealth of India, Raw material, Ca-Ci, Revised Edn, Publication And Information
Directorate, CSIR, New Delhi, 1992, 3, 6-8.

➢ Trent M, Gordon CM. Diagnosis and management of polycystic ovary syndrome in


adolescents. Pediatrics 2020;145(Suppl. 2):S210–8.

➢ Ukwenya VO, Adelakun SA, Fawepe TA, Adeagbo AS. The impact of deranged glucose
metabolism and diabetes in the pathogenesis of the novel Sars- Cov-2:Asystemic review
of the literature. Current Diabetes. Review 2021:e060821195355.

➢ Wallace IR, McKinley MC, Bell PM, Hunter SJ. Sex hormone binding globulin and insulin
resistance. Clin Endocrinol, 2013; 78(3): 321–9.

➢ Wang T, Qian YY, Huang J, et al. The relationship between insulin resistance and CpG
island methylation of LMNA gene in polycystic ovary syndrome. Cell Biochem Biophys
2013;67:1041–1047. doi:10.1007/ s12013-013- 9602-z.

COLLEGE OF PHARMACY, RIMT UNIVERSITY, MANDI GOBINDGARH, PUNJAB-147301


PAGE 22
BIBLIOGRAPHY

➢ Xu Y, Fu JF, Chen JH, et al. Sulforaphane ameliorates glucose intolerance in obese mice
via the upregulation of the insulin signaling pathway. Food Funct 2018;9(9):4.

➢ Yung Y, Maman E, Ophir L, Rubinstein N, Barzilay E, Yerushalmi GM, Hourvitz A.


Progesterone antagonist, RU486, represses LHCGR expression and LH/hCG signaling in
cultured luteinized human mural granulosa cells. Gynecol Endocrinol, 2014; 30:42–7.

COLLEGE OF PHARMACY, RIMT UNIVERSITY, MANDI GOBINDGARH, PUNJAB-147301


PAGE 23

You might also like