Next Article in Journal
Wheat Germ as Partial or Total Substitutive of Lean Meat in Low-Fat Cooked Sausages
Previous Article in Journal
Effect of Antioxidants on the Gut Microbiome Profile and Brain Functions: A Review of Randomized Controlled Trial Studies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

An Overview of the Current Scientific Evidence on the Biological Properties of Abelmoschus esculentus (L.) Moench (Okra)

1
Department of Food and Health Science, Technological and Higher Education Institute of Hong Kong, Hong Kong, China
2
Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong, China
3
University Safety Office, The Chinese University of Hong Kong, Hong Kong, China
*
Author to whom correspondence should be addressed.
Foods 2025, 14(2), 177; https://doi.org/10.3390/foods14020177
Submission received: 30 November 2024 / Revised: 28 December 2024 / Accepted: 4 January 2025 / Published: 8 January 2025
(This article belongs to the Special Issue Functional Food and Safety Evaluation: Second Edition)

Abstract

:
Abelmoschus esculentus (L.) Moench, commonly known as okra or lady’s finger, is an annual flowering plant belonging to the Malvaceae family. Okra is a native plant in Africa as well as a traditional medicine in Africa and India for treating different diseases and conditions. Today, okra is widely consumed as a vegetable and is increasingly recognized as a superfood due to its rich nutritional profile and potential pharmacological benefits. Research indicates that okra exhibits a range of biological activities, including antidiabetic, antihyperlipidemic, antifatigue, vasoprotective, hepatoprotective, antitumor, anti-inflammatory, and antimicrobial effects. Despite its promising therapeutic potential, research on the active compounds in okra and evaluating efficacy in clinical settings remains limited. This review aims to consolidate existing scientific knowledge on the biological and pharmacological properties of okra, thereby encouraging further investigation into its health benefits. Ultimately, this could pave the way for the development of functional foods or health supplements that leverage okra as a key ingredient to prevent chronic diseases and enhance overall health outcomes.

Graphical Abstract

1. Introduction

The global prevalence of chronic diseases is on the rise. A multinational survey study has demonstrated a significant increase in the percentage of teenagers aged 11 to 17 years with four or more chronic disease risk factors, soaring by approximately 30% from 2003–2007 to 2013–2017 [1]. Consequently, there has been a heightened demand for functional foods as the public becomes increasingly conscious of their consumption. Beyond providing essential nutrition, these foods can play a vital role in mitigating the development of chronic diseases and enhancing overall well-being [2,3]. Vegetables like moringa and turmeric are widely recognized as functional foods with a diverse range of pharmacological effects, including enhancing fertility and alleviating various chronic conditions such as cardiovascular diseases, diabetes, obesity, inflammatory bowel disease (IBD), acne, asthma, eczema, and allergies, supported by both clinical and preclinical studies [4,5]. Unlike moringa and turmeric, okra is an emerging functional food that has been known for its antidiabetic, antihyperlipidemic, and antifatigue effects. However, currently, there is no evidence to support the use of okra in inflammatory diseases such as IBD, asthma, and mastitis, even though okra possesses anti-inflammatory effects and antioxidative effects [6]. Therefore, further research is essential to uncover the full extent of okra’s biological activity.
The scientific name of okra is Abelmoschus esculentus (L.) Moench (Figure 1 and Figure 2). It is also known as lady’s finger, as well as gumbo. This perennial flowering plant belongs to the family of Malvaceae. Its origin is still under debate. The majority believes that it is from Africa, probably Ethiopia (Sudan), instead of India [7]. Today, okra is cultivated worldwide in the tropics, subtropics, and warm regions like South Asia (China, India, etc.), Europe, and Australia, as well as the Americas (the United States and Brazil), and is extensively consumed as a vegetable globally, especially in Africa [7,8]. Meanwhile, okra is recorded as a traditional medicine in India and Africa, for instance, in Ghana [9,10]. Traditionally, the okra pod is used to treat sexually transmitted diseases (gonorrhea and syphilis), urinary diseases (ardor urine and dysuria), dysentery, muscle spasms, catarrh, fever, diarrhea, constipation, anemia, dermal disease (pruritus), and even as a cosmetic product (lotion). It has also been used as a cordial, sudorific (to promote sweating), and aphrodisiac, with historical records suggesting its efficacy in preventing scurvy [8,11,12,13,14,15,16,17,18,19].
Okra, recognized as a superfood (functional food), is increasingly gaining recognition for its high nutritional value and diverse therapeutic effects, which are supported by scientific evidence [20,21]. Furthermore, okra’s easy availability in the market is a notable advantage. Due to its abundant cultivation, okra remains affordably priced, making it a desirable functional food option [6]. Although the consumption of okra is becoming popular, currently, there is no review summarizing both clinical and preclinical data of okra supporting its usage in different diseases. This review aims to provide an overview of the currently available scientific information on okra in both preclinical and clinical studies to draw attention from researchers to studying undiscovered biological activities of okra, its active components, and the investigating the efficacy of okra in different diseases in clinical trials. Ultimately, this could pave the way for the development of functional foods or health supplements that leverage okra as a key ingredient to prevent chronic diseases and enhance overall health outcomes.

2. Active Ingredients and Nutrition Value in Okra

Okra stands out as a functional food due to its exceptional nutritional profile. It is rich in essential nutrients, boasting a significant carbohydrate content (7 g per 100 g serving), protein (2 g per 100 g serving), dietary fiber (3.2 g per 100 g serving), an array of minerals (abundant in potassium, calcium, phosphorus, and manganese), and vitamins, while being low in fat (0.1 g per 100 g serving) [22,23] (Table 1).
A total of 35 active components have been isolated from various parts of okra, primarily from the pods and seeds. Among these components, the majority are flavonoids (16 in total) and polysaccharides (12 in total) [24,25,26,27,28,29,30]. These active components, along with their biological effects and sources of isolation, are summarized in Table 2.

3. Biological Activities of Okra

Okra has been reported to possess a wide range of biological activities, including antidiabetic, antihyperlipidemic, antifatigue, antitumor, and immunomodulating properties [46,57,58,59]. This section will provide a comprehensive overview of these biological activities and their underlying mechanisms (Table 3 and Table 4).

3.1. Antidiabetic Effect

Restoration of β-cell function, improvement in insulin resistance or sensitivity through suppression of peroxisome proliferator-activated receptor (PPAR)-γ, and enhancement of antioxidant enzymes, as well as scavenging of free radicals, inhibition of glucose absorption, retardation of carbohydrate digestion, reducing blood glucose levels, and improving glucose tolerance are the crucial working principles underlying the antihyperglycemic effect of Abelmoschus esculentus (L.) Moench fruit, seeds, and peel [38]. The detailed mechanisms of okra’s antidiabetic effect will be discussed as follows.

3.1.1. Restoration of β-Cell Function

The protective effect of Abelmoschus esculentus (L.) Moench on pancreatic islets, particularly β-cells, has become one of the key targets of recent research. Okra fruit extract has been found to reverse the streptozotocin-induced β-cells damage and prevent free fatty acid-induced apoptosis of β-cells [61,86]. For example, an in vivo study found that administration of okra fruit extract (200 mg/kg) significantly suppressed insulin levels, the homeostasis model assessment of basal insulin resistance (HOMA-IR), as well as blood glucose levels in streptozotocin-induced diabetic rats [61]. These changes might be associated with the increase in the mass of pancreas islets and the number of β-cells in diabetic rats, which was proposed to play a key role in the restoration of β-cells function [61]. Similarly, subfractions of okra fruit also showed improved glycemic control in a high-fat diet and streptozotocin-induced diabetes in rats [60]. Although subfraction 1 (F1: rich in quercetin glucosides, such as isoquercetin and pentacyclic triterpene ester) and subfraction 2 (F2: rich in polysaccharides and carbohydrates) could significantly lower blood glucose levels, HOMA-IR, and glycated hemoglobin (HbA1c), and the effects of F2 are more effective than F1. The preventative effect of okra on β islet damage was related to the antihyperglycemic effect [60], which can be further supported in vitro in the RINm5f cell line with palmitate-induced β-cell apoptosis, which demonstrates that F1 and F2 prevented free fatty acid-induced β-cell apoptosis significantly through the downregulating expression of dipeptidyl peptidase-4 (DPP-4) apoptotic signaling and restoring the expression level of glucagon-like peptide-1 receptor (GLP-1R) [86]. Both F1 and F2 decreased in the sub-G1 stage through the downregulation of the expression of pro-caspase 3 and active-caspase 3, suppressing DPP-4, as well as modulating palmitate-induced signal cascades (the one that causes β-cell apoptosis) via the downregulation of adenosine monophosphate-activated protein kinase (AMPK) and Bax, as well as the upregulation of the mammalian target of rapamycin (mTOR) and phosphoinositide 3-kinase (PI3K). However, the effect of F2 on the downregulation of AMPK and suppression of cascades is more significant than F1 [86].

3.1.2. Improvement in Insulin Resistance/Sensitivity via Suppression of PPARs Genes

Apart from the restoration of β-cell function, okra has also been shown to improve insulin sensitivity through the downregulation of PPARs gene expression. Several studies discovered that okra, particularly its polysaccharides, were antagonists of PPARs, which ameliorated insulin resistance and insulin sensitivity.
An in vivo study showed that the amelioration in insulin resistance/sensitivity in high-fat diet-induced diabetes in rats relied on the effect of okra fruit extract suppressing mRNA levels of PPAR-α and -γ in the pancreas [61]. These findings were aligned with the one in the mice with high-fat diet-induced obesity, which demonstrated that ethanol extract from okra alleviated insulin resistance via the downregulation of mRNA levels of PPAR-α and -γ in the liver (caused by obesity) significantly [25]. Similarly, okra fruit polysaccharide significantly attenuated the expression of PPAR-α, -γ, and -β/δ in adipose tissue in the mice [49].

3.1.3. Enhancement of Antioxidant Enzymes as Well as Scavenging of Free Radicals

Increasing evidence has shown that oxidative stress plays a crucial role in the development of diabetes. Excessive production of free radicals [reactive oxygen species (ROS)/reactive nitrogen species (RNS)] and weakened antioxidant defenses can cause oxidation of macromolecules and cell damage, particularly β-cells [98,99]. Studies found that okra seeds, peel, and fruit possess strong antioxidant activity and enhance antioxidant defense systems in diabetic rats [58,64]. Therefore, the ability of okra to free radical scavenging effects and restoration of the antioxidant enzyme system also plays an essential role in its antidiabetic effects.
A study investigated the in vivo antioxidant activity in okra seeds and peel, which found that okra significantly increased antioxidant enzyme levels, including superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), and reduced glutathione (GSH), as well as attenuated lipid peroxidation in the liver, pancreas, and kidney [58]. Additionally, another study showed that okra fruit also possessed excellent in vivo antioxidant activity (the ferric-reducing ability of plasma assay); it decreased the activity of erythrocyte plasma membrane redox system (PMRS), erythrocyte malondialdehyde (MDA) content (prevent lipid peroxidation), and advanced oxidation protein products (AOPP) (hinder protein oxidation); as well as increased erythrocyte GSH [64].
The okra flower, fruit, leaf, and seed (methanol extracts/enrichment fraction of water extracts) also demonstrated good scavenging free radical in both 2,2-diphenyl-1-picrylhydrazyl (DPPH) and ferric-reducing antioxidant power assays. The results also indicated that there was a positive proportional relationship between phenolic content, flavonoid content, and antioxidative activities [87]. Similarly, another study further indicated that phenolic compounds, including procyanidin B2, procyanidin B1, catechin, epicatechin, quercetin, and rutin (okra seeds do not contain catechin and epicatechin, while pulp does not have quercetin and procyanidin B2) might be the active molecules responsible for antioxidant activity in okra [37]. Moreover, four flavonoid compounds in okra fruit, 5,7,3′,4′-tetrahydroxy-4″-O-methyl flavonol-3-O-β-D-glucopyranoside, 5,7,3′,4′-tetrahydroxy flavonol-3-O-[β-D-glucopyranosyl-(1→6)]-β-D-glucopyranoside, isoquercitrin, and quercetin 3-O-gentiobioside, showed high antioxidant activity [25,27]. Last but not least, a pectic polysaccharide WOP-2, which is a rhamnogalacturonan I with type-II arabinogalactan side-chains (580KDA composed with monosaccharides Rha (21.4%), GalA (34.9%), Gal (29.6%), GlcA (4.5%), Glc (5.9%), and Ara (3.7%) was identified. It had strong free radical scavenging activity in a dose-dependent manner and was shown to boost antioxidant enzyme (SOD) levels in diabetic mice, which prevented damage in β-cells caused by peroxidation and helped restore insulin levels [40].
Okra was also found to possess a therapeutic effect on gestational diabetes in rats through suppressing oxidative stress and insulin resistance, which is achieved by restoration of antioxidant defense, such as SOD, GPx, GSH, and CAT, in the liver and pancreas [65].
The antioxidant activity in okra and its active ingredients, epically isoquercitrin and quercetin-3-O-gentiobiose, not only contributed to its antidiabetic effect but was also found to be attributed to its hepatoprotective effect, antifatigue effect, vasoprotective effect, and neuroprotective effect (for instance, reducing the risk of developing Alzheimer’s disease) [31,33,73,92].

3.1.4. Inhibition of Rate of Carbohydrate Digestion and Glucose Absorption

The antidiabetic effects of okra were also found, depending on the retardation of the rate of starch digestion and glucose absorption. In vitro studies have shown that aqueous extract from the okra peel and seeds inhibited α-glucosidase and α-amylase activities appreciably in a dose-dependent manner [38,88]. The effect of okra peel was more potent than its seeds [88]. In unripe seeds, oligomeric proanthocyanidins, which are composed of epigallocatechin and catechin extension units, were inhibitors of α-glucosidase and α-amylase [38]. However, another study found that rutin and quercetin 3-gentiobioside are also active compounds responsible for suppressing carbohydrate digestion [32].
In an in vivo study, the water-soluble fraction (dietary fiber) of okra fruit was able to reduce the intestinal absorption of glucose significantly in fasting rats. Interestingly, when okra and metformin were fed to diabetic rats, the effect of metformin on intestinal absorption of glucose vanished [54]. The effect of okra reduction in intestinal absorption of glucose was found to be concentration-dependent in an in vitro study [89]. These results suggested that okra is useful for postprandial glucose control.

3.1.5. Hypoglycemia and Improving Glucose Tolerance

The antidiabetic effects of okra also relied on the fact that it lowered fasting blood glucose levels and improved glucose tolerance. Okra fruit, seeds, and peel were found to lower blood glucose levels and HbA1c considerably in different models of diabetic rats, which were either induced by alloxan or streptozotocin [66,67].
Okra polysaccharides from its fruit were demonstrated to reduce blood glucose levels and improve glucose tolerance in mice with high-fat diet-induced obesity [49]. Isoquercitrin and quercetin 3-O-gentibiosidein in okra were responsible for the hypoglycemic effect of okra in high-fat diet-induced obesity in mice [25]. Meanwhile, a polysaccharide, rhamnogalacturonan, was identified and responsible for lowering blood glucose levels and improving glucose tolerance in diabetic mice [28].

3.1.6. Prevention of Diabetic Nephropathy

An in vitro study demonstrated that fractional extract from okra fruit, especially F1 and F2, could improve diabetic nephropathy through inhibition of diabetic renal epithelial to mesenchymal transition (EMT), and the regulation of DPP-4 and GLP-1R, as well as reducing oxidative stress and renal fibrosis in the HK-2 cell line [90]. The same study showed that F1 was rich in pentacyclic triterpene and flavonoid glycosides, such as quercetin glycosides. In contrast, F2 was mainly composed of polysaccharides of uronic acid, galactose, glucose, and myo-inositol [90].
The effect of F1 and F2 on relieving diabetic nephropathy was found to be achieved by modifying the signal involved in developing EMT. F1 significantly suppressed high glucose-induced increased levels of vimentin, angiotensin II receptor-1 (AT-1), and transforming growth factor β1 (TGF-β1), as well as DPP-4 activity and upregulated high attenuated levels of cadherin. Similarly, F2 has almost the same effect as F1 except for no significant change in the level of TGF-β1 [90]. Similarly, in vivo studies also found that both F1 and F2 could ameliorate diabetic nephropathy, where the effect of F2 was much more specific to the kidney. Even though both fractions could improve renal function and alleviate renal fibrosis, only F2 was able to reverse the DPP-4 and GLP-1R levels as well as attenuate oxidative stress in the kidney [69].

3.2. Antifatigue and Vasoprotective Effect

Recent studies suggested that okra possesses antifatigue properties, which might enhance exercise tolerance by reducing the accumulation of metabolic by-products, increasing energy reserves, and regulating energy metabolism. Additionally, okra has been shown to mitigate oxidative stress by modulating enzymatic activities involved in energy metabolism and the excitation–contraction coupling process.
An in vivo study showed that okra ethanol extract and its polysaccharides could alleviate fatigue in mice. Okra polysaccharides and ethanol extract enhanced exercise endurance in a dose-dependent manner via lowering blood lactic acid (BLA), as well as serum urea nitrogen (SUN), and increasing the hepatic glycogen (HG) notably, in which the effect of the polysaccharides was much better than the extract. The polysaccharides could also improve kidney function in mice with kidney yang deficiency [57]. Another study also found that two okra polysaccharide fractions, AEP-1 and AEP-2, possessed antifatigue activity in accordance with the previous study [71]. The same study also found that okra polysaccharides could increase muscle glycogen (MG), and the effect of AEP-1 was stronger than AEP-2. Regarding the mechanistic pathways of AEP-1 and AEP-2, their effects were related to the enhancement of the removal of BLA by decreasing the content of lactate dehydrogenase (LDH), decreasing creatine kinase (CK) in blood and improving energy metabolism via increasing succinate dehydrogenase (SDH), adenosine 5′-triphosphatase (ATPase), and energy content (ATP) in the serum, liver, and muscle in three different states (resting, dynamic, and recovery states) [71].
Other research also found that the okra seed in the pod was the part responsible for the antifatigue effect of okra, and the result aligned with the aforementioned studies. This study revealed that okra seeds significantly improved antioxidant defense enzymes (SOD and GSH-Px) and scavenge free radicals. The flavonoid compounds in okra seeds, particularly isoquercitrin and quercetin 3-O-gentiobiose, were likely to be responsible for their antifatigue activity because of their antioxidant activity [31]. Another investigation found that quercetin 3-O-gentiobiose relieved fatigue significantly by increasing gastrocnemius muscle glycogen [33].
In addition, quercetin 3-O-gentiobiose also possesses a vasoprotective effect by preventing exhaustive exercise-induced vascular endothelial dysfunction by improving aortic morphology, preventing oxidative stress damage, and suppressing inflammation. Quercetin 3-O-gentiobiose reduced the number of foam cells and aorta thickness, as well as intima–media thickness in the exhaustive swimming rats. This was due to its high antioxidant enzyme activities, its effect on decreasing inflammatory cytokines monocyte chemoattractant protein-1 (MCP-1), tumor necrosis factor alpha (TNF-α), and interleukin-6 (IL-6) significantly, and dose-dependently, its modulating effect on the LOX-1/NF-κB signaling pathway, which remarkably reduced mRNA expression and protein expression of lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1), intercellular adhesion molecule-1 (ICAM-1), and nuclear transcription factor-κB p65 (NF-κB p65) expressions in a dose-dependent manner [33].

3.3. Hepatoprotective Activity

A few studies found that okra pods and roots had a hepatoprotective effect via their excellent antioxidant activity and their ability to boost the enzymatic antioxidant defense system. In vivo and in vitro studies showed that okra roots reversed the hepatic damage induced by carbon tetrachloride (CCl4) and restored its function in HepG2 cells and rats’ livers, as okra significantly prevented the leakage of alanine transaminase (ALT), aspartate transaminase (AST), and alkaline phosphatase (ALP), lowered the level of total bilirubin, increased serum albumin, prevented accumulation of triglyceride in the liver, and improved histopathology of the liver, as well as reduced levels of TNF-α in the liver (preventing immune-mediated liver injury) [72].
Another in vivo study in rats also demonstrated the pre-treatment of rats with ethanol extract from okra pods exhibited a hepatoprotective effect, which prevented the elevation of some liver health-related biomarkers, such as serum glutamate oxaloacetate transaminase (GOT), serum glutamate pyruvate transaminase (GPT), ALP, and gamma-glutamyltransferase (GGT), as well as an increase in cholesterol and triglycerides. But unlike okra root, okra ethanol extract could not lower the level of bilirubin. It could also suppress liver inflammation and increase hepatic total protein as well as non-protein sulfhydryls [73].
Quercetin 3-O-gentiobiose and quercetin 3-O-glucosyl (1→6) glucoside isolated from okra seeds were the active compounds in okra pods for the hepatoprotective effect. These flavonoids were shown to ameliorate hepatic damage mediated by CCl4 [26]. These compounds can serve as antioxidants to scavenge ROS and upregulate endogenous antioxidant enzyme levels (CAT, GSH, and SOD) to prevent CCl4-induced oxidative stress and CCl4-induced lipid oxidative stress, as evidenced by a decrease in the level of MDA and an increase in CAT, GSH, and SOD levels [72,73].

3.4. Antihyperlipidemic Activity

Investigations found that different parts of okra (peel, seeds, and pods) could alter dyslipidemia in mice and rats, and some improvements are even comparable to the effect of the lipid-lowering medication simvastatin [60,61,67,74]. Dyslipidemia is a well-known risk factor for developing obesity that could lead to diabetes and cardiovascular disease [100,101]. Therefore, okra may be used as a dietary source for preventing these diseases. In vivo studies showed that okra seed and peel powder reversed a high-fat diet or a high-fat diet plus streptozotocin-induced abnormal lipid profile (total triglycerides, total cholesterol, and low-density lipoprotein) in rats [61,67]. Similarly, subfractions of okra extract F1 (rich in flavonoid and quercetin glycosides) and different okra extracts also suppressed high-fat diet plus streptozotocin-induced as well as tyloxapol-induced hyperlipidemia in rats and mice [60]. However, ethanol extract from okra alleviated high-fat diet-induced hepatic steatosis and macrovesicular steatosis in C57BL/6 mice, in which isoquercitrin and quercetin 3-O-gentiobioside were found to be the active compounds [25]. Apart from improving lipid profit, okra polysaccharides could also reduce the size of white adipocytes in high-fat diet-induced obesity in C57BL/6 mice [49].
The underlying mechanism of okra and its active components in antihyperlipidemic activity was revealed by these studies, showing that okra extract reduced the transcription of lipogenesis and cholesterol metabolism-related genes as well as nuclear receptor transcription factors, such as PPARs, Liver X receptors (LXR), LXR, and PPARs target genes, and adipocyte protein 2 (aP2) [25,61]. Additionally, okra polysaccharides inhibited the gene expression of LXR α/β in liver and adipose tissue, ATP-binding cassette transporter G1 (ABCG1), Apolipoprotein E (ApoE), cytochrome P450 7A1 (CYP7A1), and lipoprotein lipase (LPL), as well as PPARs (γ, α, and β/δ) in adipose tissue and mitochondrial uncoupling protein 2 (UCP2) [49]. Okra also promoted the fecal excretion of bile acid via the upregulation of the transcription of CYP7A1, while the downregulation of the transcription of the sterol regulatory element-binding protein 1c (SREBP1c) and fatty acid synthase (FAS) were also accounted for okra’s hypolipidemic activity [75].

3.5. Antitumor Activity

Various components in okra and its compounds have the ability to impede the advancement of cancer cells by inducing apoptosis, inhibiting proliferation, and causing cell cycle arrest. Additionally, the immunomodulatory properties of different components in okra may also play a role in its antitumor activity.
The lectin isolated from okra seeds showed antiproliferation and apoptosis in human breast cancer cells (MCF7) but not in skin fibroblast (CCD-1059 sk). The selective antitumor activity (cytotoxic) of lectin on MCF7 relied on its interaction with carbohydrates on the cell surface [29]. The underlying mechanism of lectin-induced apoptosis in MCF7 was mediated by the upregulation of apoptosis-related gene expression, including caspase-3 and -9, as well as p21 and the downregulation of Bcl-2 transcription, which increased the ratio of Bax to Bcl-2 r. However, no alteration was found in the survivin, apoptosis-inducing factor (AIF) and endonuclease G gene [29].
Pectic rhamnogalacturonan-I (RG-I) extracted from okra pods retarded proliferation and induced apoptosis in B16F10 Melanoma cells in the tPs culture plate and the one cultured in anti-adhesive polyHEMA substratum (3D). This was mediated by arresting the cell cycle (increased cells in the G2/M phase dramatically) as well as decreasing the protein expressions of cadherins and α5 integrin, as well as upregulating galectin-3 (Gal-3) [41].
Polysaccharides isolated from different parts of okra possessed immunomodulatory activity by promoting the maturation of dendritic cells (DCs cells), modulating cytokine secretion, and activating macrophages [76,91]. For instance, polysaccharide extract from okra fruit stimulated primary cell-rat bone marrow hematopoietic cells derived immature dendritic cells (BMHC-imDCs), which was proved by the upregulation of major histocompatibility complex (MHC) class II and Cluster of differentiation (CD) 80/86 and decreasing endocytosis activity dose-dependently. The activation of DCs increased the secretion of IL-12/ interferon gamma (IFN-γ) and decreased the secretion of IL-10. This indicated okra could trigger a type 1 T helper (TH1) response [91]. Another study showed that a water-soluble polysaccharide (OFPS11) from okra flowers could suppress the proliferation of HepG-2 cells with the aid of the immunomodulatory effect of OFPS11 on the RAW264.7 cell, which is primarily composed of galactose and rhamnose in 2.23:1 ratio [46]. The immunomodulatory effect of OFPS11 significantly increased the phagocytic activity in the macrophages in a dose-dependent manner, as well as its production of nitric oxide (NO), TNF-α, and IL-1β. These increases were caused by the upregulation of mRNA and protein expressions of inducible nitric oxide synthase (iNOS), TNF-α, and the activation of the NF-κB signaling pathway [46]. The research evaluated the immunomodulatory effect of okra polysaccharides [RPS, composed of galactose (40%), rhamnose (29.9%), galacturonic acid (13.9%), and glucuronic acid (9.4%)] and its purified fractions RPS-1 [principally consisted of galactose (33.1%), galacturonic (31.9%), and rhamnose (20%)], RPS-2 [mainly consisted of galactose (35.5%), galacturonic (31.4%), and rhamnose (20.3%)] and RPS-3 [primarily composed of galacturonic (25.1%), galactose (21.6%), galacturonic (17.8%), glucose (14.9), and rhamnose (1.8)] in vitro in RAW264.7 and RPS2 in vivo in BALB/c mice. The RPSs showed the same result as the OFPS11 in increasing NO secretion through the upregulation of iNOS in the in vitro study. The PRSs also increased the secretion of cytokines, such as TNF-α (for all RPSs), IFN-γ (for RPS-1), and IL-10 (for all RPSs), while RPS-2 significantly increased splenocyte proliferation and thymus and spleen index in vivo [76].

3.6. Neuroprotective Effect

Oxidative stress and psychological stress could cause the development of neurodegenerative diseases, such as Alzheimer’s disease (AD) [102,103]. Aqueous and methanol extract from okra seeds was found to have anti-stress and nootropic (attenuation of scopolamine-induced cognitive impairment) effects in an in vivo study (elevated plus maze task and forced swimming test (FST) was employed for anti-stress, while passive avoidance was used to determine nootropic effect) as well as demonstrated antioxidant effects [77]. Furthermore, another in vivo study also showed okra seeds and leaves have fair antidepressant activity (FST and tail suspension test) dose-dependently [78]. As a result, okra may mitigate neurodegenerative diseases and their symptoms.
An in vivo study revealed that pre-treatment of ethanol extract from okra and its flavonoid compounds (quercetin and rutin) had a neuroprotective effect and improved cognitive impairment in dexamethasone-treated ICR male mice [30]. The same study showed the pre-treatment significantly improved the performance of mice in the Morris water maze test, mitigated the morphological damage in the cornu ammonis 3 (CA3) region of the hippocampus, and reversed the decreased number of CA3 hippocampal neurons, as well as increased the average number of Brdu-positive cells per section in the histology. It also increased the expression of NR (NMDA-receptor) 2A/B protein remarkably. This indicated that pre-treatment of okra could reverse the damage in the hippocampus through enhancement of cell proliferation in the dentate gyrus (in the CA3 region) and recover the number of N-methyl-D-aspartate (NMDA) receptors [30]. Okra was once again proven to be beneficial to neurodegenerative disease. Similarly, an in vitro study revealed that ethanolic extract from okra could reduce the risk of development of AD or other neurodegenerative diseases, especially in people who express the H63D variant in the hemochromatosis (HFE) gene in the neuroblastoma SH-SY5Y cell line [92]. The same study reported that okra significantly attenuated oxidative stress (lower protein carbonyl, H2O2, and intracellular ROS), suppressed tau phosphorylation at serine 199, 202, and 396 in a dose-dependent manner, and inhibited the activity of glycogen synthase kinase-3 beta (GSKk-3β) by increasing serine 9. The mechanism behind this was believed to be related to the decrease in the intercellular iron level.

3.7. Skin Protective Effect

Okra has a historical tradition of use in cosmetics. Presently, okra seed extract has been utilized as the active ingredient of a commercial cosmetic product. An in vivo study indicated that okra significantly improved skin elasticity, firmness, texture, and density, as well as mitigated wrinkles, which was related to the protective effect of okra seeds on fibroblast growth factor-2 (FGF-2) stimulating cell proliferation and glycosaminoglycans (GAG) synthesis [80]. Another study demonstrated that okra had the potential as sunscreen, as flavonoids enrichment of okra could alleviate ultraviolet radiation-B induced oxidative stress and cytotoxicity in human dermal fibroblast adult cells (HDFs) by its good antioxidant effect in an in vitro study and intracellular ROS assay as well as its promoting effect on enzymatic antioxidant defense [SOD, CAT, GPx, and glutathione reductase (GR)] probably via reducing protein expressions of nuclear factor E2-related factor-2 (Nrf2) and hemeoxygenase-1 (HO-1) significantly in a dose-dependent manner [93].

3.8. Relief Temporomandibular Joint (TMJ) Inflammatory Hypernociception Through Its Anti-Inflammatory, Antinociceptive, and Analgesic Activity

An in vivo study found that methanolic and water extracts of okra peel possess great anti-inflammatory, analgesic, and antinociceptive activities [81]. Another study also showed that lectin (20.0 kDa) extracted from okra seeds exhibited good antinociceptive and anti-inflammatory activities [52]. Due to the discovery of antinociceptive, anti-inflammatory, and analgesic activities of lectin, recently, the efficacy of lectin from okra seeds and its involved pathways were examined in TMJ inflammatory hypernociception in rats.
In the zymosan-induced TMJ inflammatory hypernociception in rats, pre-treatment with okra lectin could lower leukocyte cell, myeloperoxidase (MPO) activity, and Evans blue dye extravasation in the synovial lavage, as well as decrease inflammatory cell influx in synovial membrane significantly. It could also lower the mechanical hypernociception in rats (less head withdrawal) as well as decrease the cytokines levels in TMJ tissue and trigeminal ganglion, including IL-1β and TNF-α, which contribute to inflammation and nociception [82]. On the other hand, okra lectin also demonstrated similar results in the formalin-induced TMJ inflammatory hypernociception model [83].
The possible molecular mechanisms of okra lectin were elucidated by these studies. Its effects were found to be mediated by the HO-1 pathway (increase HO-1 expression) but not iNOS, as well as the activation of central opioid receptors (δ and κ but not µ) [82,83].

3.9. Anti-Gastric Ulcer Effect of Okra via Its Gastroprotective Effect and Anti-Adhesive Effect of Helicobacter pylori on the Gastric Epithelial Cells

Recently, an in vivo study reported that pre-treatment with okra demonstrated a strong gastroprotective effect on the ethanol-induced model, which could improve the histology of gastric mucosa significantly (edema, hemorrhage, and inflammation scores), decrease oxidative stress (lower MDA and retention of GSH), and increase cell proliferation in the healing area [84].
Several studies found that pre-treatment with okra fruit extract, for instance, as aqueous extract with human gastric epithelia AGS cells, possessed an anti-adhesive effect on Helicobacter pylori (H. pylori), in which some of the active compounds/molecules were identified. An in situ study stated that crude polysaccharides with a rhamnogalactan backbone have strong anti-adhesive activity towards H. pylori. This effect is due to its acid subfraction of polysaccharide (AF-III with a galacturonans backbone consisting of uronic acid clusters and glucuronic acid content) and glycoprotein fraction [48]. Another study further identified that the responsible polymer in the crude polysaccharide for the anti-adhesive effect on H. pylori was acetylated rhamnogalacturonan-I polymers [50]. The mechanism of the anti-adhesive effect of okra on H. pylori was agreed to be the non-specific interaction between compounds/molecules of okra, like polysaccharides, and binding factors/sites of H. pylori, such as SabA, Laminin, lactoferrin, BabA, HpA, and fibronectin (interaction with which binding factor is unknown) [50,94]. Moreover, it is suspected that the charge of the molecules might influence the non-specific interaction [94]. Furthermore, the acetylation/esterification of rhamnogalacturonan-I polymers was necessary for its anti-adhesive effect on H. pylori [50]. Interestingly, a study found that the anti-adhesive effect of okra on H. pylori with outer membrane protein Q genotype 1 (HopQ type 1) was better than the one with either both HopQ type 1 and 2 or HopQ type 2; it also worked well on H. pylori with cytotoxin-associated gene A (CagA) [95]. Apart from the anti-adhesive effect on H. pylori, it has also been demonstrated that the methanolic extract from okra possesses bacteriostatic and bactericidal effects against clinical isolates of H. pylori.
It is well known that gastric ulcers can be caused by alcoholic consumption and infection with H. pylori. The ability of okra to prevent alcohol-induced gastric injury and the gastric attachment of H. pylori makes okra a new potential strategy for the amelioration of gastric ulcers. This is because the effectiveness of first-line treatment of H. pylori-induced gastric ulcers utilizing antibiotics is usually low due to poor bioavailability to the inner layers of gastric mucosa and the emergence of antibiotic resistance [104]. However, further investigation is required to validate the efficacy of okra in gastric ulcers.

3.10. Antimicrobial Activity

Various research studies found that okra exhibits antibacterial properties and an antifungal effect. Specifically, palmitic and stearic acids were the active compounds responsible for its antimicrobial effects [24,96].
An in vitro study showed that lyophilized and freshwater extracts from the okra pods significantly inhibited bacterial growth, including Rhodococcus opacus, Mycobacterium sp., M. aurum, Staphylococcus aureus, and Xanthobacter Py2, as evidenced by minimum inhibitory concentration (MIC) and disk diffusion [24]. The same study revealed that okra extracts suppressed the cell viability of these bacterial strains and that the antibacterial effect was not related to the alteration of bacterial protein (catalase) and denaturation of DNA. Furthermore, it revealed that the polar lipids fraction of okra (rich in palmitic acid and stearic acid) was responsible for its antibacterial effect. Another in vitro study showed that methanolic extract from okra pods significantly inhibited the growth of different clinical isolates of H. pylori and had a potent bactericidal effect on H. pylori BAA009, H. pylori BAA026, and H. pylori ATCC 43504, but the exact mechanism was not revealed [97]. Similarly, an in vitro study demonstrated that okra seeds significantly inhibited the growth of Listeria monocytogenes, Salmonella enteritidis, and S. typhimurium [96]. The same study also reported that okra possessed significant fungistatic and fungicidal effects on Aspergillus fumigatus and A. ochraceus, and the effects were superior to the positive control, ketoconazole.

4. Clinical Evidence of Okra

In recent years, there have been around 10 clinical studies investigating the efficacy and safety of okra, mainly focusing on glycemic control and lipid profile in patients with type 2 diabetes and diabetic nephropathy; however, some of them showed contradicted results [105,106,107,108,109,110,111,112,113,114] (clinical studies’ findings were summarized in Table 5). For instance, a clinical study showed that 1000 mg powdered okra supplement three times per day for three months could significantly improve glycemic control and hyperlipidemia in diabetic patients in Iran (lowering TG and TC) [113]. In contrast, another study revealed that a 1000 mg powdered okra capsule could remarkably improve glycemic control but not lipid profile in diabetic patients in Iran receiving oral hypoglycemic medication [111]. Similarly, one clinical study supported the administration of two 500 mg okra powder capsules three times per day for eight weeks, which significantly alleviated hyperlipidemia and reduced liver and kidney damage (lowering ALT, AST, and uric acid) in prediabetic patients [105]. Additionally, other studies showed that an 80 mg dried okra extract capsule per day for 10 days did not have a significant effect on renal function and lipid profile in patients with diabetic nephropathy [106,110]. The conflicting results may stem from variations in dosage and duration of the intervention. Despite the inconsistency in findings from clinical studies, meta-analyses have supported the safety of consuming okra, which can notably enhance glycemic control. Additionally, consuming ≤3000 mg/day (powdered okra) has been shown to alleviate hyperlipidemia [115].
A novel formula known as IQP-AE-103, comprising a dehydrated powder of okra pods and inulin, [116] showed a significant effect on reducing body weight and body fat in overweight and moderately obese subjects [114]. This clinical study offers promising evidence for the potential use of okra in managing obesity, warranting further clinical investigations to validate its efficacy.

5. Perspectives

Even though okra is widely consumed as food or folk medicine, the pharmacological research on it is still preliminary. Because most of the studies still examine the effect of crude extract or fraction extract from okra on its pharmacological effect, particularly on its antidiabetic effect, preventing EMT, antifatigue effect, antihyperlipidemic activity, immunomodulatory activities, anti-gastric ulcer effect, and antimicrobial effect, as well as skin protection effect. This might result from the sticky mucilage in okra hindering the isolation of bioactive molecules, or there was insufficient investigation of active components from the okra stem, flower, and leaf [42,117]. Future studies should aim to optimize extraction methods to isolate active compounds, especially polysaccharides. Additionally, more research is needed to investigate compounds isolated from the okra stem, flower, and leaf that may be responsible for the pharmacological effects of okra. For instance, identifying the specific compound responsible for modulating PPARs and improving β-cell apoptosis would be a valuable area for further exploration.
The pharmacological effects of okra have not been well studied, particularly regarding the antifatigue effect, anti-gastric ulcer effect, and antimicrobial effect. More mechanistic studies are needed to understand these effects. For example, currently, the study of the antimicrobial effect of okra mainly focused on its antibiotic activity, it will be worth studying its effects on host response, such as how it controls bacterial infection. In vitro studies showed that enhancing macrophage phagocytosis and intracellular killing of bacteria by nitric oxide and ROS in S. aureus-infected macrophages effectively remove S. aureus infections [118,119]. Hence, future studies could explore the effect of okra in S. aureus-infected macrophages. Additionally, some of the traditionally claimed pharmacological effects of okra, such as anti-scorbutic, anemia, aphrodisiac, cordial, and sudorific, lack scientific support and require further investigation. Although okra demonstrated hyperlipidemic activity, its beneficial effects on cardiovascular disease and non-alcoholic fatty liver disease (NAFLD) remain unknown and warrant examination in future studies. Furthermore, inflammatory diseases like mastitis and IBD share similar pathogenesis involving inflammation, oxidative stress, and compromised epithelial barrier [120,121]. Okra may ameliorate these conditions due to its anti-inflammatory effect and protective effect on epithelial cells and ability to suppress oxidative stress. Thus, investigating the effects of okra on inflammatory diseases in future studies may be worthwhile.
Current clinical evidence on the pharmacological effects of okra is limited, with most clinical studies focusing on okra’s efficacy in improving glycemic control and lipid profile in patients with type 2 diabetes, diabetic nephropathy, or prediabetes. Since okra demonstrated significant effects on alleviating diabetes and hyperlipidemia in clinical trials, future clinical trials may consider investigating the efficacy of okra on CVD, obesity, and NAFLD as these diseases share similar pathogenesis, such as impaired blood glucose and hyperlipidemia and are interconnected [122]. Although okra showed significant improvement in lipid profile and glycemic control in clinical studies (Table 5), it is worth mentioning that these clinical trials are mainly conducted in Iran and suggested daily consumption of ≤3000 mg of okra powder. These results may lack diversity in sociodemographics, particularly race and ethnicity, which might lead to poor generalizability and applicability of trial outcomes in diverse patient groups [123]. Therefore, future clinical studies studying the efficacy of okra in different diseases should involve diverse sociodemographic groups and optimize the daily dose of okra consumption to maximize its beneficial effect.
Apart from the direct consumption of okra to obtain its beneficial effect, there are new supplements and food products that incorporate okra as a functional ingredient, allowing the public to maintain physical well-being. For instance, a formula, IQP-AE-103, composed of dehydrated powder from okra pods and inulin, has been proven effective in controlling weight in obese subjects [114]. Similarly, okra seed flour has been incorporated into rice noodles with tapioca starch, which showed improved glycemic control in healthy individuals [124]. Furthermore, research studies demonstrated that okra polysaccharide and okra pectin have good emulsification performance and stability [125,126]. In addition, okra mucilage was reported to be a good replacement for fat in ice cream [127]. Therefore, there is likely to be an increase in food (potentially cake and salad dressings) incorporating okra as a functional ingredient.
Potential interactions between okra and other standard medications for chronic diseases, particularly diabetes, should be investigated, as a study showed that okra diminished the absorption of metformin in rats [54]. Conversely, a clinical study showed that okra did not have any interaction with common oral hypoglycemic agents, such as metformin, pioglitazone sulfonylurea, and sitagliptin [111]. Understanding these interactions could facilitate the development of functional foods or health supplements that utilize okra as a key ingredient, ultimately aiding in the prevention of chronic diseases and improving overall health outcomes.
In summary, both preclinical and clinical studies support the notion that daily consumption of okra possesses beneficial biological activities for human health. Further studies are encouraged to study active components from different parts of okra, unveil new pharmacological effects (e.g., IBD and mastitis), and evaluate its efficacy in different diseases in clinical settings for the development of functional foods or health supplements aimed at promoting public health and preventing chronic diseases.

Author Contributions

Conceptualization, C.T.-K.K. and S.-W.C.; investigation, C.T.-K.K., Y.-F.N. and H.-T.L.C.; writing original draft preparation, C.T.-K.K. and Y.-F.N.; writing, review and editing, C.T.-K.K., Y.-F.N. and H.-T.L.C.; supervision, S.-W.C.; project administration, S.-W.C. All authors contributed equally to this work. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the Faculty Development Scheme (Research Grants Council, Hong Kong), grant number: UGC/FDS25/M03/21, Research Matching Grant (Research Grants Council, Hong Kong), grant number: RMG/042, and a donation from the Far East Consortium International Limited, grant number: RMG/030a.

Data Availability Statement

No new data were created or analyzed in this review. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

List of Abbreviations

AbbreviationsDefinitions
ABCG1ATP-binding cassette transporter G1
ABTS2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid
AEAbelmoschus esculentus
AIFApoptosis-inducing factor
ALPAlkaline phosphatase
ALTAlanine transaminase
AktProtein kinase B
AMPAdenosine 5′-monophosphate
AMPKAdenosine monophosphate-activated protein kinase
AOPPAdvanced oxidation protein products
aP2Adipocyte protein 2
ApoEApolipoprotein E
ASTAspartate transaminase
AT-1Angiotensin II receptor-1
ATPaseAdenosine 5′-TriPhosphatase
BaxB-cell lymphoma protein 2 associated X
Bcl-2B-cell lymphoma 2
BLABlood lactic acid
BMHC-imDCsRat bone marrow hematopoietic cells derived immature dendritic cells
BrdUBromodeoxyuridine
CA3Cornu Ammonis 3
CATCatalase
CCl4Carbon tetrachloride
CDCluster of differentiation
CKCreatine kinase
CYP7A1Cytochrome P450 7A1
DCs cellDendritic cells
DPP-4Dipeptidyl peptidase-4
DPPH2,2-Diphenyl-1-picrylhydrazyl
EMTEpithelial-mesenchymal transition
FASFatty acid synthase
FGF-2Fibroblast growth factor-2
FRAPFerric reducing ability of plasma
FSTForced swimming test
GAGGlycosaminoglycans
Gal-3Galectin-3
GGTGamma glutamyltransferase
GLP-1RGlucagon like peptide-1 receptor
GOTGlutamate oxaloacetate transaminase
GPTGlutamate pyruvate transaminase
GPxGlutathione peroxidase
GRGlutathione reductase
GSHGlutathione
GSH-PxGlutathione peroxidase
GSK-3βGlycogen synthase kinase-3 beta
HbA1cGlycated hemoglobin
HDFHuman dermal fibroblast adult cell
HDLHigh-density lipoprotein
HDLCHigh-density lipoprotein-cholesterol
HFEHemochromatosis protein
HGHepatic glycogen
HO-1hemeoxygenase-1
HOMA-IRHomeostasis model assessment of insulin resistance
ICAM-1Intercellular adhesion molecule-1
IFN-γInterferon gamma
IL-6Interleukin-6
IBDInflammatory bowel disease
iNOSInducible nitric oxide synthase
LDHLactate dehydrogenase
LDLLow-density lipoprotein
LDL-cLow-density lipoprotein-cholesterol
LOX-1Lectin-like oxidized low-density lipoprotein receptor 1
LPLLipoprotein lipase
LXRLiver X receptors
MAPKMitogen-activated protein kinase
MCP-1Monocyte chemoattractant protein-1
MDAMalondialdehyde
MGMuscle glycogen
MHCMajor histocompatibility complex
MICMinimum inhibitory concentration
MPOMyeloperoxidase
mRNAMessenger ribonucleic acid
mTORMammalian target of rapamycin
NAFLDNon-alcoholic fatty liver disease
NF-κBNuclear transcription factor-κB
NLRP3Nucleotide-binding domain and leucine-rich repeat containing family Pyrin domain containing 3
NMDAN-methyl-D-aspartate
NONitric oxide
Non-HDLCNon-high-density lipoprotein-cholesterol
NRNMDA-receptor
Nrf2Nuclear factor E2-related factor-2
OAOleic acid
Ox-LDLOxidized low-density lipoprotein
PCNAProliferating cell nuclear antigen
PI3KPhosphoinositide 3-kinase
PMRSPlasma membrane redox system
PPARPeroxisome proliferator-activated receptor
PTP1BProtein tyrosine phosphatase 1B
RG-IRhamnogalacturonan-I
SDHSuccinate dehydrogenase
SODSuperoxide dismutase
SREBP1cSterol regulatory element-binding protein 1c
SUNSerum urea nitrogen
TBARSThiobarbituric acid reactive substances
TCTotal cholesterol
TGTriglyceride
TGF-β1Transforming growth factor β1
TH1Type 1 T helper
TMJTemporomandibular joint
TNF-αTumor necrosis factor alpha
TLR4Toll-like receptor 4
TUNELTerminal deoxynucleotidyl transferase dUTP nick end labeling
UCP2Uncoupling protein 2
UV-BUltraviolet B radiation
VLDLVery-low-density lipoprotein

References

  1. Biswas, T.; Townsend, N.; Huda, M.M.; Maravilla, J.; Begum, T.; Pervin, S.; Ghosh, A.; Mahumud, R.A.; Islam, S.; Anwar, N.; et al. Prevalence of multiple non-communicable diseases risk factors among adolescents in 140 countries: A population-based study. eClinicalMedicine 2022, 52, 101591. [Google Scholar] [CrossRef]
  2. Kapsak, W.R.M.S.R.D.; Rahavi, E.B.R.D.; Childs, N.M.P.; White, C. Functional Foods: Consumer Attitudes, Perceptions, and Behaviors in a Growing Market. J. Am. Diet. Assoc. 2011, 111, 804–810. [Google Scholar] [CrossRef]
  3. Rashidinejad, A. The road ahead for functional foods: Promising opportunities amidst industry challenges. Future Postharvest Food 2024, 1, 266–273. [Google Scholar] [CrossRef]
  4. Fuloria, S.; Mehta, J.; Chandel, A.; Sekar, M.; Rani, N.N.I.M.; Begum, M.Y.; Subramaniyan, V.; Chidambaram, K.; Thangavelu, L.; Nordin, R.; et al. A Comprehensive Review on the Therapeutic Potential of Curcuma longa Linn. in Relation to its Major Active Constituent Curcumin. Front. Pharmacol. 2022, 13, 820806. [Google Scholar] [CrossRef]
  5. Pareek, A.; Pant, M.; Gupta, M.M.; Kashania, P.; Ratan, Y.; Jain, V.; Pareek, A.; Chuturgoon, A.A. Moringa oleifera: An Updated Comprehensive Review of Its Pharmacological Activities, Ethnomedicinal, Phytopharmaceutical Formulation, Clinical, Phytochemical, and Toxicological Aspects. Int. J. Mol. Sci. 2023, 24, 2098. [Google Scholar] [CrossRef]
  6. Dantas, T.L.; Alonso Buriti, F.C.; Florentino, E.R. Okra (Abelmoschus esculentus L.) as a Potential Functional Food Source of Mucilage and Bioactive Compounds with Technological Applications and Health Benefits. Plants 2021, 10, 1683. [Google Scholar] [CrossRef]
  7. Council, N.R. Lost Crops of Africa: Volume II: Vegetables, 1st ed.; National Academies Press: Washington, DC, USA, 2006. [Google Scholar]
  8. Iwu, M.M. Handbook of African Medicinal Plants, 2nd ed.; CRC Press: Boca Raton, FL, USA; Taylor & Francis Group: Abingdon, UK, 2014. [Google Scholar]
  9. Ezuruike, U.F.; Prieto, J.M. The use of plants in the traditional management of diabetes in Nigeria: Pharmacological and toxicological considerations. J. Ethnopharmacol. 2014, 155, 857–924. [Google Scholar] [PubMed]
  10. Lim, T.K. Edible Medicinal And Non Medicinal Plants: Volume 3, Fruits, 1st ed.; Springer: Dordrecht, The Netherlands, 2012. [Google Scholar]
  11. Esakkimuthu, S.; Mutheeswaran, S.; Arvinth, S.; Paulraj, M.G.; Pandikumar, P.; Ignacimuthu, S. Quantitative ethnomedicinal survey of medicinal plants given for cardiometabolic diseases by the non-institutionally trained siddha practitioners of Tiruvallur district, Tamil Nadu, India. J. Ethnopharmacol. 2016, 186, 329–342. [Google Scholar] [CrossRef] [PubMed]
  12. Sivasankari, B.; Anandharaj, M.; Gunasekaran, P. An ethnobotanical study of indigenous knowledge on medicinal plants used by the village peoples of Thoppampatti, Dindigul district, Tamilnadu, India. J. Ethnopharmacol. 2014, 153, 408–423. [Google Scholar] [CrossRef]
  13. Khare, C.P.; Khare, C.P. Indian Medicinal Plants: An Illustrated Dictionary, 2007 ed.; Springer: New York, NY, USA, 2007. [Google Scholar]
  14. Upadhyay, B.; Parveen; Dhaker, A.K.; Kumar, A. Ethnomedicinal and ethnopharmaco-statistical studies of Eastern Rajasthan, India. J. Ethnopharmacol. 2010, 129, 64–86. [Google Scholar] [CrossRef] [PubMed]
  15. Warrier, P.K.; Nambiar, V.P.K.; Ramankutty, C. Indian Medicinal Plants: A Compendium of 500 Species; Sangam Books Limited: London, UK, 1993. [Google Scholar]
  16. Abo, K.A.; Fred-Jaiyesimi, A.A.; Jaiyesimi, A.E.A. Ethnobotanical studies of medicinal plants used in the management of diabetes mellitus in South Western Nigeria. J. Ethnopharmacol. 2008, 115, 67–71. [Google Scholar] [CrossRef]
  17. Moret, E.S.; Voeks, R.; Rashford, J. Trans-Atlantic Diaspora Ethnobotany: Legacies of West African and Iberian Mediterranean Migration in Central Cuba; Springer: New York, NY, USA, 2013; pp. 217–245. [Google Scholar]
  18. Odugbemi, T. Outlines and Pictures of Medicinal Plants from Nigeria; University of Lagos Press: Tolu Odugbemi, Nigeria, 2008; 283p. [Google Scholar]
  19. Quattrocchi, U. CRC World Dictionary of Medicinal and Poisonous Plants: Common Names, Scientific Names, Eponyms, Synonyms, and Etymology (5 Volume Set), 1st ed.; Taylor & Francis Group: Milton, MA, USA, 2012. [Google Scholar]
  20. Muhammad, I.; Matazu, I.K.; Yaradua, I.A.; Yau, S.; Nasir, A.; Bilbis, S.L.; Abbas, Y.A. Development of Okra-Based Antidiabetic Nutraceutical Formulation from Abelmoschus esculentus (L.) Moench (Ex-maradi Variety). Trop. J. Nat. Prod. Res. (TJNPR) 2018, 2, 80–86. [Google Scholar] [CrossRef]
  21. Fernández-Ríos, A.; Laso, J.; Hoehn, D.; Amo-Setién, F.J.; Abajas-Bustillo, R.; Ortego, C.; Fullana-i-Palmer, P.; Bala, A.; Batlle-Bayer, L.; Balcells, M.; et al. A critical review of superfoods from a holistic nutritional and environmental approach. J. Clean. Prod. 2022, 379, 134491. [Google Scholar] [CrossRef]
  22. Elkhalifa, A.E.O.; Alshammari, E.; Adnan, M.; Alcantara, J.C.; Awadelkareem, A.M.; Eltoum, N.E.; Mehmood, K.; Panda, B.P.; Ashraf, S.A. Okra (Abelmoschus Esculentus) as a Potential Dietary Medicine with Nutraceutical Importance for Sustainable Health Applications. Molecules 2021, 26, 696. [Google Scholar] [CrossRef]
  23. Das, S.; Nandi, G.; Ghosh, L. Okra and its various applications in drug delivery, food technology, health care and pharmacological aspects-a review. J. Pharm. Sci. Res. 2019, 11, 2139–2147. [Google Scholar]
  24. de Carvalho, C.C.C.R.; Cruz, P.A.; da Fonseca, M.M.R.; Xavier-Filho, L. Antibacterial Properties of the Extract of Abelmoschus esculentus. Biotechnol. Bioprocess Eng. 2011, 16, 971–977. [Google Scholar] [CrossRef]
  25. Fan, S.; Zhang, Y.; Sun, Q.; Yu, L.; Li, M.; Zheng, B.; Wu, X.; Yang, B.; Li, Y.; Huang, C. Extract of okra lowers blood glucose and serum lipids in high-fat diet-induced obese C57BL/6 mice. J. Nutr. Biochem. 2014, 25, 702–709. [Google Scholar] [CrossRef] [PubMed]
  26. Hu, L.; Yu, W.; Li, Y.; Prasad, K.N.; Tang, Z.; Carvalho, J.C.T. Antioxidant Activity of Extract and Its Major Constituents from Okra Seed on Rat Hepatocytes Injured by Carbon Tetrachloride. BioMed Res. Int. 2014, 2014, 341291. [Google Scholar] [CrossRef]
  27. Liao, H.; Liu, H.; Yuan, K. A new flavonol glycoside from the Abelmoschus esculentus Linn. Pharmacogn. Mag. 2012, 8, 12–15. [Google Scholar]
  28. Liu, J.; Zhao, Y.; Wu, Q.; John, A.; Jiang, Y.; Yang, J.; Liu, H.; Yang, B. Structure characterisation of polysaccharides in vegetable “okra” and evaluation of hypoglycemic activity. Food Chem. 2018, 242, 211–216. [Google Scholar] [CrossRef] [PubMed]
  29. Monte, L.G.; Santi-Gadelha, T.; Reis, L.B.; Braganhol, E.; Prietsch, R.F.; Dellagostin, O.A.; e Lacerda, R.R.; Gadelha, C.A.A.; Conceição, F.R.; Pinto, L.S. Lectin of Abelmoschus esculentus (okra) promotes selective antitumor effects in human breast cancer cells. Biotechnol. Lett. 2014, 36, 461–469. [Google Scholar] [CrossRef] [PubMed]
  30. Tongjaroenbuangam, W.; Ruksee, N.; Chantiratikul, P.; Pakdeenarong, N.; Kongbuntad, W.; Govitrapong, P. Neuroprotective effects of quercetin, rutin and okra (Abelmoschus esculentus Linn.) in dexamethasone-treated mice. Neurochem. Int. 2011, 59, 677–685. [Google Scholar] [CrossRef] [PubMed]
  31. Xia, F.; Zhong, Y.; Li, M.; Chang, Q.; Liao, Y.; Liu, X.; Pan, R. Antioxidant and Anti-Fatigue Constituents of Okra. Nutrients 2015, 7, 8846–8858. [Google Scholar] [CrossRef] [PubMed]
  32. Shen, D.-D.; Li, X.; Qin, Y.-L.; Li, M.-T.; Han, Q.-H.; Zhou, J.; Lin, S.; Zhao, L.; Zhang, Q.; Qin, W.; et al. Physicochemical properties, phenolic profiles, antioxidant capacities, and inhibitory effects on digestive enzymes of okra (Abelmoschus esculentus) fruit at different maturation stages. J. Food Sci. Technol. 2019, 56, 1275–1286. [Google Scholar] [CrossRef] [PubMed]
  33. Lin, Y.; Liu, H.-L.; Fang, J.; Yu, C.-H.; Xiong, Y.-K.; Yuan, K. Anti-fatigue and vasoprotective effects of quercetin-3-O-gentiobiose on oxidative stress and vascular endothelial dysfunction induced by endurance swimming in rats. Food Chem. Toxicol. 2014, 68, 290–296. [Google Scholar] [CrossRef]
  34. Chaemsawang, W.; Prasongchean, W.; Papadopoulos, K.I.; Ritthidej, G.; Sukrong, S.; Wattanaarsakit, P. The Effect of Okra (Abelmoschus esculentus (L.) Moench) Seed Extract on Human Cancer Cell Lines Delivered in Its Native Form and Loaded in Polymeric Micelles. Int. J. Biomater. 2019, 2019, 9404383. [Google Scholar] [CrossRef] [PubMed]
  35. Ping, M.H. Hyperin Controls the Development and Therapy of Gastric Cancer via Regulating Wnt/β-Catenin Signaling. Cancer Manag. Res. 2020, 12, 11773–11782. [Google Scholar] [CrossRef] [PubMed]
  36. Yang, J.; Chen, X.; Rao, S.; Li, Y.; Zang, Y.; Zhu, B. Identification and Quantification of Flavonoids in Okra (Abelmoschus esculentus L. Moench) and Antiproliferative Activity In Vitro of Four Main Components Identified. Metabolites 2022, 12, 483. [Google Scholar] [CrossRef]
  37. Khomsug, P.; Thongjaroe, W.; Pakdeenaro, N.; Suttajit, M.; Chantirati, P. Antioxidative Activities and Phenolic Content of Extracts from Okra (Abelmoschus esculentus L.). Res. J. Biol. Sci. 2010, 5, 310–313. [Google Scholar] [CrossRef]
  38. Lu, Y.; Demleitner, M.F.; Song, L.; Rychlik, M.; Huang, D. Oligomeric proanthocyanidins are the active compounds in Abelmoschus esculentus Moench for its α-amylase and α-glucosidase inhibition activity. J. Funct. Foods 2016, 20, 463–471. [Google Scholar] [CrossRef]
  39. Pan, L.-C.; Sun, Y.-Y.; Zhang, X.-L.; Zhu, Z.-Y.; Liu, C.-Y.; Sun, H.-Q.; Geng, X.-Q.; Jiang, W.; Wang, J.-H. Structure, antioxidant property and protection on PC12 of a polysaccharide isolated and screened from Abelmoschus esculentus L. Moench (okra). Nat. Prod. Res. 2021, 36, 1441–1447. [Google Scholar] [CrossRef] [PubMed]
  40. Zhang, T.; Xiang, J.; Zheng, G.; Yan, R.; Min, X. Preliminary characterization and anti-hyperglycemic activity of a pectic polysaccharide from okra (Abelmoschus esculentus (L.) Moench). J. Funct. Foods 2018, 41, 19–24. [Google Scholar] [CrossRef]
  41. Vayssade, M.; Sengkhamparn, N.; Verhoef, R.; Delaigue, C.; Goundiam, O.; Vigneron, P.; Voragen, A.G.J.; Schols, H.A.; Nagel, M.-D. Antiproliferative and proapoptotic actions of okra pectin on B16F10 melanoma cells. Phytother. Res. 2010, 24, 982–989. [Google Scholar] [CrossRef] [PubMed]
  42. Li, Y.; Deng, Y.; Li, Z.; Liu, Z.; Piao, M.; Cui, X. Composition, physicochemical properties, and anti-fatigue activity of water-soluble okra (Abelmoschus esculentus) stem pectins. Int. J. Biol. Macromol. 2020, 165, 2630–2639. [Google Scholar] [CrossRef]
  43. Liao, Z.; Li, Y.; Liao, L.; Shi, Q.; Kong, Y.; Hu, J.; Cai, Y. Structural characterization and anti-lipotoxicity effects of a pectin from okra (Abelmoschus esculentus (L.) Moench). Int. J. Biol. Macromol. 2023, 238, 124111. [Google Scholar] [CrossRef] [PubMed]
  44. Wang, K.; Li, M.; Wen, X.; Chen, X.; He, Z.; Ni, Y. Optimization of ultrasound-assisted extraction of okra (Abelmoschus esculentus (L.) Moench) polysaccharides based on response surface methodology and antioxidant activity. Int. J. Biol. Macromol. 2018, 114, 1056–1063. [Google Scholar] [CrossRef]
  45. Xiong, B.; Zhang, W.; Wu, Z.; Liu, R.; Yang, C.; Hui, A.; Huang, X.; Xian, Z. Preparation, characterization, antioxidant and anti-inflammatory activities of acid-soluble pectin from okra (Abelmoschus esculentus L.). Int. J. Biol. Macromol. 2021, 181, 824–834. [Google Scholar] [CrossRef] [PubMed]
  46. Zheng, W.; Zhao, T.; Xiangyang, W.U.; Feng, W.; Wang, W.; Ye, Z.O.U.; Zheng, D.; Takase, M.; Qian, L.I.; Huiyu, W.U.; et al. Purification, characterization and immunomodulating activity of a polysaccharide from flowers of Abelmoschus esculentus. Carbohydr. Polym. 2014, 106, 335–342. [Google Scholar] [CrossRef] [PubMed]
  47. Liu, Y.; Ye, Y.; Hu, X.; Wang, J. Structural characterization and anti-inflammatory activity of a polysaccharide from the lignified okra. Carbohydr. Polym. 2021, 265, 118081. [Google Scholar] [CrossRef]
  48. Lengsfeld, C.; Titgemeyer, F.; Faller, G.; Hensel, A. Glycosylated Compounds from Okra Inhibit Adhesion of Helicobacter pylori to Human Gastric Mucosa. J. Agric. Food Chem. 2004, 52, 1495–1503. [Google Scholar] [CrossRef] [PubMed]
  49. Fan, S.; Guo, L.; Zhang, Y.; Sun, Q.; Yang, B.; Huang, C. Okra polysaccharide improves metabolic disorders in high-fat diet-induced obese C57BL/6 mice. Mol. Nutr. Food Res. 2013, 57, 2075–2078. [Google Scholar] [CrossRef] [PubMed]
  50. Thöle, C.; Brandt, S.; Ahmed, N.; Hensel, A. Acetylated Rhamnogalacturonans from Immature Fruits of Abelmoschus esculentus Inhibit the Adhesion of Helicobacter pylori to Human Gastric Cells by Interaction with Outer Membrane Proteins. Molecules 2015, 20, 16770–16787. [Google Scholar] [CrossRef]
  51. Ijarotimi, O.S.; Akinola-Ige, A.O.; Oluwajuyitan, T.D. Okra seeds proteins: Amino acid profile, free radical scavenging activities and inhibition of diabetes and hypertensive converting enzymes indices. Meas. Food 2023, 11, 100101. [Google Scholar] [CrossRef]
  52. de Sousa Ferreira Soares, G.; Assreuy, A.M.S.; de Almeida Gadelha, C.A.; de Morais Gomes, V.; Delatorre, P.; da Conceição Simões, R.; Cavada, B.S.; Leite, J.F.; Nagano, C.S.; Pinto, N.V.; et al. Purification and Biological Activities of Abelmoschus esculentus Seed Lectin. Protein J. 2012, 31, 674–680. [Google Scholar] [CrossRef] [PubMed]
  53. Musthafa, S.A.; Muthu, K.; Vijayakumar, S.; George, S.J.; Murali, S.; Govindaraj, J.; Munuswamy-Ramanujam, G. Lectin isolated from Abelmoschus esculentus induces caspase mediated apoptosis in human U87 glioblastoma cell lines and modulates the expression of circadian clock genes. Toxicon 2021, 202, 98–109. [Google Scholar] [CrossRef] [PubMed]
  54. Khatun, H.; Rahman, M.A.; Biswas, M.; Islam, M.A.U.; Murata, Y.; Pongjanyakul, T. Water-soluble Fraction of Abelmoschus esculentus L Interacts with Glucose and Metformin Hydrochloride and Alters Their Absorption Kinetics after Coadministration in Rats. ISRN Pharm. 2011, 2011, 260537. [Google Scholar] [CrossRef] [PubMed]
  55. Daliu, P.; Annunziata, G.; Tenore, G.C.; Santini, A. Abscisic acid identification in Okra, Abelmoschus esculentus L. (Moench): Perspective nutraceutical use for the treatment of diabetes. Nat. Prod. Res. 2020, 34, 3–9. [Google Scholar] [CrossRef] [PubMed]
  56. Guo, G.; Xu, W.; Zhang, H.; Hu, X.; Chen, Y.; He, X.; Huang, K.; Ma, S.; Fu, J. Characteristics and antioxidant activities of seed oil from okra (Abelmoschus esculentus L.). Food Sci. Nutr. 2024, 12, 2393–2407. [Google Scholar] [CrossRef] [PubMed]
  57. Li, Y.-X.; Yang, Z.-H.; Lin, Y.; Han, W.; Jia, S.-S.; Yuan, K. Antifatigue Effects of Ethanol Extracts and Polysaccharides Isolated from Abelmoschus esculentus. Pharmacogn. Mag. 2016, 12, 219–224. [Google Scholar] [CrossRef]
  58. Sabitha, V.; Ramachandran, S.; Naveen, K.R.; Panneerselvam, K. Investigation of in vivo antioxidant property of Abelmoschus esculentus (L) moench. fruit seed and peel powders in streptozotocin-induced diabetic rats. J. Ayurveda Integr. Med. 2012, 3, 188–193. [Google Scholar] [PubMed]
  59. Tomoda, M.; Shimizu, N.; Gonda, R.; Kanari, M.; Yamada, H.; Hikino, H. Anticomplementary and hypoglycemic activity of Okra and Hibiscus mucilages. Carbohydr. Res. 1989, 190, 323–328. [Google Scholar] [CrossRef] [PubMed]
  60. Huang, C.-N.; Wang, C.-J.; Lin, C.-L.; Lin, H.-T.; Peng, C.-H. The nutraceutical benefits of subfractions of Abelmoschus esculentus in treating type 2 diabetes mellitus. PLoS ONE 2017, 12, e0189065. [Google Scholar] [CrossRef] [PubMed]
  61. Erfani Majd, N.; Tabandeh, M.R.; Shahriari, A.; Soleimani, Z. Okra (Abelmoscus esculentus) Improved Islets Structure, and Down-Regulated PPARs Gene Expression in Pancreas of High-Fat Diet and Streptozotocin-Induced Diabetic Rats. Cell J. 2018, 20, 31–40. [Google Scholar] [PubMed]
  62. Nasrollahi, Z.; ShahaniPour, K.; Monajemi, R.; Ahadi, A.M. Abelmoschus esculentus (L.) Moench improved blood glucose, lipid, and down-regulated PPAR-α, PTP1B genes expression in diabetic rats. J. Food Biochem. 2022, 46, e14097. [Google Scholar] [CrossRef]
  63. Nasrollahi, Z.; ShahaniPour, K.; Monajemi, R.; Ahadi, A.M. Effect of quercetin and Abelmoschus esculentus (L.) Moench on lipids metabolism and blood glucose through AMPK-α in diabetic rats (HFD/STZ). J. Food Biochem. 2022, 46, e14506. [Google Scholar] [CrossRef] [PubMed]
  64. Mishra, N.; Kumar, D.; Rizvi, S.I. Protective Effect of Abelmoschus esculentus Against Alloxan-induced Diabetes in Wistar Strain Rats. J. Diet. Suppl. 2016, 13, 634–646. [Google Scholar] [CrossRef]
  65. Tian, Z.-H.; Miao, F.-T.; Zhang, X.; Wang, Q.-H.; Lei, N.; Guo, L.-C. Therapeutic effect of okra extract on gestational diabetes mellitus rats induced by streptozotocin. Asian Pac. J. Trop. Med. 2015, 8, 1010–1013. [Google Scholar] [CrossRef] [PubMed]
  66. Ben-Chioma, A.E.; Tamuno-Emine, D.G.; Dan, D.B. The Effect of Abelmoschus esculentus in Alloxan- Induced Diabetic Wistar Rat. Int. J. Sci. Res. (IJSR) 2015, 4, 540–543. [Google Scholar]
  67. Sabitha, V.; Ramachandran, S.; Naveen, K.R.; Panneerselvam, K. Antidiabetic and antihyperlipidemic potential of Abelmoschus esculentus (L.) Moench. in streptozotocin-induced diabetic rats. J. Pharm. Bioallied Sci. 2011, 3, 397–402. [Google Scholar] [PubMed]
  68. Liao, Z.; Zhang, J.; Liu, B.; Yan, T.; Xu, F.; Xiao, F.; Wu, B.; Bi, K.; Jia, Y. Polysaccharide from Okra (Abelmoschus esculentus (L.) Moench) Improves Antioxidant Capacity via PI3K/AKT Pathways and Nrf2 Translocation in a Type 2 Diabetes Model. Molecules 2019, 24, 1906. [Google Scholar] [CrossRef] [PubMed]
  69. Peng, C.-H.; Lin, H.-C.; Lin, C.-L.; Wang, C.-J.; Huang, C.-N. Abelmoschus esculentus subfractions improved nephropathy with regulating dipeptidyl peptidase-4 and type 1 glucagon-like peptide receptor in type 2 diabetic rats. J. Food Drug Anal. 2019, 27, 135–144. [Google Scholar] [CrossRef] [PubMed]
  70. Alblihd, M.A.; Alsharif, K.F.; Hamad, A.A.; Ali, F.A.Z.; Hussein, M.T.; Alhegaili, A.S.; Hassan, M.A.; Al-Amer, O.M.; Albezrah, N.K.A.; Almalki, A.A.; et al. Okra [Abelmoschus esculentus (L.) Moench] improved blood glucose and restored histopathological alterations in splenic tissues in a rat model with streptozotocin-induced type 1 diabetes through CD8+ T cells and NF-kβ expression. Front. Vet. Sci. 2023, 10, 1268968. [Google Scholar] [CrossRef]
  71. Gao, H.; Zhang, W.; Wang, B.; Hui, A.; Du, B.; Wang, T.; Meng, L.; Bian, H.; Wu, Z. Purification, characterization and anti-fatigue activity of polysaccharide fractions from okra (Abelmoschus esculentus (L.) Moench). Food Funct. 2018, 9, 188–211. [Google Scholar] [CrossRef] [PubMed]
  72. Saravanan, S.; Pandikumar, P.; Pazhanivel, N.; Paulraj, M.G.; Ignacimuthu, S. Hepatoprotective role of Abelmoschus esculentus (Linn.) Moench., on carbon tetrachloride-induced liver injury. Toxicol. Mech. Methods 2013, 23, 528–536. [Google Scholar] [CrossRef] [PubMed]
  73. Alqasoumi, S.I. ’Okra’ Hibiscus esculentus L.: A study of its hepatoprotective activity. Saudi Pharm. J. 2012, 20, 135–141. [Google Scholar] [CrossRef] [PubMed]
  74. Huynh Ngoc, T.; Nguyen Ngoc, Q.; Tran, A.; Vo Phung, N. Hypolipidemic effect of extracts from Abelmoschus esculentus L. (malvaceae) on tyloxapol-induced hyperlipidemia in mice. Mahidol Univ. J. Pharm. Sci. 2008, 35, 42–46. [Google Scholar]
  75. Wang, H.; Chen, G.; Ren, D.; Yang, S.-T. Hypolipidemic Activity of Okra is Mediated Through Inhibition of Lipogenesis and Upregulation of Cholesterol Degradation. Phytother. Res. 2014, 28, 268–273. [Google Scholar] [CrossRef] [PubMed]
  76. Chen, H.; Jiao, H.; Cheng, Y.; Xu, K.; Jia, X.; Shi, Q.; Guo, S.; Wang, M.; Du, L.; Wang, F. In Vitro and In Vivo Immunomodulatory Activity of Okra (Abelmoschus esculentus L.) Polysaccharides. J. Med. Food 2016, 19, 253–265. [Google Scholar] [CrossRef] [PubMed]
  77. Ramarao, N.; Desu, B.S.R.; Gaddam, D.P.; Bonam, S.R.; Doreddula, S.K.; Pandy, V.; Da Rocha, J.B.T. Phytochemical Analysis, Antioxidant, Antistress, and Nootropic Activities of Aqueous and Methanolic Seed Extracts of Ladies Finger (Abelmoschus esculentus L.) in Mice. Sci. World J. 2014, 2014, 519848. [Google Scholar]
  78. Ebrahimzadeh, M.A.; Nabavi, S.M.; Nabavi, S.F. Antidepressant activity of Hibiscus esculentus L. Eur. Rev. Med. Pharmacol. Sci. 2013, 17, 2609–2612. [Google Scholar] [PubMed]
  79. Yoldaş, M.A.; Bekdaş, M.; Danış, A.; Çetinkaya, A.; Düzcü, S.E.; Alışık, M.; Kocabey, H.; Türel, İ.; Dinçel, G.K. Protective and therapeutic effects of okra seed in acute nontraumatic brain injury. Int. J. Neurosci. 2023, 1–10. [Google Scholar] [CrossRef]
  80. Rival, D.; Bonnet, S.; Sohm, B.; Perrier, E. A Hibiscus Abelmoschus seed extract as a protective active ingredient to favour FGF-2 activity in skin. Int. J. Cosmet. Sci. 2009, 31, 419–426. [Google Scholar] [CrossRef]
  81. Naim, Z.; Billah, M.; Ibrahim, M.; Debnath, D.; Masud Rana, S.; Arefin, P.; Emdadul Hasan Mukul, M. Anti-Inflammatory, Analgesic and Anti-Nociceptive Efficacy of Peel of Abelmoschus esculentus Fruits in Laboratory Animal. Curr. Drug Ther. 2015, 10, 113–121. [Google Scholar] [CrossRef]
  82. Freitas, R.S.; do Val, D.R.; Fernandes, M.E.F.; Gomes, F.I.F.; de Lacerda, J.T.J.G.; SantiGadelha, T.; de Almeida Gadelha, C.A.; de Paulo Teixeira Pinto, V.; Cristino-Filho, G.; Pereira, K.M.A.; et al. Lectin from Abelmoschus esculentus reduces zymosan-induced temporomandibular joint inflammatory hypernociception in rats via heme oxygenase-1 pathway integrity and tnf-α and il-1β suppression. Int. Immunopharmacol. 2016, 38, 313–323. [Google Scholar] [CrossRef]
  83. Alves, S.M.; Freitas, R.S.; do Val, D.R.; Vieira, L.V.; de Assis, E.L.; Gomes, F.I.F.; Gadelha, C.A.d.A.; Gadelha, T.S.; de Lacerda, J.T.J.G.; Clemente-Napimoga, J.T.; et al. The efficacy of a lectin from Abelmoschus Esculentus depends on central opioid receptor activation to reduce temporomandibular joint hypernociception in rats. Biomed. Pharmacother. 2018, 101, 478–484. [Google Scholar] [CrossRef] [PubMed]
  84. Ortaç, D.; Cemek, M.; Karaca, T.; Büyükokuroğlu, M.E.; Özdemir, Z.Ö.; Kocaman, A.T.; Göneş, S. In vivo anti-ulcerogenic effect of okra (Abelmoschus esculentus) on ethanol-induced acute gastric mucosal lesions. Pharm. Biol. 2018, 56, 165–175. [Google Scholar] [CrossRef] [PubMed]
  85. Yan, T.; Nian, T.; Liao, Z.; Xiao, F.; Wu, B.; Bi, K.; He, B.; Jia, Y. Antidepressant effects of a polysaccharide from okra (Abelmoschus esculentus (L) Moench) by anti-inflammation and rebalancing the gut microbiota. Int. J. Biol. Macromol. 2020, 144, 427–440. [Google Scholar] [CrossRef]
  86. Huang, C.-N.; Wang, C.-J.; Lee, Y.-J.; Peng, C.-H. Active subfractions of Abelmoschus esculentus substantially prevent free fatty acid-induced β cell apoptosis via inhibiting dipeptidyl peptidase. PLoS ONE 2017, 12, e0180285. [Google Scholar] [CrossRef] [PubMed]
  87. Liao, H.; Dong, W.; Shi, X.; Liu, H.; Yuan, K. Analysis and comparison of the active components and antioxidant activities of extracts from Abelmoschus esculentus L. Pharmacogn. Mag. 2012, 8, 156–161. [Google Scholar] [PubMed]
  88. Sabitha, V.; Panneerselvam, K.; Ramachandran, S. In vitro α–glucosidase and α–amylase enzyme inhibitory effects in aqueous extracts of Abelmoscus esculentus (L.) Moench. Asian Pac. J. Trop. Biomed. 2012, 2, S162–S164. [Google Scholar] [CrossRef]
  89. Khatun, H.; Rahman, M.A.; Biswas, M.; Islam, M.A.U. In vitro study of the effects of viscous soluble dietary fibers of Abelmoschus esculentus L. in lowering intestinal glucose absorption. Bangladesh Pharm. J. 2010, 13, 35–40. [Google Scholar]
  90. Peng, C.-H.; Chyau, C.-C.; Wang, C.-J.; Lin, H.-T.; Huang, C.-N.; Ker, Y.-B. Abelmoschus esculentus fractions potently inhibited the pathogenic targets associated with diabetic renal epithelial to mesenchymal transition. Food Funct. 2016, 7, 728–740. [Google Scholar] [CrossRef] [PubMed]
  91. Sheu, S.-C.; Lai, M.-H. Composition analysis and immuno-modulatory effect of okra (Abelmoschus esculentus L.) extract. Food Chem. 2012, 134, 1906–1911. [Google Scholar] [CrossRef]
  92. Mairuae, N.; Connor, J.R.; Lee, S.Y.; Cheepsunthorn, P.; Tongjaroenbuangam, W. The effects of okra (Abelmoschus esculentus Linn.) on the cellular events associated with Alzheimer’s disease in a stably expressed HFE neuroblastoma SH-SY5Y cell line. Neurosci. Lett. 2015, 603, 6–11. [Google Scholar] [CrossRef] [PubMed]
  93. Patwardhan, J.; Bhatt, P. Flavonoids Derived from Abelmoschus esculentus Attenuates UV-B Induced Cell Damage in Human Dermal Fibroblasts Through Nrf2-ARE Pathway. Pharmacogn. Mag. 2016, 12, S129–S138. [Google Scholar]
  94. Messing, J.; Thole, C.; Niehues, M.; Shevtsova, A.; Glocker, E.; Boren, T.; Hensel, A. Antiadhesive Properties of Abelmoschus esculentus (Okra) Immature Fruit Extract against Helicobacter pylori Adhesion. PLoS ONE 2014, 9, e84836. [Google Scholar] [CrossRef]
  95. Yakoob, J.; Abbas, Z.; Mehmood, M.H.; Tariq, K.; Saleem, S.A.; Awan, S.; Malik, A.; Hamid, S.; Khan, R.; Jafri, W. Helicobacter pylori outer membrane protein Q genotypes and their susceptibility to anti-adhesive phytotherapeutic agents. J. Integr. Med. 2017, 15, 398–406. [Google Scholar] [CrossRef] [PubMed]
  96. Petropoulos, S.; Fernandes, Â.; Barros, L.; Ciric, A.; Sokovic, M.; Ferreira, I.C.F.R. The chemical composition, nutritional value and antimicrobial properties of Abelmoschus esculentus seeds. Food Funct. 2017, 8, 4733–4743. [Google Scholar] [CrossRef]
  97. Olorunnipa, T.A.; Igbokwe, C.C.; Lawal, T.O.; Adeniyi, B.A.; Mahady, G.B. Anti-helicobacter pylori activity of Abelmoschus esculentus L. moench (okra): An in vitro study. Clin Microb. 2013, 2, 132. [Google Scholar]
  98. Asmat, U.; Abad, K.; Ismail, K. Diabetes mellitus and oxidative stress—A concise review. Saudi Pharm. J. 2016, 24, 547–553. [Google Scholar] [CrossRef]
  99. Evans, J.L.; Goldfine, I.D.; Maddux, B.A.; Grodsky, G.M. Are Oxidative Stress−Activated Signaling Pathways Mediators of Insulin Resistance and β-Cell Dysfunction? Diabetes 2003, 52, 1–8. [Google Scholar] [CrossRef] [PubMed]
  100. Donath, M.Y.; Ehses, J.A.; Maedler, K.; Schumann, D.M.; Ellingsgaard, H.; Eppler, E.; Reinecke, M. Mechanisms of β-Cell Death in Type 2 Diabetes. Diabetes 2005, 54, S108–S113. [Google Scholar] [CrossRef] [PubMed]
  101. Gaal, L.F.v.; Mertens, I.L.; Block, C.E.d. Mechanisms linking obesity with cardiovascular disease. Nature 2006, 444, 875–880. [Google Scholar] [CrossRef]
  102. Esch, T.; Stefano, G.B.; Fricchione, G.L.; Benson, H. The role of stress in neurodegenerative diseases and mental disorders. Neuro-Endocrinol. Lett. 2002, 23, 199–208. [Google Scholar]
  103. Kim, G.H.; Kim, J.E.; Rhie, S.J.; Yoon, S. The Role of Oxidative Stress in Neurodegenerative Diseases. Exp. Neurobiol. 2015, 24, 325–340. [Google Scholar] [CrossRef] [PubMed]
  104. Gupta, A.; Shetty, S.; Mutalik, S.; Nandakumar, K.; Mathew, E.M.; Jha, A.; Mishra, B.; Rajpurohit, S.; Ravi, G.; Saha, M.; et al. Treatment of H. pylori infection and gastric ulcer: Need for novel Pharmaceutical formulation. Heliyon 2023, 9, e20406. [Google Scholar] [CrossRef]
  105. Afsharmanesh, M.R.; Mansourian, A.R.; saghaeian Jazi, M.; Ghaffary, S.; Eshghinia, S.; Behnampour, N.; Jafari, S.M. Okra (Abelmoschus esculentus) Intake Improves Lipid Profile and Liver Transaminases in Pre-diabetic Adults: A Randomized Double-blinded Trial. Jundishapur J. Nat. Pharm. Prod. 2024, 19, e143074. [Google Scholar] [CrossRef]
  106. Bahreini, N.; Saghafi-Asl, M.; Nikpayam, O.; Safaei, E.; Sadra, V.; Fakhr, L.; Beyrampour-Basmenj, H.; Asgharian, P.; Asghari- Jafarabadi, M. Effects of dried okra extract on lipid profile, renal function and some RAGE-related inflammatory genes expression in patients with diabetic nephropathy: A randomized controlled trial. Complement. Ther. Med. 2024, 81, 103027. [Google Scholar] [CrossRef]
  107. Khodija, U.; Wiboworini, B.; Kartikasari, L. Comparing the Effect of Steamed and Boiled Okra (Abelmoschus esculentus) on Fasting Blood Glucose among Type 2 Diabetes Mellitus Patients with Hypercholesterolemia. Int. J. Nutr. Sci. 2020, 5, 65–71. [Google Scholar]
  108. Moradi, A.; Tarrahi, M.-J.; Ghasempour, S.; Shafiepour, M.; Clark, C.C.T.; Safavi, S.-M. The effect of okra (Abelmoschus esculentus) on lipid profiles and glycemic indices in Type 2 diabetic adults: Randomized double blinded trials. Phytother. Res. 2020, 34, 3325–3332. [Google Scholar] [CrossRef] [PubMed]
  109. Nikpayam, O.; Safaei, E.; Bahreyni, N.; Sadra, V.; Saghafi-Asl, M.; Fakhr, L. The effect of Abelmoschus esculentus L. (Okra) extract supplementation on dietary intake, appetite, anthropometric measures, and body composition in patients with diabetic nephropathy. Health Promot. Perspect. 2022, 12, 169–177. [Google Scholar] [CrossRef] [PubMed]
  110. Nikpayam, O.; Saghafi-Asl, M.; Safaei, E.; Bahreyni, N.; Sadra, V.; Asgharian, P. The effect of Abelmoschus esculentus L. (Okra) extract supplementation on glycaemic control, inflammation, kidney function and expression of PPAR-α, PPAR-γ, TGF-β and Nrf-2 genes in patients with diabetic nephropathy: A triple-blind, randomised, placebo-controlled trial. Br. J. Nutr. 2024, 131, 648–657. [Google Scholar] [PubMed]
  111. Saatchi, A.; Aghamohammadzadeh, N.; Beheshtirouy, S.; Javadzadeh, Y.; Afshar, F.H.; Ghaffary, S. Anti-hyperglycemic effect of Abelmoschus culentesus (Okra) on patients with diabetes type 2: A randomized clinical trial. Phytother. Res. 2022, 36, 1644–1651. [Google Scholar] [CrossRef]
  112. Salarfard, M.; Abedian, Z.; Mazlum, S.R.; Rakhshandeh, H.; Akhlaghi, F. The effect of okra powder on blood glucose levels in women with gestational diabetes mellitus: A non-blinded randomized controlled trial. Nurs. Midwifery Stud. 2023, 12, 62–68. [Google Scholar]
  113. Tavakolizadeh, M.; Peyrovi, S.; Ghasemi-Moghaddam, H.; Bahadori, A.; Mohkami, Z.; Sotoudeh, M.; Ziaee, M. Clinical efficacy and safety of okra (Abelmoschus esculentus (L.) Moench) in type 2 diabetic patients: A randomized, double-blind, placebo-controlled, clinical trial. Acta Diabetol. 2023, 60, 1685–1695. [Google Scholar] [CrossRef] [PubMed]
  114. Uebelhack, R.; Bongartz, U.; Seibt, S.; Bothe, G.; Chong, P.W.; De Costa, P.; Wszelaki, N. Double-Blind, Randomized, Three-Armed, Placebo-Controlled, Clinical Investigation to Evaluate the Benefit and Tolerability of Two Dosages of IQP-AE-103 in Reducing Body Weight in Overweight and Moderately Obese Subjects. J. Obes. 2019, 2019, 3412952. [Google Scholar] [PubMed]
  115. Bahari, H.; Shahraki Jazinaki, M.; Rahnama, I.; Aghakhani, L.; Amini, M.R.; Malekahmadi, M. The cardiometabolic benefits of okra-based treatment in prediabetes and diabetes: A systematic review and meta-analysis of randomized controlled trials. Front. Nutr. 2024, 11, 1454286. [Google Scholar] [CrossRef]
  116. Peng, L.V.; Cooper, J.; De Costa, P.; Chong, P.W. Microbiota Composition and Diversity in Weight Loss Population After the Intake of IQP-AE-103 in a Double-Blind, Randomized, Placebo-Controlled Study. Front. Nutr. 2022, 9, 790045. [Google Scholar] [CrossRef]
  117. Kontogiorgos, V.; Margelou, I.; Georgiadis, N.; Ritzoulis, C. Rheological characterization of okra pectins. Food Hydrocoll. 2012, 29, 356–362. [Google Scholar] [CrossRef]
  118. da Silva, R.A.G.; Stocks, C.J.; Hu, G.; Kline, K.A.; Chen, J. Bosutinib Stimulates Macrophage Survival, Phagocytosis, and Intracellular Killing of Bacteria. ACS Infect. Dis. 2024, 10, 1725–1738. [Google Scholar] [CrossRef]
  119. Kwok, C.T.-K.; Chow, F.W.-N.; Cheung, K.Y.-C.; Zhang, X.-Y.; Mok, D.K.-W.; Kwan, Y.-W.; Chan, G.H.-H.; Leung, G.P.-H.; Cheung, K.-W.; Lee, S.M.-Y.; et al. Medulla Tetrapanacis water extract alleviates inflammation and infection by regulating macrophage polarization through MAPK signaling pathway. Inflammopharmacology 2024, 32, 393–404. [Google Scholar] [CrossRef] [PubMed]
  120. Kwok, C.T.-K.; Hu, Y.; Tsoi, B.; Wong, F.; Hau, P.-T.; Tam, E.W.-T.; Mok, D.K.-W.; Kwan, Y.-W.; Leung, G.P.-H.; Lee, S.M.-Y.; et al. Medulla Tetrapanacis water extract ameliorates mastitis by suppressing bacterial internalization and inflammation via MAPKs signaling in vitro and in vivo. Food Front. 2024, 1–16. [Google Scholar] [CrossRef]
  121. Zhu, M.-Z.; Yang, M.-F.; Song, Y.; Xu, H.-M.; Xu, J.; Yue, N.-N.; Zhang, Y.; Tian, C.-M.; Shi, R.-Y.; Liang, Y.-J.; et al. Exploring the efficacy of herbal medicinal products as oral therapy for inflammatory bowel disease. Biomed. Pharmacother. 2023, 165, 115266. [Google Scholar] [CrossRef]
  122. Gutiérrez-Cuevas, J.; Santos, A.; Armendariz-Borunda, J. Pathophysiological Molecular Mechanisms of Obesity: A Link between MAFLD and NASH with Cardiovascular Diseases. Int. J. Mol. Sci. 2021, 22, 11629. [Google Scholar] [CrossRef]
  123. Lee, L.K.; Narang, C.; Rees, C.A.; Thiagarajan, R.R.; Melvin, P.; Ward, V.; Bourgeois, F.T. Reporting and Representation of Participant Race and Ethnicity in National Institutes of Health–Funded Pediatric Clinical Trials. JAMA Netw. Open 2023, 6, e2331316. [Google Scholar] [CrossRef] [PubMed]
  124. Yang, D.; Lew, H.L.; Mak, Y.Y.; Ou, S.J.L.; Lim, J.A.; Lu, Y.; Seah, C.L.Y.; Tan, M.Q.H.; Huang, D.; Tai, E.S.; et al. Incorporation of okra (Abelmoschus esculentus (L.) Moench) seed powder into fresh rice noodles with tapioca starch improves postprandial glycemia, insulinemia and satiety in healthy human volunteers. J. Funct. Foods 2023, 100, 105382. [Google Scholar] [CrossRef]
  125. Lv, Y.; Cai, X.; Shi, N.; Gao, H.; Zhang, Z.; Yan, M.; Li, Y. Emulsification performance and stabilization mechanism of okra polysaccharides with different structural properties. Food Hydrocoll. 2024, 153, 109997. [Google Scholar] [CrossRef]
  126. Olawuyi, I.F.; Park, J.J.; Park, G.D.; Lee, W.Y. Enzymatic Hydrolysis Modifies Emulsifying Properties of Okra Pectin. Foods 2022, 11, 1497. [Google Scholar] [CrossRef] [PubMed]
  127. Aziz, N.S.; Sofian-Seng, N.-S.; Yusop, S.M.; Kasim, K.F.; Mohd Razali, N.S. Functionality of Okra Gum as a Novel Carbohydrate-based Fat Replacer in Ice Cream. Food Sci. Technol. Res. 2018, 24, 519–530. [Google Scholar] [CrossRef]
Figure 1. Fruit of okra.
Figure 1. Fruit of okra.
Foods 14 00177 g001
Figure 2. Cross section of okra fruit with seeds.
Figure 2. Cross section of okra fruit with seeds.
Foods 14 00177 g002
Table 1. Summary of nutrients in okra.
Table 1. Summary of nutrients in okra.
ConstituentsReference
Carbohydrates[22]
Protein[22]
Dietary fiber[22]
Starch[22]
Sugar[22]
Fat[22]
Total omega-3 fatty acids[22]
Total omega-6 fatty acids[22]
Calcium[22]
Phosphorus[22]
Magnesium[22]
Copper[22]
Selenium[22]
Manganese[22]
Zinc[22]
Sodium[22]
Iron[22]
β-carotene [23]
Nicotinic Acid[23]
Riboflavin[23]
Thiamine[23]
Vitamin A[23]
Vitamin C[23]
Vitamin K [23]
Vitamin B complex[23]
Table 2. Summary of active components in okra.
Table 2. Summary of active components in okra.
Compound NameClassBiological
Activity
Isolated from Part of the PlantReferences
Quercetin 3-O-glucosyl (1→6) glucoside (QDG)FlavonoidsAntioxidant, hepatoprotective Seed[26]
Quercetin-3-O-gentiobioseFlavonoidsAntioxidant and antifatigue
Antidiabetic
Vasoprotective
Pod[31,32,33]
Isoquercitrin = quercetin 3-O-glucoside (QG).FlavonoidsAntioxidant
Antifatigue
Anticancer
Antidiabetic
Antihyperlipidemic
Hepatoprotective
Pod and seed[25,26,31,34]
RutinFlavonoidsAntioxidant
Antidiabetic
Neuroprotective
Pod[30,32]
QuercetinFlavonoidsNeuroprotective Pod[30]
Quercetin-3-gentiobiosideFlavonoidsAntitumorPod[35,36]
Quercetin-3-sambubiosideFlavonoidsAntitumorPod[36]
Quercetin-3-malonylglucosideFlavonoidsAntitumorPod[36]
CatechinFlavonoidsAntioxidantPod[37]
EpicatechinFlavonoidsAntioxidantPod[37]
Proanthocyanidins: oligomeric (epi)gallocatechinFlavonoidsAntidiabeticSeed[38]
Procyanidin B1FlavonoidsAntioxidantSeed[37]
Procyanidin B2FlavonoidsAntioxidantSeed[37]
5,7,3′,4′-tetrahydroxy flavonol-3-O-[β-D-glucopyranosyl-(1→6)]-β-D-glucopyranosideFlavonoidsAntioxidantPod[27]
5,7,3′,4′-tetrahydroxy-4″-O-methyl flavonol -3-O-β-D-glucopyranosideFlavonoidsAntioxidantPod[27]
Pectic polysaccharide AeP-P-2PolysaccharideAntioxidant
Neuroprotective
Pod[39]
Pectic polysaccharide WOP-2PolysaccharideAntidiabeticPod[40]
Pectic rhamnogalacturonanPolysaccharideAntitumorPod[41]
Water soluble pectinPolysaccharideAntifatigueStem[42]
Pectin OP-1PolysaccharideAntihyperlipidemic
Hepatoprotective
Pod[43]
Water-soluble polysaccharidePolysaccharideAntioxidantPod[44]
Acid-soluble pectinPolysaccharideAntiinflammatory
Antioxidant
Pod[45]
Polysaccharide OFPS11PolysaccharideAntiinflammatory Flower[46]
Polysaccharide AP1-bPolysaccharideAntiinflammatory Pod[47]
Acidic soluble polysaccharidePolysaccharideAntimicrobialPod[48]
PolysaccharidePolysaccharideAntihyperlipidemic
Antidiabetic
Pod[49]
RhamnogalacturonanPolysaccharideAntidiabetic
Antimicrobial
Pod[28,50]
Protein hydrolysateProteinAntioxidant
Antidiabetic
Antihyperlipidemic
Seed[51]
LectinProteinAntitumor
Anti-inflammatory
Antinociceptive
Seed
Pod
[29,52,53]
Soluble dietary fiberDietary fiberAntidiabeticPod[54]
Abscisic acidPlant hormonesAntidiabeticPod[55]
Linoleic acidFatty acidsAntioxidantSeed[56]
Oleic acidFatty acidsAntioxidantSeed[56]
Palmitic acidFatty acidsAntimicrobialPod[24]
Stearic acidFatty acidsAntimicrobialPod[24]
Table 3. Summary of therapeutic effects of okra in in vivo experiments.
Table 3. Summary of therapeutic effects of okra in in vivo experiments.
Type of Therapeutic EffectsType of ExperimentsTesting SubjectsDescription of the Effects References
Antidiabetic effect
Restoration of β-cell function
In vivoSD rats↓ Exacerbation of β islets → ↓ HbA1, HOMA-IR, and serum glucose levels.[60]
In vivoFemale Wistar rats↓ PPAR-α and –γ mRNA in pancreas → ↑ β-cell in large and small islet in pancreas and ↑ reduced islet’s size, pancreatic disruption, and vacuolization.[61]
In vivoMale Wistar rats↓ Pancreatic beta cell damage, also contain oxidative factors → repair beta cell and ↑ insulin levels.[62]
Improving insulin resistance/sensitivity/glucose tolerance
In vivoFemale Wistar rats↓ PPAR-α and –γ mRNA in pancreas → ↓ HOMA-IR, fasting blood glucose, and ↑ serum insulin.[61]
In vivoFemale C57BL/6 mice↓ PPAR-α and –γ mRNA expression in liver, → ↓ HOMA-IR, blood glucose, fasting blood glucose, and serum insulin.[25]
In vivoC57BL/6 mice↓ PPAR-α, -γ and –β/δ mRNA expression in adipose tissue → ↓ blood glucose and ↑ insulin sensitivity and glucose tolerance.[49]
In vivoMale Wistar rats↓ PTP1B and PPAR-α expressions in liver tissues →↓ HOMA-IR, blood glucose, and fasting blood glucose.[62]
In vivoMale Wistar rats↑ AMPK-α activation, ↓ PEPCK ex-pression → ↑ insulin level → ↑ insulin sensitivity.[63]
Antioxidant activity
In vivoMale Wistar albino rats↑ SOD, CAT, GPx, and GSH levels and ↓ lipid peroxidation (TBARS) in liver, kidney, and pancreases.
↓ Blood glucose.
[58]
In vivoMale Wistar rats↑ Erythrocyte GSH level and FRAP content.
↓ Erythrocyte PMRS activity.
↓ Erythrocyte MDA and plasma AOPP.
[64]
In vivoMale ICR mice↓ Fasting blood glucose and serum MDA.
↑ SOD activity and serum insulin levels.
[40]
Gestational diabetes
In vivoFemale and male SD rats↑ SOD, GPx, GSH, and CAT content in liver and pancreas → ↓ fasting blood glucose, HbA1c, fasting insulin, and ↑ hepatic glycogen.[65]
Inhibition of rate of carbohydrate digestion and glucose absorption
In vivoLong Evans rats↓ Glucose absorption → ↓ blood glucose level.[54]
Hypoglycemia
In vivoMale Wistar albino rats↓ Blood glucose level.[66]
In vivoMale Wistar albino rats↓ Blood glucose level and HbA1c.[67]
In vivoMale C57BL/6 mice↓ Blood glucose level and glucose tolerance.[28]
In vivoMale SPF grade C57BL/6 mice↓ Fasting blood glucose level.[68]
Diabetic nephropathy
In vivoMale SD rats↓ Urine albumin excretion → improve renal function.
↓ Creatinine clearance rate → ↓ hyperfiltration → improve renal function.
↓ Matrix deposition → ↓ renal fibrosis.
↓ Kidney DPP-4 and ↑ GLP-1R expression.
↓ Serum and kidney TBARS.
[69]
Restoration of diabetic-induced splenic damage
In vivoMale Wistar rats↓ Reduction of white pulp, ↑ active red pulp, and ↑ hemosiderin deposition → ↑ effect on restoring the normal immunological function of the spleen.[70]
Antifatigue effectIn vivoMale Kunming mice↑ Weight-loaded swimming endurance time.
↑ HG content.
↓ SUN and BLA content.
[57]
In vivoMale Kunming mice↑ SDH, ATP, and ATPase levels and ↓ LDH and CK levels → ↑ swimming time, ↓ SUN and BLA content, and ↑ HG and MG content.[71]
In vivoMale ICR miceFRAP and reducing power as well as ↓ hepatic MDA and ↑ SOD and GSH-Px → ↑ swimming time, ↓ BLA and SUN content, and ↑ HG content.[31]
In vivoMale SD rat↑ Swimming endurance time.
↓ BLA, SUN, and MDA levels.
↑ HG, MG, SOD, and GSH-Px levels.
[33]
Vasoprotective effectIn vivoMale SD rat↓ Serum MDA level.
↑ SOD and GSH-Px levels → ↓ serum MCP-1, IL-6, and TNF-α levels.
↓ Ox-LDL, LOX-1, and NF-κB p65 expression in aortic tissues.
↓ Ox-LDL, LOX-1, and mRNA expression in aortic tissues → endothelial dysfunction ↓ foam cell in aorta, aorta thickness, and intima–medial thickness.
[33]
Hepatoprotective effect
Antioxidant activity
In vivoMale Wistar rats ↑ Hepatic CAT, SOD, and GSH in rats → ↓ hepatic TG, MDA, and TNF-α, serum AST, ALT, ALP, and total bilirubin content in rats, ↑ serum Albumin in rats, as well as ↓ steatosis, inflammation, and necrosis in rat liver.[72]
In vivoWistar albino rats↓ Serum GOT, GPT, ALP, and GGT levels.
↓ Serum TC and TG levels.
↓ Hepatic MDA and non-protein sulfhydryls (NP-SH) and total protein (TP).
↓ Liver inflammation.
[73]
Antihyperlipidemia effectIn vivoFemale Wistar rats↓ PPAR-α and –γ mRNA in pancreas → ↓ serum TG and TC.[61]
In vivoFemale C57BL/6 mice↓ PPAR-α and -γ and aP2 mRNA expression in liver → ↓ TG → ↓ hepatic steatosis.[25]
In vivoC57BL/6 mice↓ PPAR-α, -γ, -β/δ, and UCP2.
mRNA expression in adipose tissue and LXR and its target ABCG1, ApoE, CYP7A1, and LPL mRNA expression in liver → ↓ serum TC, LDL-c, and ↑ HDL-C.
↓ Size of white adipocytes.
[49]
Mice white adipocytes tissue
In vivoSD rats↓ TG and FFA.
↑ HDL/LDL ratio and HDL.
[60]
In vivoMale Wistar albino rats↓ TC, TG, LDL, and VLDL.
↑ HDL.
[67]
In vivoddY mice↓ Serum TC and TG.[74]
In vivoMale C57BL/6J mice↑ CYP7A1 mRNA expression and ↓ SREBP1c and FAS mRNA expression → ↓ serum TG, TC non-HDL-C, non-HDL-C/HDL-C, and hepatic TG, TC, and ↑ fecal bile acid (bile acid excretion).[75]
Antitumor activity
Immunomodulatory activity
In vivoBALB/c inbred mice↑ Serum TNF-α, IFN-γ, and ↓ IL-10 levels in mice.
↑ Thymus and spleen index and ↑ splenocyte proliferation in mice.
[76]
Neuroprotective effectIn vivoAdult male Swiss albino mice↓ Step-down latency → memory impairment.
↓ Acute restraint stress-induced change in biochemical parameters, e.g., plasma corticosterone, TC, TG, and glucose.
↓ Immobility time.
↑ Time spent and number of entries in open arms of elevated plus arms.
[77]
In vivoMale Swiss albino mice↓ Duration of immobility in forced swimming test and tail suspension tests → antidepressant activity.[78]
In vivoMale ICR mice↓ Escape latency time and ↑ time spent om target quadrant → ↑ learning and ↓ memory impairment.
↑ NR2A/B protein expression.
↑ Average number of BrdU-positive cell per section → ↑ dentate gyrus cell proliferation.
↑ Number of CA3 hippocampal neurons and ↓ morphological damage in the CA3 region.
[30]
In vivoMale Wistar rat↓ Malondialdehyde level and ↓ matrix membrane metalloproteinase-9 level.[79]
Skin protective effectIn vivoNormal women↑ Skin elasticity, firmness, texture, density and ↓ wrinkle in vivo.[80]
Anti-temporomandibular joint (TMJ) inflammatory hypernociception
Anti-inflammation
In vivoSwiss albino mice↓ Carrageenan induced paw edema.[81]
In vivoWistar rats [52]
In vivoMale Wistar rats↓ TNF-αand IL-1βand ↑ HO-1 expression in TMJ tissue → ↓ TNF-α and IL-1β in TMJ tissue and trigeminal ganglion.
↓ Leukocyte cells, MPO activity, and evans blue extravasation in TMJ synovial lavage.
↓ Inflammatory cell influx (↓ inflammatory cell and edema in synovial membrane.
[82]
In vivoMale Wistar rats↓ Evans blue extravasation.
↓ TNF-α in TMJ tissue, trigeminal ganglion, and subnucleus caudalis.
[83]
Analgesic activity
In vivoSwiss albino mice↓ Acetic acid induced writhing.[81]
In vivoMale Swiss albino mice↓ Acetic acid induced abdominal writhing.[52]
Antinociceptive activity
In vivoSwiss albino mice↓ Licking activity.[81]
In vivoMale Wistar rats↑ Head withdrawal threshold → ↓ mechanical hypernociception.[82]
In vivoMale Wistar ratsActivation of central opioid receptors (δ and κ but not µ) → ↓ nociceptive behavior.[83]
Anti-gastric ulcer effect
Gastroprotective effect
In vivoMale Wistar rats↓ Ulcer formation.
↓ Blood MDA and GSH levels.
↑ Serum β—carotene and retinol levels.
↑ PCNA-positive nuclei marker → ↑ cell proliferation in gastric mucosal healing area.
↓ TUNEL positive apoptotic cell.
↓ Gastric damage (↓ edema, hemorrhage, and inflammation scores).
[84]
Antidepressive effect
Anti-inflammatory effect
In vivoMale C57BL/6 mice↓ Toll-like receptor 4 (TLR4)/NF-κB, ↓ NLRP3 inflammasome, and Akt/PI3K pathways, →↓ inflammation.
↑ Activation of MAPK pathways →↑ anti-inflammatory effect → the bidirectional communication of microbiota-gut-brain axis via regulation of inflammation response.
[85]
Key: ↑ = activate/enhance/increase; ↓ = decrease/inhibit/reduce; → = lead to.
Table 4. Summary of therapeutic effects of okra in in vitro experiments.
Table 4. Summary of therapeutic effects of okra in in vitro experiments.
Type of Therapeutic EffectsType of ExperimentsTesting SubjectsDescription of the Effects References
Antidiabetic effect
Restoration of β-cell function
In vitroRINm5F cell↓ % subG1.
↓ Procaspase and caspase 3, DPP-4, AMPK, and Bax expression.
↑ GLP-1R, mTOR, and PI3K expression.
↓ apoptosis.
[86]
Antioxidant activity
In vitroN.A.Good antioxidant activity in DPPH, ABTS, and FRAP.[25]
In vitroN.A.Good antioxidant activity in DPPH and FRAP.[87]
In vitroN.A.High antioxidant activity in DPPH and ABTS.[37]
In vitroN.A.Strong antioxidant activity in DPPH and FRAP.[27]
In vitroN.A.High scavenging activity on superoxide and hydroxyl radical.[40]
In vitroN.A.Good antioxidant activity in DPPH.[62]
Inhibition of rate of carbohydrate digestion and glucose absorption
In vitroα-glucosidase and α-amylase↓ Activity of α-glucosidase and α-amylase.[38,88]
In vitroDiffusion system↓ Glucose diffusion.[89]
Diabetic nephropathy
In vitroHK-2↓ Vimentin, AT-1, TGF-β1, and DPP-4 expression.
↑ cadherin expression.
[90]
Antifatigue effectIn vitro N.A.Good antioxidant activity in DPPH, FRAP, and reducing power.[31]
Hepatoprotective effect
Antioxidant activity
In vitroN.A.
HepG2
High in DPPH, hydroxy radical scavenging activity, and total antioxidant capacity.
↑ GSH in HePG2 and → ↓ ALT, AST, and MDA in HepG2.
[72]
In vitroN.A.Strong reducing power and DPPH, superoxide, and hydroxyl radical scavenging activity
↓ MDA content.
↓ GPT and GOT activity.
↑ SOD and CAT activity.
[26]
In vitroBRL-3A
Antilipotoxicity activity
In vitroHepG2 cells↓ OA-induced lipid accumulation, ROS formation, apoptosis, leakage of transaminases, and inflammatory cytokine secretion →↓ lipotoxicity.
↑ Activation of Adenosine 5′-monophosphate (AMP)-activated protein kinase pathway → ↓ lipotoxicity.
[43]
Antihyperlipidemia effectIn vitroN.A.High bile acid binding capacity.[75]
Antilipotoxicity activity
In vitroHepG2 cells↓ OA-induced lipid accumulation, ROS formation, apoptosis, leakage of transaminases, and inflammatory cytokine secretion →↓ lipotoxicity.
↑ Activation of Adenosine 5′-monophosphate (AMP)-activated protein kinase pathway → ↓ lipotoxicity.
[43]
Antitumor activity
Antiproliferation and apoptosis
In vitroMCF7 and CCD-1059 sk↓ Cell growth % in MCF7 but not CCD-1059 sk.
↑ Caspase-3 and -9 mRNA expression.
↑ p21 mRNA expression and BAX/Bcl-2 expression.
↓ Bcl-2 mRNA expression → ↑ apoptosis in MCF7.
↑ Necrosis in MCF7 depend on interaction with cell surface-expressed carbohydrates.
[29]
In vitroHighly metastatic B16F10↓ Proliferation indices and ↑ % apoptosis cells.
↑ % of cells in G2/M and ↓ % of cells in G1.
↓ Cadherins and α5 integrin expression.
↑ Gal-3 expression.
[41]
Immunomodulatory activity
In vitroBMHC-imDCs↑ Cell size, polymorphic nuclei, dendritic protrusions → ↑ dendritic cell maturation.
↑ MHC class II and CD80/86 expression on the cell surface.
↓ endocytosis activity.
↑ IL-12, IFN-γ, and ↓ IL-10 level → ↑ TH1 response.
[91]
In vitroHepG2 and RAW 264.7↑ NF-κB p65 expression →
↑ iNOS expression and iNOS and TNF-α mRNA expression.
↑ NO, TNF-α, and IL-1β levels.
↑ Phagocytic activity of macrophage.
↑ Macrophage response → ↓ proliferation of HepG2.
[46]
In vitroRAW 264.7↑ RAW 264.7 proliferation.
↑ iNOS expression in RAW 264.7 → ↑ NO level.
↑ TNF-α, IFN-γ, and IL-10 levels in RAW 264.7.
[76]
Neuroprotective effectIn vitroN.A.Good antioxidant activity in FRAP, DPPH, β-Carotene-Linoleic acid, and good chelating effect on ferrous ions. [77]
In vitroSH-SY5Y (wild type and H63D HFE forms)↓ Protein carbonyl l, H2O2, and intracellular ROS levels in cells.
↓ Tau ps199, 202, and 396, and GSK-3β expression.
↓ Intracellular iron in cells.
[92]
Skin protective effectIn vitroFibroblast↑ Protection % of FGF-2 placed in physiological conditions and concentration of FGF-2 in cells.
↑ Sulphated GAG synthesis in fibroblast.
↑ Fibroblast cell proliferation.
[80]
N.A.Good antioxidant capacity in DPPH, ABTS, and FRAP.
↓ UV-B radiation induced cytotoxicity, DNA damage (nongenotoxic), as well as loss of cell membrane integrity and apoptosis.
↓ Nrf2 and HO-1 protein and mRNA expression → ↓ intracellular ROS and depletion of SOD, CAT, GPx, and GR.
[93]
In vitroHDF
Anti-gastric ulcer effect
Anti-adhesive effect of H. pylori to gastric mucosa
In vitroH. pylori and human gastric mucosaInteractions of compounds from okra with bacterial surface structure → ↓ adhesion of H. pylori in human gastric mucosa.[48]
In vitroH. pylori and
human gastric epithelia AGS cell
↓ Bacteria binding to SabA, laminin, lactoferrin, BabA, and HpA binding site → ↓ Adhesion of H. pylori in human gastric epithelia AGS cells.
Esterification → ↑ anti-adhesive activity.
[50]
In vitroH. pylori and human adherent gastric adenocarcinoma epithelia cells↓ binding to BabA, SabA, and fibronectin binding adhesin → ↓ adhesion of H. pylori in AGS.[94]
In vitroH. pyloriH. pylori strains with HopQ genotype or CagA → ↓ adhesion activities.[95]
Antimicrobial activity
Antibacterial activity
In vitroBacillus cereus and Micrococcus flavus
Staphylococcus aureus, Listeria monocytogenes, Escherichia coli, Enterobacter cloacaea, Salmonella enteritidis, and S. typhimurium
Bacteriostatic activity of different genotypes of okra were lower than streptomycin but comparable to ampicillin especially Listeria monocytogenes, Salmonella typhimurium, and Salmonella enteritidis.[96]
In vitroRhodococcus erythrolis R. opacus, Mycobacterium sp., M. aurum, Staphylococcus aureus, Escherichia coli, Xanthobacter Py2, and Pseudomonas aeruginosaLow minimum inhibitory concentration against S.aureus, Mycobacterium sp., Mycobacterium aurum, and X. Py2.
Large inhibition area on the above-mentioned bacteria strains.
↓ Cell viability of bacterial strains.
[24]
In vitroH. pylori strainsHad zone of inhibition → susceptible to okra.
Moderately high MIC.
Showed time dose-dependent bactericidal effect.
[97]
Antifungal activity
In vitroAspergillus fumigatus, A.versicolor, A. ochraceus, A. niger, Cladosporium cladosporioides, Penicillium funiculosum, and P. verrucosumDifferent genotypes of okra showed better or comparable fungistatic and fungicidal activity than ketoconazole, while bifonazole was much more effective than them.[96]
Key: ↑ = activate/enhance/increase; ↓ = decrease/inhibit/reduce; → = lead to.
Table 5. Summary of clinical studies on okra.
Table 5. Summary of clinical studies on okra.
Study DesignSubjectsInterventionDescription of the FindingsReferences
Randomized, double-blind, placebo-controlled clinical trial94 patients with type II diabetes (aged 40–60) in IranTreatment: 1000 mg powdered okra thrice per day for 3 months
Placebo: with the same dosage
Improved glycemic control: ↓ hba1c, fasting blood glucose (FBG), HOMA-IR, and insulin levels
Improved hyperlipidemia: ↓ TG and TC
Alleviated inflammation: ↓ high-sensitivity C-reactive protein (hs-CRP)
No reported adverse effects
[113]
Randomized double-blinded, single-center, plcebo-controlled clinical trial48 patients with type II diabetes (aged 30–75) in IranTreatment: 10 g okra powder (equivalent to 100 g fresh okra) blended in 150 g yogurt (twice per day lunch and dinner) for 8 weeks
Placebo: yogurt with consumable color
Improved glycemic control: ↓ Fasting plasma glucose (FPG), HOMA-IR, and ↑ Quantitative insulin sensitivity checkindex (QUICKI
TC, TG LDL-C, LDL-C/ HDL-C ratio
No reported adverse effects
[108]
Randomized, non-blinded controlled trial60 women with gestational diabetes mellitus (aged 18–35) in IranTreatment: 3 g of okra skin and seed powder twice per day for 4 weeks.
Control: intervention
Improved glycemic control after 2- and 4-week consumption: ↓ fbg and postprandial blood glucose (ppg)[112]
Clinical trial40 patients with type II diabetes and hypercholesterolemia (aged 45–65) in IndonesiaTreatment 1: 40 g boiled okra per day for 2 weeks
Treatment 2: 40 g stream okra per day for 2 weeks
Control: no intervention
Improved glycemic control (both treatments): ↓ fbg[107]
Randomized, double-blinded, placebo-controlled clinical trial70 patients with pre-diabetes (aged 30–55) in IranTreatment: 2 capsules of 500 mg okra (composed with okra powder + magnesium stearate in 10 to 1 ratio) thrice per day for 8 weeks
Placebo: 2 capsules of 500 mg placebo capsules (composed of carboxymethyl cellulose + magnesium stearate in 10 to 1 ratio) thrice per day for 8 weeks
Improved hyperlipidemia: ↓ TC, LDL-C, and ↑ HDL-C
Reduced liver and kidney damage: ↓ ALT, AST, and uric acid
No side effect
[105]
Randomized, double-blind, placebo-controlled clinical trial99 patients with diabetes (aged above 18) receiving oral hypoglycemic medications in IranTreatment: 1000 mg powdered okra capsule every 6 h for 8 weeks
Placebo: microcrystalline cellulose capsule every 6 h for 8 weeks
Improved glycemic control: ↓ FBG, blood sugar, and hba1c
No side effect
No significant effect on lipid profile
[111]
Randomized, triple-blind, placebo-controlled clinical trial55 patients with diabetic nephropathy (aged 40–70) in IranTreatment: capsule containing 80 mg dried okra extract per day for 10 weeks
Placebo: capsule of carboxymethylcellulose per day for 10 weeks
No significant effect on renal function indices, lipid profile, and inflammation[106]
Randomized, triple-blind, placebo-controlled clinical trial55 patients with diabetic nephropathy (aged 40–70) in IranTreatment: capsule containing 80 mg dried okra extract per day for 10 weeks
Placebo: capsule of carbox-ymethylcellulose per day for 10 weeks
↓ Energy and carbohydrate intake[109]
Randomized, triple-blind, placebo-controlled clinical trial55 patients with diabetic nephropathy (aged 40–70) in IranTreatment: capsule containing 80 mg dried okra extract per day for 10 weeks
Placebo: capsule of carbox-ymethylcellulose per day for 10 weeks
Improved glycemic control: ↓FBG, HOMA-IR, and hba1c (in treatment group but not significant between group)
No significant effect on renal function, inflammation
[110]
Randomized, double-blind, three-armed, placebo-controlled clinical trial101 overweight to moderately obese adults (aged 18–65) in GermanyTreatment 1: high dose IQP-AE-103 (330 mg dehydrated okra powder and 85 mg inulin) thrice per day after meal for 12 weeks
Treatment 2: low dose IQP-AE-103 (165 mg dehydrated okra powder and 42.5 mg inulin) for 12 weeks
Placebo: capsules containing standard excipients for 12 weeks
Improved anthropometric measures ↓ weight loss, BMI, waist circumference, and hip circumference (both dosage of IQP-AE-103)
↓ Body Fat
↓ Feeling of hunger in 66% subjects (high dosage)
No side effects reported
[114]
Key: ↑ = activate/enhance/increase; ↓ = decrease/inhibit/reduce.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kwok, C.T.-K.; Ng, Y.-F.; Chan, H.-T.L.; Chan, S.-W. An Overview of the Current Scientific Evidence on the Biological Properties of Abelmoschus esculentus (L.) Moench (Okra). Foods 2025, 14, 177. https://doi.org/10.3390/foods14020177

AMA Style

Kwok CT-K, Ng Y-F, Chan H-TL, Chan S-W. An Overview of the Current Scientific Evidence on the Biological Properties of Abelmoschus esculentus (L.) Moench (Okra). Foods. 2025; 14(2):177. https://doi.org/10.3390/foods14020177

Chicago/Turabian Style

Kwok, Carsten Tsun-Ka, Yam-Fung Ng, Hei-Tung Lydia Chan, and Shun-Wan Chan. 2025. "An Overview of the Current Scientific Evidence on the Biological Properties of Abelmoschus esculentus (L.) Moench (Okra)" Foods 14, no. 2: 177. https://doi.org/10.3390/foods14020177

APA Style

Kwok, C. T.-K., Ng, Y.-F., Chan, H.-T. L., & Chan, S.-W. (2025). An Overview of the Current Scientific Evidence on the Biological Properties of Abelmoschus esculentus (L.) Moench (Okra). Foods, 14(2), 177. https://doi.org/10.3390/foods14020177

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop