20
www.wjpps.com Vol 8, Issue 8, 2019. 1615 Prabhavalkar et al. World Journal of Pharmacy and Pharmaceutical Sciences EXPERIMENTAL ANIMAL MODELS FOR GASTROINTESTINAL ULCER DISEASE Mrunal Patil a and Kedar S. Prabhavalkar a, * a Department of Pharmacology, SVKM’s Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India. ABSTRACT Gastric ulcer is one of the most serious gastrointestinal disease. Ulcers are mainly caused by imbalance between the gastroduodenal mucosal defensive factors versus aggressive factors. In gastric ulcer disease, animal models have helped to understand the basic mechanisms responsible for the formation of gastric ulcers and its treatment. The limited number of animal models for the study of gastric ulcer has hindered the process of research in gastrointestinal disorders. Therefore it is important to review the available literature for animal models of gastric ulcer. Available models include primates, rats, mice, rabbits, cats, guinea pigs, ferrets and pigs. The main aim is to provide scientific information about different animal models to induce gastric ulcers as well as for the screening of antiulcer activity. In this review, we summarize different experimental animal models used in clinical research during past few decades to carry out antiulcerative activity of new agents as well as its underlying mechanisms. KEYWORDS: Gastric ulcer; animal models; NSAIDs; antiulcerative activity; diethyldithiocarbamate; COX. 1.0 INTRODUCTION Gastric ulcer is a common gastrointestinal disorder, which includes both gastric and duodenal ulcers affecting many people throughout the world. [1] It affects around 3%10% of the global population. [2] About 10% of the world population is on high risk of developing gastric ulcer at some point in their lifetime. [3] Globally, an estimated 15 mortality was recorded per year out of every 15,000 complications of gastric ulcer. [4] It can be defined as a damage to the WORLD JOURNAL OF PHARMACY AND PHARMACEUTICAL SCIENCES SJIF Impact Factor 7.421 Volume 8, Issue 8, 1615-1634 Review Article ISSN 2278 – 4357 *Corresponding Author Dr. Kedar S. Prabhavalkar Department of Pharmacology, SVKM’s Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India. Article Received on 17 June 2019, Revised on 07 July 2019, Accepted on 28 July 2019 DOI: 10.20959/wjpps20198-14539

EXPERIMENTAL ANIMAL MODELS FOR GASTROINTESTINAL …

  • Upload
    others

  • View
    4

  • Download
    0

Embed Size (px)

Citation preview

www.wjpps.com Vol 8, Issue 8, 2019.

1615

Prabhavalkar et al. World Journal of Pharmacy and Pharmaceutical Sciences

EXPERIMENTAL ANIMAL MODELS FOR GASTROINTESTINAL

ULCER DISEASE

Mrunal Patila and Kedar S. Prabhavalkar

a,*

aDepartment of Pharmacology, SVKM’s Dr. Bhanuben Nanavati College of Pharmacy, Vile

Parle (W), Mumbai, India.

ABSTRACT

Gastric ulcer is one of the most serious gastrointestinal disease. Ulcers

are mainly caused by imbalance between the gastroduodenal mucosal

defensive factors versus aggressive factors. In gastric ulcer disease,

animal models have helped to understand the basic mechanisms

responsible for the formation of gastric ulcers and its treatment. The

limited number of animal models for the study of gastric ulcer has

hindered the process of research in gastrointestinal disorders.

Therefore it is important to review the available literature for animal

models of gastric ulcer. Available models include primates, rats, mice,

rabbits, cats, guinea pigs, ferrets and pigs. The main aim is to provide

scientific information about different animal models to induce gastric

ulcers as well as for the screening of antiulcer activity. In this review,

we summarize different experimental animal models used in clinical research during past few

decades to carry out antiulcerative activity of new agents as well as its underlying

mechanisms.

KEYWORDS: Gastric ulcer; animal models; NSAIDs; antiulcerative activity;

diethyldithiocarbamate; COX.

1.0 INTRODUCTION

Gastric ulcer is a common gastrointestinal disorder, which includes both gastric and duodenal

ulcers affecting many people throughout the world.[1]

It affects around 3%–10% of the global

population.[2]

About 10% of the world population is on high risk of developing gastric ulcer

at some point in their lifetime.[3]

Globally, an estimated 15 mortality was recorded per year

out of every 15,000 complications of gastric ulcer.[4]

It can be defined as a damage to the

WORLD JOURNAL OF PHARMACY AND PHARMACEUTICAL SCIENCES

SJIF Impact Factor 7.421

Volume 8, Issue 8, 1615-1634 Review Article ISSN 2278 – 4357

*Corresponding Author

Dr. Kedar S.

Prabhavalkar

Department of

Pharmacology, SVKM’s Dr.

Bhanuben Nanavati College

of Pharmacy, Vile Parle

(W), Mumbai, India.

Article Received on

17 June 2019,

Revised on 07 July 2019,

Accepted on 28 July 2019

DOI: 10.20959/wjpps20198-14539

www.wjpps.com Vol 8, Issue 8, 2019.

1616

Prabhavalkar et al. World Journal of Pharmacy and Pharmaceutical Sciences

mucosa that ruptures the muscle layer and forms an injury followed by inflammation.[2], [5]

Generally normal gastric mucosa is exposed to the offensive factors. This may result from an

imbalance between the defensive factors and the offensive factors present in the gastric

mucosal layer.[6]

The defensive factors like mucin, adequate blood flow, nitric oxide,

prostaglandin secretion, bicarbonate and growth factors and the offensive factors include

increased secretion of hydrochloric acid and pepsin, reactive oxygen species, improper

dietary habits, administration of non-steroidal anti-inflammatory drugs, consumption of

alcohol, stressful conditions and infection with Helicobacter pylori infection.[7], [8]

So the

imbalance between these offensive factors and the defensive factors is the main cause for the

pathogenesis of gastric ulcers.[9]

The gastric acid, reactive oxygen species (ROS), and

inflammatory cytokines are continuously in contact with gastric mucosa and can lead to

gastric tissue damage.[10]

Nowadays, the drug therapy for gastric ulcer is mainly based on the

reduction in acid secretion and the protection of gastric mucosa.[11]

The drugs which are

commonly used for the treatment of gastric ulcers consist of antacids, anticholinergics, proton

pump inhibitors and H2- receptor antagonists.[12], [13]

However, the current therapy is not

completely effective and continuous use of these drugs can produce side effects[14]

such as

hypersensitivity, hematopoietic changes, gynecomastia and arrhythmia.[15]

The search for a

new, effective and affordable ulcer treatment, reducing its side effects, current studies are

moving towards natural products.[16]

Hence for the screening of novel antiulcer agents,

suitable animal models are necessary, which can mimic the conditions and show resemblance

to that of the human disease state. Therefore it is important to review the available literature

for animal models of gastric ulcer to screen antiulcer agents. Researchers have used some

technologies to generate transgenic rats[17]

, cats[18]

, dogs[19]

, rabbits, pigs, sheep[20]

, goats,

cattle, chickens[21]

, zebrafish[22]

and non-human primates[23]

as animal models to study

different disease mechanisms.

The main aim is to provide scientific information about different animal models to induce

gastric ulcers as well as for the screening of antiulcer activity. Therefore in this review,

authors putforth different experimental animal models used in clinical research during past

few decades to carry out antiulcer activity of new agents as well as its underlying

mechanisms.

www.wjpps.com Vol 8, Issue 8, 2019.

1617

Prabhavalkar et al. World Journal of Pharmacy and Pharmaceutical Sciences

2.0 Animal models

To study the actual pathogenesis and mechanisms of gastric ulcer in humans, rodent models

are helpful.[24]

Many scientists are using different animal models to induce gastric ulcer such

as pylorus ligation model, alcohol induced ulcers, NSAIDs induced ulcers, stress ulcers,

histamine induced gastric ulcers, cysteamine induced duodenal ulcers. These models could

also serve as effective tool for the better understanding of pathophysiological mechanisms,

like antisecretory, gastroprotective, antioxidant and healing of ulcers. But the limited number

of animal models for the study of gastric ulcer has hindered the process of research in

gastrointestinal disorders.

Certain conditions should be fulfilled while selecting the proper experimental animal model

for the screening of antiulcer agents.

It should be simple and reproducible.

It should be easy for the quantification of the results.

It should induce a proper ulceration at targeted site.

It should involve different mechanisms by which ulcers are produced.

Gastric ulcer can be induced by various methods and can be classified as (A) Physical

methods, (B) Chemical methods and (C) Surgical methods. These models are listed in

Table 1.

Table 1. List of Animal models.

Methods Animal models

Physical methods

Stress induced ulcers

Water immersion stress induced ulcers

Cold restraint stress induced ulcers

Chemical methods

NSAIDs induced gastric ulcers

Ethanol induced gastric ulcers

HCl/ethanol induced gastric ulcers

Acetic acid induced gastric ulcer

Serotonin induced gastric ulcer

Reserpine induced gastric ulcers

Diethyldithiocarbamate induced gastric ulcers

Histamine induced duodenal ulcers

Cysteamine HCl induced duodenal ulcers

Surgical methods

Pylorus ligation induced gastric ulcers

Ischemis reperfusion induced gastric ulcers

Mucosectomy induced gastric ulcers

www.wjpps.com Vol 8, Issue 8, 2019.

1618

Prabhavalkar et al. World Journal of Pharmacy and Pharmaceutical Sciences

The principle along with their underlying mechanism of action is described below and a brief

procedure for the same is given in table 2.

2.1 Physical methods

Certain physical methods are used to induce gastric ulcers in experimental animals. Generally

physical methods are responsible for the production of oxidative stress and increases reactive

oxygen species in the stomch.

2.1.1 Water-immersion stress or cold-resistant stress induced gastric ulcers

Physical stress and psychological stress can cause gastric ulcers in humans. Hence various

stressors are responsible for the production of gastric ulcers in animal models.[25] This

model involves the restraint technique developed by Brodie and Hanson[26]

employed with

coupling of new method developed by Levine i.e. cold water immersion method which

induces gastric ulcers in a synergistic manner. Gastric ulcers induced by water-immersion

stress or cold water-restraint stress or cold-restraint stress in animals are completely resemble

to human ulcer condition by histopathogically.[27]

This model is widely used to study the

gastroprotective and healing effect of test agents, especially those with mucus enhancing and

cytoprotective properties, for gastric ulcers in rats. The pathophysiology behind stress-

induced gastric ulcers is complex and difficult to understand. Generally these ulcers are

produced due to the release of histamine, which resulting into increased acid secretion and

reduced mucus production[22]

and poor gastric blood flow.[28]

Excessive production of

reactive oxygen species and inhibition of prostaglandin synthesis also promote stress induced

ulcer formation.[29], [30]

Increased gastrointestinal motility produced by stress results into

formation of folds in the stomach[31]

which are more susceptible to damage gastric mucosa

when they come in contact with gastric acid. Increased vagal activity is also one of the factor

responsible for the production of stress-induced ulcers.[26]

As mucus plays a crucial role in

the protection of stomach wall and enhances healing of gastric ulcers, this model is suggested

for evaluating mucus enhancing and cytoprotective agents.

www.wjpps.com Vol 8, Issue 8, 2019.

1619

Prabhavalkar et al. World Journal of Pharmacy and Pharmaceutical Sciences

Table 2. Animal models of gastric and duodenal mucosal lesions and ulcers.

Assay Types of

animals Method of induction

Types of

lesions References

Stress induced gastric

ulcers

Water immersion stress

Rat

Animals are placed in

a stress cage and

immerse it vertically

in water bath (15–

20∘C) upto the level

of the xiphoid for 17

hours to induce stress

ulcer.

Gastric,

Acute,

Chronic

[32], [33]

Cold restraint stress Rat

Animals are

restrained in plastic

cages in a ventilated

refrigerator at 2-3∘C

for 2-4 hours and

then sacrificed.

Gastric,

Acute,

Chronic

[34]

NSAIDs induced ulcers

Aspirin

Rat or Mouse

24-36 hrs fasted

animals receive a

single dose of aspirin

(125-150 mg/kg)

administered by oral

route and after 4 hrs

animals are sacrificed

for ulcer

examination.

Gastric,

Acute

[28], [35]–[38]

Indomethacin Rat or Hamster

Animals receive a

single dose of

indomethacin in the

range of 40-100

mg/kg

subcutaneously and

sacrificed after 5 hrs.

Gastric,

Acute,

Chronic

[28], [37], [39]–[42]

Ethanol induced ulcers

(Absolute ethanol) Rat

After 24-36 hrs of

fasting, animals

receive ethanol

(1ml/rat) orally and

sacrificed after 1 hr

to check gastric

lesions.

Gastric,

Acute [41], [43]–[46]

HCl/ethanol induced

Ulcers Rat

Animals receive a

mixture of 1.0 mL

EtOH/HCl (60 mL

ethanol, 1.7 mL

hydrochloric acid,

38.3 mL distilled

Gastric,

Acute [47]

www.wjpps.com Vol 8, Issue 8, 2019.

1620

Prabhavalkar et al. World Journal of Pharmacy and Pharmaceutical Sciences

water) to induce

gastric ulcer.

Acetic acid induced

gastric ulcers Rat, Cat

After an acute

laparotomy, stomach

is exposed and 20%

acetic acid (0.03 mL)

is injected into the

sub-serosal layer of

the antrum at

multiple sites. After

24 hrs animals are

sacrificed.

Gastric,

Chronic [48]–[50]

Serotonin induced

ulcers Rat

After 30 min of drug

treatment, Serotonin

Creatinine Sulphate

(20-50 mg/kg) is

administered

subcutaneously and

after 6 hours animals

are sacrificed.

Gastric,

Acute [51], [52]

Reserpine induced

ulcers Rat

After 36 hrs of

fasting, reserpine (5–

8mg/kg) is

administered

intraperitoneally and

after 24 hrs animals

are sacrificed.

Gastric,

Acute [53]

Diethyldithiocarbamate

inducd ulcers Rat

In this model, acute

glandular lesions are

induced by

subcutaneous

injection of 1 mL of

diethyldithiocarbama

te (800mg/kg) in

saline followed by

oral dose of 1 mL of

0.1N HCl.

Gastric,

Acute [54], [55]

Histamine induced

ulcers Rat

Histamine phosphate

(40–100mg/kg) is

administered

subcutaneously and

after 2 hours animals

are sacrificed.

Gastric,

Duodenal,

Acute

[56], [57]

Cysteamine HCl

induced ulcers

Guinea pig,

Rat,

Mouse

A single dose of

cysteamine

hydrochloride

(400mg/kg p.o.) is

sufficient to induce

acute duodenal ulcers

in experimental rats.

Duodenal,

Acute [35], [36], [40]

www.wjpps.com Vol 8, Issue 8, 2019.

1621

Prabhavalkar et al. World Journal of Pharmacy and Pharmaceutical Sciences

Pylorus ligation induced

ulcers

Rat

After an acute

laparotomy, pyloric

end of animals

stomach is ligated for

4 hrs. Then animals

are sacrificed for

ulcerative lesions

examination.

Gastric,

Acute [38]

Ischemia reperfusion

induced ulcers Rats

A laparotomy is

carried out under

anesthesia, and the

superior mesenteric

artery (SMA) is

clamped with a

bulldog clip for 30

min (The ischemic

stage). After that,

bulldog clip is

removed to permit

reoxygenation of the

gastric tissue for 15

minutes (the

reperfusion stage).

Then animals are

sacrificed for

ulcerative lesions

examination.

Gastric,

Intestinal

Acute

[58], [59]

Mucosectomy induced

ulcers

Rabbits

Guinea pigs

After a median

laparotomy, 0.2 mL

of isotonic saline is

injected into the

submucosal layer of

the upper corpus. A

diameter of 7-10 mm

of the swollen

mucosal layer is

resected with

scissors. After the

procedure, animals

are placed to

individual cages for

recovery.

Gastric,

Acute [60]–[62]

2.2 Chemical methods

Various chemicals are used to induce ulcers because they directly ruptures the gastric mucosa

(Ethanol, Acetic acid) or some other mechanisms are also involved in it.

www.wjpps.com Vol 8, Issue 8, 2019.

1622

Prabhavalkar et al. World Journal of Pharmacy and Pharmaceutical Sciences

2.2.1 NSAID’s induced gastric ulcers

Gastric ulcers are known to be induced by excessive use of several Non-Steroidal Anti-

Inflammatory Drugs (NSAID’s) like aspirin, indomethacin, and ibuprofen.[63]

This is the most

common model used for inducing gastric ulcer. NSAID’s are responsible for the production

of gastric ulcers by inhibiting prostaglandin synthesis via cyclooxygenase (COX)

pathway.[64], [65]

Prostaglandins plays an important and protective role in the production of mucus and

secretion of bicarbonate, which maintains mucosal blood flow and regulates mucosal cell

integrity.[66]

Thus the suppression of production of prostaglandins by NSAIDs leads to the

gastric mucosal damage and gastric ulcers. The mechanism behind NSAIDs -induced gastric

ulceration includes blocking activity of the cyclooxygenase enzymes (COX-1 and COX-2),

by NSAIDs, which further leads to the decreased mucus and bicarbonate secretion, impaired

platelet aggregation, decreased mucosal blood flow, changes in microvascular structures

leading to epithelia damage.[65]

Increased reactive oxygen species (ROS) and lipid

peroxidation (LPO) are also responsible for gastric mucosal damage.[67]

NSAID’s specially

those are having chemically acidic nature, can exert direct cytotoxic effects on epithelial

cells, which disrupt surface active phospholipids on the mucosal surface thus making the

mucosa more susceptible to damage by luminal acid.[68]

Thus this model is commonly used

for the evaluation of antisecretory and cytoprotective agents. Specially, most commonly used

ulcerogen used for the induction of gastric ulcers are aspirin and indomethacin. A correct

route and an appropriate vehicle (e.g. water, 1% carboxymethyl cellulose) is selected for

administration of ulcerogen.

2.2.2 Ethanol induce gastric ulcers

Ethanol has been commonly used as a damaging agent to gastric mucosa for the induction of

gastric ulcers. Administration of absolute ethanol (>99%) has been used as a reproducible

method to induce gastric mucosal damage in experimental animals.[69]

The pathology behind

ethanol-induced gastric ulcer generally involves three main parameters: inflammatory

response, oxidative stress and apoptosis.

In the gastrointestinal tract, the motility of the esophagus, stomach, and gut as well as the

capacity of gut absorption can be severely affected by alcohol exposure. It can cause severe

mucosal damage and even carcinogenesis specially gastric cancer.[70]

Ethanol causes severe

gastric damage by producing several instabilities in the gastric mucosal layer[71]

such as a

www.wjpps.com Vol 8, Issue 8, 2019.

1623

Prabhavalkar et al. World Journal of Pharmacy and Pharmaceutical Sciences

decreased bicarbonate secretion, gastric blood flow and mucus production. Ethanol can

stimulate gastric acid secretion, resulting in microvascular injuries which facilitate vascular

permeability, through release of gastrin and histamine from sensitive nerve terminals present

in the gastric mucosa.[72]

Ethanol ruptures gastric mucosal integrity through exfoliation of

cells, which leads to increase in mucosal permeability and in somehow causes bleeding.[73],

[74] Intra-gastric administration of absolute ethanol results in severe gastric mucosal injury

characterized by disturbances in microcirculation, mast cell secretary products, inhibition of

prostaglandin synthesis, reduction in mucus production and reactive oxygen species.[75]

Neutrophil infiltration in the gastric mucosa is also responsible for the production of lesions

induced by absolute ethanol.[76]

Oxidative stress plays a significant role in alcohol-induced gastric mucosal damage.[77]

Ethanol is also known to increase cellular oxidative stress[78]

and produce changes in gastric

cell calcium levels[79]

which may lead to the pathogenesis of gastric mucosal injury.

However, it is not a suitable animal model to evaluate antisecretory activity and ulceration

dependent on acid secretion, because this model is independent of gastric acid secretion.

Ethanol directly increases the levels of free radicals that can alter the cell structure and

function[75]

and can also gives its direct toxic effects on the gastric mucosa resulting in

reduced bicarbonate secretion and gastric mucus production.[80]

Hence this model is more

preferable for evaluating the gastroprotective potential of test materials which has

cytoprotective as well as antioxidant activities.

2.2.3 HCl/ethanol induced gastric lesions

This model is considered to be an advanced model of an absolute ethanol induced ulcer

model. Instead of ethanol only, a mixture of HCl and ethanol is used to induce ulceration.

Direct necrotizing action of HCl/ethanol is the pathogenesis for gastric lesions on the gastric

mucosa. Hence the combination of ethanol with HCl leads to induction and progression of

gastric injury.[81]

2.2.4 Acetic acid-induced gastric ulcers

Chronicity of the gastric ulcer disease is one of the least understood aspects for researchers.

Takagi et al.,[82]

developed an animal model to induce chronic gastric ulcers by sub-mucosal

injection of acetic acid and he also reported healing process of ulcers for extended time

intervals after the ulcer formation. The experimental gastric ulcers induced by acetic acid are

www.wjpps.com Vol 8, Issue 8, 2019.

1624

Prabhavalkar et al. World Journal of Pharmacy and Pharmaceutical Sciences

considered as chronic ulcers due to its prolonged steadiness in gastric mucosa and

resemblance to human chronic ulcer both physiologically and histologically.

To overcome certain problems like adherence of ulcer to the adjacent organs such as the liver,

method given by Takagi, was modified and the one that is currently most commonly used is

developed by Okabe and Pfeiffer.[83]

This method consists of intraluminal application of

acetic acid solution. Thus acetic acid induced gastric ulcer model is most suitable for

evaluating antisecretory and cytoprotective activity of various test agents against chronic

gastric ulcers.[82], [84]

This model is easy and reliable to produce round and deep ulcers in the

stomach and duodenum of mice, rats, Mongolian gerbils, guinea pigs, cats, dogs, miniature

pigs, and monkeys.[49], [85]

2.2.5 Serotonin-induced gastric ulcers

Serotonin is a vasoconstrictor. It causes disturbance in gastric mucosal microcirculation and

reduces blood flow to gastric mucosa resulting in acute gastric mucosal damage which causes

gastric ulcers.[86]

2.2.6 Reserpine-Induced Peptic Ulcer

Reserpine is also used for induction of gastric ulcers by scientist. Reserpine causes

degranulation of mast cells resulting into release of histamine, mediated by cholinergic

system[87]

and this histamine is responsible for formation of gastric ulcer. Although this

model is dependent on gastric acid secretion, gastric hypermotility also plays an important

role in the induction of gastric mucosal lesions.[53]

2.2.7 Diethyldithiocarbamate induced gastric ulcers

This model is useful to assess the antioxidant activity of test drug which is mediates

gastroprotection by preventing gastric damage[54]

and also to evaluate the cytoprotective

potential of test agents. It is reported that antral lesions are induced by diethyldithiocarbamate

through the mobilization of superoxide and hydroxyl radicals. Superoxide radical and

hydroxyl radicals play a pathogenic role in the induction of this ulcer.[55]

2.2.8 Histamine-induced gastric ulcers

Release of histamine in stomach also causes gastric ulcers and hence histamine can be used

for inducing gastric ulcers in animals. On this basis histamine induced gastric ulcer model is

developed.[57]

Mast cells secretes histamine, which gets bind with receptors which are present

www.wjpps.com Vol 8, Issue 8, 2019.

1625

Prabhavalkar et al. World Journal of Pharmacy and Pharmaceutical Sciences

on the surface of parietal cells, resulting into activation of adenylate cyclase, which is

responsible for the conversion of ATP into c-AMP. This process of conversion of AMP to c-

AMP enhances gastric acid secretion from parietal cells. Histamine produces disturbances in

gastric mucosal layer subsequently cause severe damage in gastric mucosa.[88]

The mechanism by which histamine produces gastric ulcers is its potent acid stimulating and

vasodilatory effect in animals. Vasodilating capability of histamine causes increase in

vascular permeability.[89]

This model is commonly used for evaluation of antisecretory effect

of test drug which acts as H2 receptor antagonists.

2.2.9 Cysteamine-Induced Duodenal Ulcers

Selye and Szabo[32]

depicted the technique for acceptance of duodenal ulcers in rodents by

utilizing cysteamine HCl. Duodenal ulcers incited by cysteamine in rodents has been

generally utilized as a model of duodenal ulcer. Cysteamine prompted ulcer looks like

duodenal ulcer in human as for its area, histopathology and a few parts of pathophysiology

too. The genuine component associated with generation of ulcer by cysteamine has not been

completely known. But it is reported that Cysteamine is responsible for the excessive

secretion of gastric acid and inhibits the alkaline mucus secretion from Brunner’s glands in

the proximal duodenum resulting in the formation of duodenal ulcer. Cysteamine is also

responsible for excessive pepsin secretion in the gastric mucosa[90]

and consequently causes

decrease in the production in defensive factors like bicarbonate and mucus.[91]

From different

studies, it was reported that cysteamine also reduces somatostatin bioavailability and elevates

serum gastrin levels, which is associated with an increase in gastric acid secretion.[92]

Additionally certain transcription factors like early growth response factor-1, hypoxia-

inducible factor-1 and their target genes assumes a vital role in the pathogenesis of

cysteamine-induced duodenal ulcers.[57]

2.3 Surgical methods

By performing a surgical method we can induce gastric ulcers in experimental animals.

2.3.1 Pylorus-ligation induced peptic ulcer

Shay (1945) developed an animal model for the investigation of gastric secretory activity of a

drug by ligating the pylorus end of the stomach which causes accumulation of gastric acid in

the stomach and produces ulcers. This excessive gastric acid is responsible for auto digestion

of gastric mucosal layer which cause breakdown of the gastric mucosa. So obstruction in

www.wjpps.com Vol 8, Issue 8, 2019.

1626

Prabhavalkar et al. World Journal of Pharmacy and Pharmaceutical Sciences

pylorus causes mucosal digestion by an increase in acid pepsin pool. This model is

commonly used for the evaluation of cytoprotective and antisecretory effect of a drug which

increases secretion of mucus and reduce secretion of gastric acid, respectively.[57], [93]

2.3.2 Ischemia-Reperfusion induced gastric ulcer

Gastric mucosa is highly sensitive to ischemia or ischemic shock. Hence reperfusion

followed by ischemia causes formation of free radicals which is responsible for gastric

mucosal injury.[57]

This involves damage to the muscularis mucosac which is developed by

clamping the celiac artery. Intestinal injury is generally induced by 60 min clamping of the

superior mesenteric artery followed by 60 min reperfusion.[59]

2.3.3 Mucosectomy induced gastric ulcer

This is also called as mucosal resection method where gastric mucosa is exposed by median

laparotomy. Normal saline is injected into the submucosal layer of stomach and swollen

mucosal area is resected with scissors.[61]

Endoscopic resections have certain advantages over

conventional methods. They are more economical and less invasive.[62]

3.0 CONCLUSION

Actually it is not possible to understand the actual mechanism of any disease in human. No

one can predict which is the actual factor or mediator responsible for disease progression.

Without knowing this basic information, it is difficult to design treatment strategies for any

disease. In this review, we tried to cover several animal models of gastrointestinal ulcers that

are widely used in clinical research. Here we also reviewed available experimental animal

models of gastrointestinal ulcers for the evaluation new medicinal compounds having

potential antiulcerative as well as gastroprotective activity. In each model. we have discussed

the underlying mechanism behind the production of gastric ulcer. This will definitely help

scientists and investigators to select a suitable animal model of gastric ulcer for the evaluation

of antiulcerative activity of the test compound. Animal models also plays an important role in

pre-clinical research, specially for the identification of drug targets.

4.0 Abbreviations

ROS- Reactive Oxygen Species

NSAIDs- Non-Steroidal Anti Inflammatory Drugs

HCl- Hydrochloric Acid; SMA- Superior Mesenteric Artery

COX- cyclooxygenase

www.wjpps.com Vol 8, Issue 8, 2019.

1627

Prabhavalkar et al. World Journal of Pharmacy and Pharmaceutical Sciences

COX 1- Cyclooxygenase 1

COX 2- Cyclooxygenase 2

ATP- Adenosine triphosphate

c-AMP- Cyclic Adenosine monophosphate.

5.0 Conflict of interest

Authors have no conflict of interest.

6.0 ACKNOWLEDGEMENT

Authors are thankful to Dr. Lokesh kumar Bhatt, HOD Pharmacology department and Dr.

Munira Momin, Principal, SVKM’s Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle,

Mumbai for their kind support.

REFERENCES

1. N. Rasheed et al., ―Differential response of A 68930 and sulpiride in stress-induced

gastric ulcers in rats,‖ Eur. J. Pharmacol., 2010; 643(1): 121–128.

2. J. M. Kirsch and C. Hirsch-Reilly, ―Peptic ulcer disease,‖ Acute Care Gen. Surg. Work.

Manag., 2017; 38(3): 159–164.

3. F. M. Snowden, ―Emerging and reemerging diseases: A historical perspective,‖ Immunol.

Rev., 2008; 225(1): 9–26.

4. H. M. A. Sidahmed et al., ―Phytomedicine Pyranocycloartobiloxanthone A , a novel

gastroprotective compound from Artocarpus obtusus Jarret , against ethanol-induced

acute gastric ulcer in vivo,‖ Eur. J. Integr. Med., 2013; 20(10): 834–843.

5. P. Aro et al., ―Peptic ulcer disease in a general adult population: The kalixanda study: A

random population-based study,‖ Am. J. Epidemiol., 2006; 163(11): 1025–1034.

6. A. Aslan et al., ―The inhibition of gastric mucosal lesion, oxidative stress and neutrophil-

infiltration in rats by the lichen constituent diffractaic acid,‖ Phytomedicine, 2005; 13(8):

584–590.

7. M. Lemos et al., ―Copaifera langsdorffii: Evaluation of potential gastroprotective of

extract and isolated compounds obtained from leaves,‖ Brazilian J. Pharmacogn., 2015;

25(3): 238–245.

8. K. Sowndhararajan and S. Chul, ―Protective effect of ethyl acetate fraction of Acacia

ferruginea DC. against ethanol-induced gastric ulcer in rats,‖ J. Ethnopharmacol., 2013;

148(1): 175–181.

9. M. K. Choudhary, S. H. Bodakhe, and S. K. Gupta, ―Assessment of the Antiulcer

www.wjpps.com Vol 8, Issue 8, 2019.

1628

Prabhavalkar et al. World Journal of Pharmacy and Pharmaceutical Sciences

Potential of Moringa oleifera Root-Bark Extract in Rats,‖ J. Acupunct. Meridian Stud.,

2013; 6(4): 214–220.

10. A. Bhattacharyya, R. Chattopadhyay, S. Mitra, and S. E. Crowe, ―Oxidative Stress: An

Essential Factor in the Pathogenesis of Gastrointestinal Mucosal Diseases,‖ Physiol. Rev.,

2014; 94(2): 329–354.

11. P. Malfertheiner, F. K. L. Chan, and K. E. L. Mccoll, ―Peptic ulcer disease,‖ Lancet,

2009; 374(9699): 1449–1461.

12. A. E. Bighetti et al., ―Antiulcerogenic activity of a crude hydroalcoholic extract and

coumarin isolated from Mikania laevigata Schultz Bip,‖ Phytomedicine, 2005; 12(1–2):

72–77.

13. V. Lakshmi et al., ―Gedunin and Photogedunin of Xylocarpus granatum show significant

anti-secretory effects and protect the gastric mucosa of peptic ulcer in rats,‖

Phytomedicine, 2010; 17(8–9): 569–574.

14. V. Bassi, O. Fattoruso, M. T. Polistina, and C. Santinelli, ― Graves’ disease shows a

significant increase in the Helicobacter pylori recurrence ,‖ Clin. Endocrinol. (Oxf).,

2014; 81(5): 784–785.

15. S. Chanda, Y. Baravalia, and M. Kaneria, ―Protective effect of Polyalthia longifolia var .

pendula leaves on ethanol and ethanol / HCl induced ulcer in rats and its antimicrobial

potency,‖ Asian Pac. J. Trop. Med., 2011; 4(9): 673–679.

16. Mohd. Akhtar, M. A. Ahmad, S. Sumbul, and Mohd. Asif, ―Role of phenolic compounds

in peptic ulcer: An overview,‖ J. Pharm. Bioallied Sci., 2011; 3(3): 361.

17. W. E. Filipiak and T. L. Saunders, ―Advances in transgenic rat production,‖ Transgenic

Res., 2006; 15(6): 673–686.

18. E. Poeschla, T. Otoi, P. Wongsrikeao, D. Saenz, and T. Rinkoski, ―Antiviral restriction

factor transgenesis in the domestic cat,‖ Nat. Methods, 2011; 8(10): 853–859.

19. S. G. Hong et al., ―Generation of red fluorescent protein transgenic dogs,‖ Genesis, 2009;

47(5): 314–322.

20. R. E. Hammer et al., ―Production of transgenic rabbits, sheep and pigs by

microinjection,‖ Nature, 1985; 315(6021): 680–683.

21. I. M. Mansuy and U. Suter, ―Special Review Series — Gene Manipulation and

Integrative Physiology Mouse genetics in cell biology,‖ Exp. Physiol., 2000; 85(6):

713–31.

22. S. Higashijima, Y. Hotta, and H. Okamoto, ―Visualization of cranial motor neurons in

live transgenic zebrafish expressing green fluorescent protein under the control of the

www.wjpps.com Vol 8, Issue 8, 2019.

1629

Prabhavalkar et al. World Journal of Pharmacy and Pharmaceutical Sciences

islet-1 promoter/enhancer.,‖ J. Neurosci., 2000; 20(1): 206–18.

23. [E. Sasaki et al., ―Generation of transgenic non-human primates with germline

transmission,‖ Nature, 2009; 459(7246): 523–527.

24. N. Choudhary, L. K. Bhatt, and K. S. Prabhavalkar, ―Experimental animal models for

rheumatoid arthritis,‖ Immunopharmacol. Immunotoxicol., 2018; 0(0): 1–8.

25. [N. Sezgin, N. Gürbüz, S. Demirbilek, İ. Gürses, and A. Karaman, ―Protective effect of

polyunsaturated phosphatidylcholine pretreatment on stress ulcer formation in rats,‖ J.

Pediatr. Surg., 2003; 39(1): 57–62.

26. D. A. Brodie and H. M. Hanson, ―A Study of the Factors Involved in the Production of

Gastric Ulcers by the Restraint Technique,‖ Gastroenterology, 1960; 38(3): 353–360.

27. P. C. Konturek et al., ―Pioglitazone, a specific ligand of peroxisome proliferator-activated

receptor-gamma, accelerates gastric ulcer healing in rat,‖ Eur. J. Pharmacol., 2003;

472(3): 213–220.

28. P. H. Guth, ―Gastric blood flow in restraint stress,‖ Am. J. Dig. Dis., 1972; 17(9):

807–813.

29. H. Kitagawa, M. Fujiwara, and Y. Osumi, ―Effects of water-immersion stress on gastric

secretion and mucosal blood flow in rats,‖ Gastroenterology, 1979; 77(2): 298–302.

30. P. Tamashiro Filho et al., ―Evaluation of antiulcer activity and mechanism of action of

methanol stem bark extract of Lafoensia pacari A. St.-Hil. (Lytraceae) in experimental

animals,‖ J. Ethnopharmacol., 2012; 144(3): 497–505.

31. M. N. Peters and C. T. Richardson, ―Stressful Life Events, Acid Hypersecretion, and

Ulcer Disease,‖ Gastroenterology, 1983; 84(1): 114–119.

32. H. Selye and S. Szabo, ―Experimental model for production of perforating duodenal

ulcers by cysteamine in the Rat,‖ Nature, 1973; 244(5416): 458–459.

33. S. Guo, Q. Gao, Q. Jiao, W. Hao, X. Gao, and J. M. Cao, ―Gastric mucosal damage in

water immersion stress: Mechanism and prevention with GHRP-6,‖ World J.

Gastroenterol., 2012; 18(24): 3145–3155.

34. M. A. Morsy, G. H. Heeba, S. A. Abdelwahab, and R. R. Rofaeil, ―Protective effects of

nebivolol against cold restraint stress-induced gastric ulcer in rats: Role of NO, HO-1,

and COX-1,2,‖ Nitric Oxide - Biol. Chem., 2012; 27(2): 117–122.

35. S. Szabo, ―Animal model of human disease. Cysteamine induced acute and chronic

duodenal ulcer in the rat,‖ Am J Pathol, 1978; 93(1): 273–276.

36. S. Szabo and C. H. Cho, ―From Cysteamine to MPTP: Structure-Activity Studies with

Duodenal Ulcerogens,‖ Toxicol. Pathol., 1988; 16(2): 205–211.

www.wjpps.com Vol 8, Issue 8, 2019.

1630

Prabhavalkar et al. World Journal of Pharmacy and Pharmaceutical Sciences

37. B. J. R. WHITTLE, ―Mechanisms Underlying Gastric Mucosal Damage Induced By

Indomethacin and Bile‐Salts, and the Actions of Prostaglandins,‖ Br. J. Pharmacol.,

1977; 60(3): 455–460.

38. Q. Li et al., ―Activity of Brucea javanica oil emulsion against gastric ulcers in rodents,‖

Asian J. Pharm. Sci., 2018; 13(3): 279–288.

39. J. Liang et al., ―Prophylactic efficacy of patchoulene epoxide against ethanol-induced

gastric ulcer in rats: Influence on oxidative stress, inflammation and apoptosis,‖ Chem.

Biol. Interact., 2018; 283, no. November 2017: 30–37.

40. S. Szabo and C. H. Cho, ―Animal models for studying the role of eicosanoids in peptic

ulcer disease,‖ Eicosanoids Gastrointest. Tract, 1988; b(2): 75–102.

41. S. Szabo and C. H. Cho, ―From Cysteamine to MPTP: Structure-Activity Studies with

Duodenal Ulcerogens,‖ Toxicol. Pathol., 2009; 16(2): 205–211.

42. J. Z. Wu et al., ―Protective role of β-patchoulene from Pogostemon cablin against

indomethacin-induced gastric ulcer in rats: Involvement of anti-inflammation and

angiogenesis,‖ Phytomedicine, 2018; 39, no. November 2017: 111–118.

43. S. Szabo and C. H. Cho, ―Animal models for studying the role of eicosanoids in peptic

ulcer disease,‖ Eicosanoids Gastrointest. Tract, 2011; b(2): 75–102.

44. J. Y. C. Chow, L. Ma, and C. H. Cho, ―Effect of cigarette smoke on ethanol-induced

gastric mucosal lesions: The role of nitric oxide and neutrophils,‖ Eur. J. Pharmacol.,

1998; 342(2–3): 253–260.

45. J. K. S. Ko and C. H. Cho, ―Adaptive gastric mucosal cytoprotection in rats: Different

modes of action by three mild irritants,‖ Digestion, 1996; 57(1): 54–59.

46. I. Boutemine et al., ―Gastro-protective , therapeutic and anti-inflammatory activities of

Pistacia lentiscus L. fatty oil against ethanol-induced gastric ulcers in rats,‖ J.

Ethnopharmacol., 2018; 224(January): 273–282.

47. P. Baiubon, P. Kunanusorn, P. Khonsung, N. Chiranthanut, A. Panthong, and C.

Rujjanawate, ―Gastroprotective activity of the rhizome ethanol extract of Zingiber

simaoense Y. Y. Qian in rats,‖ J. Ethnopharmacol., 2016; 194(July): 571–576.

48. Keijiro Takagi and Susumu Okabe, ―Studies of the mechanisms involved in the

production of stress and stress-atropine ulcers in rats,‖ Eur. J. Pharmacol., 1970; 10(3):

378–384.

49. S. Okabe, J. L. A. Roth, and C. J. Pfeiffer, ―Differential healing periods of the acetic acid

ulcer model in rats and cats,‖ Experientia, 1971; 27(2): 146–148.

50. S. K. Bhattamisra et al., ―Protective activity of geraniol against acetic acid and

www.wjpps.com Vol 8, Issue 8, 2019.

1631

Prabhavalkar et al. World Journal of Pharmacy and Pharmaceutical Sciences

Helicobacter pylori- induced gastric ulcers in rats,‖ J. Tradit. Complement. Med., 2018;

1–9.

51. I. H. M. MAIN and B. J. R. WHITTLE, ―Investigation of the Vasodilator and

Antisecretory Role of Prostaglandins in the Rat Gastric Mucosa By Use of Non‐Steroidal

Anti‐Inflammatory Drugs,‖ Br. J. Pharmacol., 1975; 53(2): 217–224.

52. S. Okabe, Y. Takata, K. Takeuchi, T. Naganuma, and K. Takagi, ―Effects of

carbenoxolone Na on acute and chronic gastric ulcer models in experimental animals,‖

Am. J. Dig. Dis., 1976; 21(8): 618–625.

53. M. B. Gupta, K. K. Tangri, and K. P. Bhargava, ―Mechanism of ulcerogenic activity of

reserpine in albino rats,‖ Eur. J. Pharmacol., 1974; 27(2): 269–271.

54. T. Takemoto et al., ―Role of active oxygen species in diethyldithiocarbamate-induced

gastric ulcer in the rat,‖ Experientia, 2005; 46(3): 281–283.

55. A. S. Salim, ―Protection against stress-induced acute gastric mucosal injury by free

radical scavengers,‖ Intensive Care Med., 1991; 17(8): 455–460.

56. M. E. Parsons, ―Histamine and the pathogenesis of duodenal ulcer disease,‖ Gut, 1985;

26(11): 1159–1164.

57. M. B. Adinortey, C. Ansah, I. Galyuon, and A. Nyarko, ―In Vivo Models Used for

Evaluation of Potential Antigastroduodenal Ulcer Agents ,‖ Ulcers, 2013; 2013: 1–12.

58. A. T. Salami, O. A. Odukanmi, O. F. Faniyan, T. P. Omayone, and S. B. Olaleye, ―Seeds

of Buchholzia coriacea in Diet Mitigate Ischemic Reperfusion–Induced Gastric

Ulceration in Experimental Rats,‖ J. Diet. Suppl., 2018; 15(6): 842–859.

59. A. Zeng et al., ―Gastrin attenuates ischemia-reperfusion-induced intestinal injury in rats,‖

Exp. Biol. Med., 2016; 241(8): 873–881.

60. A. Onen, Z. Kanay, C. Guzel, D. Kurt, and K. Ceylan, ―The effects of allopurinol on

stomach mucosal barrier of rats subjected to ischemia-reperfusion,‖ Turkish J. Med. Sci.,

2000; 30(5): 449–451.

61. J. H. Maeng, E. Lee, D. H. Lee, and S.-G. Yang, ―Rabbit gastric ulcer models:

comparison and evaluation of acetic acid-induced ulcer and mucosectomy-induced ulcer,‖

Lab. Anim. Res., 2013; 29(2): 96.

62. J. H. Maeng et al., ―Endoscopic application of EGF-chitosan hydrogel for precipitated

healing of GI peptic ulcers and mucosectomy-induced ulcers,‖ J. Mater. Sci. Mater. Med.,

2014; 25(2): 573–582.

63. K. D. Rainsford, ―The effects of 5-1ipoxygenase inhibitors and leukotriene antagonists on

the development of gastric lesions induced by nonsteroidal antiinflammatory drugs in

www.wjpps.com Vol 8, Issue 8, 2019.

1632

Prabhavalkar et al. World Journal of Pharmacy and Pharmaceutical Sciences

mice,‖ Agents Actions, 1987; 21: 316–319.

64. H. Matsui, O. Shimokawa, T. Kaneko, Y. Nagano, K. Rai, and I. Hyodo, ―The

pathophysiology of non-steroidal anti-inflammatory drug (NSAID)-induced mucosal

injuries in stomach and intestine,‖ J. Clin. Biochem. Nutr, 2011; 48(2): 107–111.

65. J. L. Wallace, W. McKnight, B. K. Reuter, and N. Vergnolle, ―NSAID-Induced gastric

damage in rats: Requirement for inhibition of both cyclooxygenase 1 and 2,‖

Gastroenterology, 2000; 119(3): 706–714.

66. [Hayllar and I. Bjarnason, ―NSAIDs, Cox-2 inhibitors, and the gut,‖ Lancet, 1995;

346(8982): 1105–1106.

67. B. J. R. Whittle, ―Gastrointestinal effects of nonsteroidal anti-inflammatory drugs,‖

Fundam. Clin. Pharmacol., 2003; 17(3): 301–313.

68. J. L. Wallace, ―Prostaglandins, NSAIDs, and Gastric Mucosal Protection: Why Doesn’t

the Stomach Digest Itself?,‖ Physiol. Rev., 2008; 88(4): 1547–1565.

69. C. Yu, X. T. Mei, Y. P. Zheng, and D. H. Xu, ―Gastroprotective effect of taurine zinc

solid dispersions against absolute ethanol-induced gastric lesions is mediated by

enhancement of antioxidant activity and endogenous PGE2production and attenuation of

NO production,‖ Eur. J. Pharmacol., 2014; 740: 329–336.

70. C. Bode and J. C. Bode, ―Alcohol’s role in gastrointestinal tract disorders.,‖ Alcohol

Health Res. World, 1997; 21(1): 76–83.

71. A. R. M. S. Brito et al., ―Antiulcerogenic action of ethanolic extract of the resin from

Virola surinamensis Warb. (Myristicaceae),‖ J. Ethnopharmacol., 2008; 122(2): 406–409.

72. A. B. Albino de Almeida et al., ―Antiulcerogenic effect and cytotoxic activity of semi-

synthetic crotonin obtained from Croton cajucara Benth.,‖ Eur. J. Pharmacol., 2003;

472(3): 205–212.

73. M. Guslandi, ―Effects of ethanol on the gastric mucosa,‖ Dig Dis, 1987; 5(1): 21–32.

74. G. G. Ortiz, G. Nisticò, B. Poeggeler, E. Sewerynek, D. Melchiorri, and R. J. Reiter, ―

Suppressive effect of melatonin administration on ethanol-induced gastroduodenal injury

in rats in vivo ,‖ Br. J. Pharmacol., 2006; 121(2): 264–270.

75. A. . Guseva et al., ―Antiulcer effects of amylin: a review,‖ Pathophysiology, 2003; 11(1):

1–6.

76. C. La Casa, I. Villegas, C. Alarcón De La Lastra, V. Motilva, and M. J. Martín Calero,

―Evidence for protective and antioxidant properties of rutin, a natural flavone, against

ethanol induced gastric lesions,‖ J. Ethnopharmacol., 2000; 71(1–2): 45–53.

77. P. Arda-Pirincci, S. Bolkent, and R. Yanardag, ―The role of zinc sulfate and

www.wjpps.com Vol 8, Issue 8, 2019.

1633

Prabhavalkar et al. World Journal of Pharmacy and Pharmaceutical Sciences

metallothionein in protection against ethanol-induced gastric damage in rats,‖ Dig. Dis.

Sci., 2006; 51(12): 2353–2360.

78. M. G. Repetto and S. F. Llesuy, ―Antioxidant properties of natural compounds used in

popular medicine for gastric ulcers,‖ Brazilian J. Med. Biol. Res., 2002; 35(5): 523–534.

79. S. H. Wong, C. H. Cho, and C. W. Ogle, ―CALCIUM AND ETHANOL-INDUCED

GASTRIC MUCOSAL DAMAGE IN RATS Department of Pharmacology , Faculty of

Medicine , University of Hong Kong , Hong Kong,‖ Pharmacol. Res., 1991; 23(1):

71–79.

80. E. Marhuenda, M. J. Martin, and C. D. La Alarcon Lastra, ―Antiulcerogenic activity of

aescine in different experimental models,‖ Phyther. Res., 1993; 7(1): 13–16.

81. P. J. Oates and J. P. Hakkinen, ―Studies on the mechanism of ethanol-induced gastric

damage in rats,‖ Gastroenterology, 1988; 94(1): 10–21.

82. K. Takagi, S. Okabe, and R. Saziki, ―A new method for the production of chronic gastric

ulcer in rats and the effect of several drugs on its healing.,‖ Jpn. J. Pharmacol., 1969;

19(3): 418–26.

83. S. Okabe and C. J. Pfeiffer, ―Chronicity of acetic acid ulcer in the rat stomach,‖ Am. J.

Dig. Dis., 1972; 17(7): 619–629.

84. S. Okabe, J. L. A. Roth, and C. J. Pfeiffer, ―A method for experimental, penetrating

gastric and duodenal ulcers in rats - Observations on normal healing,‖ Am. J. Dig. Dis.,

1971; 16(3): 277–284.

85. S. Okabe and K. Amagase, ―An Overview of Acetic Acid Ulcer Models<br>&mdash;The

History and State of the Art of Peptic Ulcer Research&mdash;,‖ Biol. Pharm. Bull.

Pharm. Bull., 2005; 28(8): 1321–1341.

86. S. Singh and D. K. Majumdar, ―Evaluation of the gastric antiulcer activity of fixed oil of

Ocimum sanctum (Holy Basil),‖ J. Ethnopharmacol., 1999; 65(1): 13–19.

87. S. Singh, ―Evaluation of gastric anti-ulcer activity of fixed oil of Ocimum basilicum Linn.

and its possible mechanism of action,‖ Indian J. Exp. Biol., 1999; 37(3): 253–257.

88. L. E. Sander et al., ―Selective expression of histamine receptors H1R, H2R, and H4R, but

not H3R, in the human intestinal tract,‖ Gut, 2006; 55(4): 498–504.

89. C. H. I. H. I. N. Cho and C. J. Pfeiffer, ―Gastrointestinal Ulceration in the Guinea Pig in

Response to Dimaprit , Histamine , and Ha- and H2-Blocking Agents,‖ Dig. Dis. Sci.,

1981; 26(4): 306–311.

90. H. Tamaki, Y. Onoda, and T. Kashida, ―Gastric Secretion and Duodenal Ulcer Formation

Induced By Cysteamine in Rats,‖ Jpn. J. Pharmacol., 2008; 28(4): 647–649.

www.wjpps.com Vol 8, Issue 8, 2019.

1634

Prabhavalkar et al. World Journal of Pharmacy and Pharmaceutical Sciences

91. D. Stiel, D. J. Murray, and T. J. Peters, ―Mucosal Enzyme Activities, with Special

Reference to Enzymes Implicated in Bicarbonate Secretion, in the Duodenum of Rats

with Cysteamine-Induced Ulcers,‖ Clin. Sci., 2015; 64(3): 341–347.

92. S. Szabo, X. Deng, T. Khomenko, G. D. McLaren, H. Ishikawa, and G. J. Anderson,

―Role of iron in the pathogenesis of cysteamine-induced duodenal ulceration in rats,‖ Am.

J. Physiol. Liver Physiol., 2009; 296(6): G1277–G1286.

93. F. A. Carone and R. E. Cooke, ―Effect of Potassium Deficiency on Gastric Secretion in

the Rat,‖ Am. J. Physiol. Content, 2017; 172(3): 684–688.