Next Article in Journal
Effect of Tetrahedrally Coordinated Al on the Surface Acidity of Mg-Al Binary Mixed Oxides
Next Article in Special Issue
Methanol Extracts from Cirsium japonicum DC. var. australe Kitam. and Their Active Components Reduce Intracellular Oxidative Stress in Caenorhabditis elegans
Previous Article in Journal
Cepharanthine Exerts Antioxidant and Anti-Inflammatory Effects in Lipopolysaccharide (LPS)-Induced Macrophages and DSS-Induced Colitis Mice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Yellow Himalayan Raspberry (Rubus ellipticus Sm.): Ethnomedicinal, Nutraceutical, and Pharmacological Aspects

by
Ananda Lamichhane
1,
Gopal Lamichhane
2 and
Hari Prasad Devkota
3,4,5,*
1
Collage of Pharmacy, Kyungpook National University, Daegu 41566, Republic of Korea
2
Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078, USA
3
Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
4
Headquarters for Admissions and Education, Kumamoto University, Kurokami, 2-39-1, Chuo-ku, Kumamoto 860-8555, Japan
5
Pharmacy Program, Gandaki University, Pokhara 33700, Nepal
*
Author to whom correspondence should be addressed.
Molecules 2023, 28(16), 6071; https://doi.org/10.3390/molecules28166071
Submission received: 27 June 2023 / Revised: 7 August 2023 / Accepted: 8 August 2023 / Published: 15 August 2023

Abstract

:
Yellow Himalayan raspberry (Rubus ellipticus Sm., Rosaceae) is a native species of the Indian subcontinent, Southern China, and the Philippines, which has been historically used as a traditional medicine and food. All of the parts of this plant have been used in traditional medicine to treat respiratory ailments, diabetes, and gastrointestinal disorder, and as an anti-infective agent. The scientific evaluation revealed a richness of macronutrients, micronutrients, and minerals in the fruits, indicating its potential use as a nutraceutical. Furthermore, this plant has been found to be rich in various secondary metabolites, including polyphenols, flavonoids, anthocyanins, tannins, and terpenoids. Ascorbic acid, kaempferol, gallic acid, and catechin are some of the compounds found in this plant, which have been widely discussed for their health benefits. Furthermore, various extracts and compounds obtained from R. ellipticus have shown antioxidant, antidiabetic, anticancer, anti-inflammatory, nephroprotective, antipyretic, anticonvulsant, and anti-infective activities investigated through different study models. These findings in the literature have validated some of the widespread uses of the fruits in folk medicinal systems and the consumption of this nutritious wild fruit by local communities. In conclusion, R. ellipticus holds strong potential for its development as a nutraceutical. It can also improve the nutritional status of villagers and uplift the economy if properly utilized and marketed.

1. Introduction

Medicinal plants serve as an integral component of healthcare in large populations throughout the world. The incorporation of medicinal plants in traditional medicine, spices, nutraceuticals, and as a source of lead compounds has helped humanity fight against many health ailments. Only a fraction of plants have been scientifically documented and studied, and there are many higher plants that are yet to be discovered [1,2]. This diversity of plants and secondary metabolites present in these plants indicates tremendous possibilities for drug discovery and development [3,4]. Currently, scientists are more inclined toward investigating and gathering scientific evidence of ethnomedicinal plants to discover new lead compounds [5,6,7]. This approach, on one hand, would help validate the use of these plants as traditional medicine and nutraceuticals, and on the other hand, increases the likelihood of the discovery of lead compounds compared to random screening [8,9]. Furthermore, polyherbal formulation synergistic therapeutic effects and reduced adverse effects due to individual ingredients can be developed with low doses of multiple ingredients for the management of diseases [10,11,12]. Many bioactive compounds with beneficial effects for various chronic diseases such as diabetes [13], hypercholesterolemia [14,15], high blood pressure [16], cirrhosis [17,18], chronic obstructive pulmonary disease (COPD) [19,20], cancer [21], bacterial infections, aging-related degenerative diseases, and autoimmune diseases [22] have been identified. Research on natural products is still ongoing, searching for pharmacologically active extracts, herbal formulations, and, possibly, newer lead compounds by which to develop them as an effective therapeutic intervention [23]. The growing interest in plant-derived herbal medicine and nutraceuticals with bioactive polyphenols, terpenoids, vitamins, amino acids, and minerals is another reason for the growing research on natural products [24,25,26]. Although researchers have worked on a synthetic approach to develop various derivatives, such as amino sugars, iminosugars, and glycopeptides, as glycosidase inhibitors to discover potent antidiabetic compounds [27,28], the potential of plants as a source of lead compounds cannot be ignored. This is because there are still many unstudied plants that might contain such compounds, indicating the importance of in-depth studies on medicinal plants. Many plant-derived formulations have already been marketed; however, we still need to resolve the challenges regarding the identification of bioactive molecules, the depth study of their pharmacological properties, as well as the design of an effective formulation and drug delivery system using those herbal medicines as well [29,30].
Rubus ellipticus Sm., commonly known as the yellow Himalayan raspberry (Figure 1), is an evergreen shrub belonging to the Rosaceae family. There are approximately 600–800 species belonging to the Rubus genus, categorized into 12 sub-genera [31,32]. Although different species of the Rubus genus are cultivated worldwide, R. ellipticus is the most abundant and wild-growing fruit species on the Indian subcontinent [33]. It is native to the Indian subcontinent, Southern China, and the Philippines, and it is also distributed in Thailand, Sri Lanka, Jamaica, Ecuador, as well as the Central Coast of Australia [34,35,36]. It is locally called “Ainselu” in Nepal and “Hisalu” in India [31,37] and has been reported to be distributed in the hilly and Himalayan region at an altitude of 2600 m. It is a shrub of about 1–3 m in height, covered with glandular hair and a needle-like hook on its stem. The plant is well known for its golden-yellow subglobose fruit, with high nutritional value. Upon maturity, the fruit changes from a green to golden or yellow color, increasing both its esthetic and medicinal value. In Tibetan medicine, matured fruits of R. ellipticus are used to treat coughs and fever [32]. Its roots and shoots are well-known renal tonic, anti-diuretic agents. Recently, this plant has been referred to as a high-nutrient food, and dried fresh leaves are used in powders and other forms. The leaves are rich in many bioactive compounds, including phenols [38], amino acids [39,40], flavonoids [38,41], and terpenoids [42]. In this study, we aim to examine all of the traditional and modern uses of R. ellipticus, including its ethnomedicinal uses, nutritional value, bioactive chemical constituents, and pharmacological activities reported in the literature.

2. Methods

More than 100 research publications were used as the primary source of data for this review paper. Original research articles, review papers, books, book chapters, reports, and short communications were used as the data source from different database sites like Google Scholar, Scopus, and Web of Science. Various keywords such as ethnomedicinal uses of Rubus ellipticus, in vitro and in vivo studies of R. ellipticus, chemical constituents of R. ellipticus, nutritional importance of R. ellipticus, and different pharmacological properties of R. ellipticus were used to search the required information for the database. Ethnomedicinal uses, bioactive chemical constituents, and nutritional composition were especially taken into consideration and the reported pharmaceutical activities of such contained bioactive chemicals were extracted from the database to prepare this manuscript.

3. Botanical Description and Distribution

Rubus ellipticus is a shrub about 1–3 m tall with purplish brown or brownish branches, covered in sparse, curved prickles and dense, purplish brown bristles or glandular hairs. The leaves are composed of three leaflets, with the terminal leaflet having a longer petiole than the lateral leaflets, and the petiole is 2–6 cm long. The leaves have stipules that are linear and 7–11 mm long, and the blade of the leaflets is elliptic or obovate, with the terminal leaflet being larger than the lateral leaflets. The inflorescences are terminal, with dense glomerate racemes that have several (10 or more) flowers. The flowers are 1–1.5 cm in diameter, bisexual, and have white or pink petals that are longer than the sepals. The aggregate fruit is golden yellow and subglobose, with a diameter of approximately 1 cm. It blooms from March to April and bears fruit from April to May [43,44,45].
It grows quickly in both open, sunny areas and dense rainforests and thrives in moist forests in the high-altitude forests of the Himalaya region [46]. It grows along highways, hillsides, thickets, slopes, mountain valleys, and sparsely populated woodlands between 300 and 2600 meters above sea level in an area with annual rainfall between 2000 and 6500 mm [47]. The plant is commonly found in Southern Asia, Southwestern China, Myanmar, Bhutan, Laos, Pakistan, Sri-Lanka, Nepal, Philippines, Vietnam, Thailand, and India. The plant is also found in a certain area of Australia’s Central Coastal New South Wales, Southern Queensland [47] and some countries of Africa [48]. Within India, it is found in Assam, Sikkim, Tamil Nadu, Kerala, and Maharashtra states [32].

4. Ethnomedicinal Uses

The Rubus genus offers opportunities for the farmers not only economically but also medicinally. Its fruits provide extra income to farmers in rural areas without investment, thereby helping to uplift their economic status [49]. According to Tibetan traditional medicine, the leaves and fruits of R. ellipticus can be used treat a variety of conditions such as bronchitis, nausea, ulcers, and diabetes, as well as for its antimicrobial, carminative, and tonic properties [37,50,51]. The root bark is used to treat diarrhea, dysentery, fractured bones, and as an abortifacient and emmenagogue [52]. The shoot of the plant is sometimes chewed to soothe stomach discomfort, and a root decoction can be consumed to warm the stomach [53,54,55,56]. This plant comes under the top ten wild edible medicinal plants of the Tanahun District of Nepal, owing to the widespread use of its fruits and root in folk medicine. Additionally, the juice of R. ellipticus is used for making squash and jams because of its flavor and color [50]. Details of ethnomedicinal uses of R. ellipticus are mentioned in Table 1.
Table 1. Ethnomedicinal uses of R. ellipticus Sm.
Table 1. Ethnomedicinal uses of R. ellipticus Sm.
Parts of Plants UsedDose/FormulationUsed in DiseaseReferences
BarkBark juice is consumedCommon cold and blood disorders[57]
Paste mixed with water and consumedAs an antidiuretic and renal tonic[32,55]
-Common cold [57,58]
FruitRaw fruit is consumedAbdominal pain[59]
DecoctionDysentery[60]
Juice is consumedDiabetes[61,62]
Eaten rawDiarrhea and as diuretics[63]
Juice is consumedSore throat and cold[64]
Juice is consumedCardiac and blood-related diseases[62]
10–20 g fruits, 3 times a dayGastritis, antacid, diarrhea, and dysentery[65]
Juice is consumed Indigestion[66]
Juice is consumed Fever and cough [60]
Ripe fruit1 Teaspoon decoction 3 times a dayFood poisoning[67,68]
DecoctionLoss of appetite, general debility, continuous vomiting after eating[67,68]
Eaten rawAs an aperient[69,70]
-Constipation[65]
Taken as juice and in raw formMouth ulcer[69]
LeavesJuice of 20 leaflets consumedAs a febrifuge[67,68]
Juice/powdered is consumedvomiting[59]
Juice is consumedMouth ulcer and gastrointestinal disorders[69,70]
Leaves and fruitsConsumed as juice, powder, or in raw formGastrointestinal problems and mouth disorders[71]
Root5–10 g of the crushed root as a juiceUsed to reduce fever[67,68]
PasteApplied on wound[60,72]
Powder/JuiceFever and diarrhea[73,74]
Root paste is mixed with various other plants and 1 spoon (fresh) or 1/2 spoon (dry) is consumed with 1 glass of water once a dayMental disorder[64]
Paste applied as a poulticeParalysis, Bone facture[50,64,75]
Paste applied as a poulticeColic pain, aggression[75]
Root paste and ash of Eleusine coracana mixed and applied externally, once a dayWound healing[76]
Juice is takenGastrointestinal and respiratory problems[77]
Decoction is prepared together with other plants and consumedTyphoid and stomach pain, Respiratory tract infection, Gastrointestinal tract infection[77]
Decoction is consumed Used to kill stomach worms and cure other gastric problems [74]
PowderRhinitis and sinusitis[77]
10–20 mL of juice takenDiarrhea, cholera, gastritis, sore throat[78]
Decoction is consumedTyphoid fever[79]
Juice is consumedUrinary tract infection[57]
Root and fruit Eaten rawExcessive thirst and weakness[73]
Root and shoot-Colic pains, antiprotozoal activity against Entamoeba histolytica, hypoglycemic activity[64]
Root and young shoots Paste is taken orallyThroat pain[80]
Whole partRaw/Juice is consumedHypothermia[75,81]
Raw/Juice is consumedAs an astringent and tonic[82]
-Epilepsy[83]
Crushed plant parts along with Osbeckia nepalensis is applied with to skinDermatitis[77]

5. Nutritional Composition

Many studies have reported macronutrients such as carbohydrates, protein, fat, fiber, and several minerals such as calcium, phosphorus, and copper on fruits of the Rubus species. Table 2 and Table 3 below show a comparative representation of the profile of macronutrients and micronutrients of R. ellipticus, respectively. Values for R. fruticosus, another species of Rubus, are also provided, but some of these values are for residues after extracting juice. However, it should be noted that the nutritional composition, total phytochemical content, and compounds isolated vary based on various factors such as environmental factors, ripening stage, processing after collection such as drying, extraction solvents and methods, and analysis methods [84,85,86,87,88,89].

6. Phytochemical Constituents

Rubus species possess diverse secondary metabolites including phenolics, flavonoids, anthocyanins, and tannins. Table 4 shows the values of total phenolic, flavonoid, anthocyanin, and tannin contents observed in R. ellipticus and R. fruticosus. The main compounds isolated/identified from R. ellipticus are listed in Table 5 and structures of main compounds are provided in Figure 2. With respect to another species of Rubus, the main compounds reported from R. fruticosus were campesterol, rubinic acid, rubitic acid, kaempferol, stigmasterol, matairesinol, tocopherol, morin, etc. [90,96].
Table 4. Major phytochemicals contents present in R. ellipticus and R. fruticosus.
Table 4. Major phytochemicals contents present in R. ellipticus and R. fruticosus.
PhytochemicalsValue for R. ellipticusValue for R. fruticosus
Total phenol content343.75 ± 2.21 µg GAE/mg [35]412.38 ± 18.78 mg GAE/100 g fresh weight [97]
Total flavonoid content433.5 ± 13.39 mg CE/100 g [41]77.77 mg QE/100 g [31]
Total anthocyanin content1.71 ± 0.08 CGE/100g [98]152 mg/100 g [31]
Total tannin content628.32 ± 3.17 mg TAE/g [31] 6.50 ± 3.1% of dry matter [99]
µg GAE/mg: Microgram gallic acid equivalent per milligram of extract; mg CE/100 g: milligram catechin.
Table 5. List of bioactive chemical constituents isolated from R. ellipticus.
Table 5. List of bioactive chemical constituents isolated from R. ellipticus.
Phytochemical ClassesBioactive CompoundPlant PartReferences
Flavonoids/ChalconesQuercetin Fruit[100]
RutinFruit[85]
Quercetin 3-O-glucuronideFruit[101]
PhloridzinFruit[85,102]
Kaempferol Leaves[38,103]
Catechin
Epicatechin
Epigallocatechin
Fruit[104,105,106]
Chrysin
Cyanidin
Pelargonidin
Fruit[107]
Phenolic acids/Organic AcidsGallic acid Fruit and Leaves[31,41]
Malic acid Fruit[64]
Ellagic acidFruit and Leaves[38,102]
Chlorogenic acidFruit and Leaves[31]
Citric acid Fruit[32]
Ascorbic acid Fruit[32,98]
Acuminatic acid Root[108]
Quinic acid Fruit[64]
Caffeic acid Leaves and Fruit[31,41]
m-Coumaric acid Fruit[56]
p-Coumaric acidFruit[56,85]
EllagitanninsLambertianin C
Sanguiin H6
Fruit[101]
Triterpenes and SterolsTormentic acid
Miquelianin
Leaves and Root[34,102]
Euscaphic acid Root[42]
β-Sitosterol
β-Sitosterol-β-D-glucoside
Leaves and Root[102,109]
β-CaroteneFruits and Leaves[33,85]
Rosamutin
Sericic acid
Buergericic acid
Root[42]
OleananeLeaves[110]
Ursolic acid Root[108]
CampesterolLeaves and Root[34,102]
Niga-ichgoside-F1 [28-β-Glucopyranosyl ester of 19 α-hydroxyasiatic acid]Leaves and Fruits[111]
Octacosanol
Octacosanic acid
Root[53,102]
3-β-Hydroxy-urs-12,18-diene-28-oic-acid-3-O-(β-D-glucopyranosyl (1-4)-α-L-arabinopyranosideFruit and Aerial Part[53,64]
24-DeoxysericosideWhole Plant[111]
Amino acidsTyrosine
Hydroxy proline
Serine
Histidine
Leucine
Fruit[39]
Figure 2. Chemical structure of major phytochemicals found in R. ellipticus plant.
Figure 2. Chemical structure of major phytochemicals found in R. ellipticus plant.
Molecules 28 06071 g002

7. Biological Activities

7.1. Antioxidant Activity

Rubus species are enriched with various bioactive phytochemicals, especially anthocyanin, phenolic, and flavonoid compounds. All these components play an important role in combating various diseases by scavenging reactive oxygen species (ROS) [35,112]. There are various mechanisms through which R. ellipticus exhibits its antioxidant potential depending upon the bioactive chemical constituents present on the plant. These include scavenging free radicals and ROS, thereby reducing oxidative stress and cellular damage, neutralizing the free radicals by donating electrons, quenching singlet oxygen, and chelation of the metal ions [113]. According to Badhani et al., a high level of various types of chemical constituents including 19.80 mg/100 g fresh weight (fw) of ascorbic acid and 0.99 mg/100 g fw of β-carotene contributes to the antioxidant properties of R. ellipticus [33]. For instance, half maximum inhibitory concentration (IC50,) values of R. ellipticus fruit extract from the α, α-diphenyl-β-picrylhydrazyl (DPPH) assay, ferric ion reducing antioxidant power (FRAP) assay, and 2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) (ABST) assay were found to be 32.8 ± 0.5 µg/mL, 73.1 ± 0.5 µM Fe2+ equivalent, and 39.2 ± 1.1 µg/mL, respectively, revealing the strong antioxidant property of R. ellipticus [114]. Likewise, George et al. reported the significant potential of methanolic leaf extract to scavenge DPPH free radicals with an IC50 value of 6.96 ± 2.32 µg/mL, which was comparable to standard BHA, BHT, and quercetin with IC50 values of 4.88 ± 1.45 µg/mL, 13.18 ± 1.43 µg/mL, and 4.12 ± 1.67 µg/mL, respectively [115].

7.2. Antimalarial Activity

Sachdeva et al. reported the potent anti-malarial activity of R. ellipticus leaf extract in both in vitro and in vivo models. From an in vitro study, they found the antimalarial efficacy of R. ellipticus with an IC50 value of 14.26 μg/mL against the Plasmodium falciparum INDO strain (PfINDO). They observed 64% of inhibition against Plasmodium berghei at a dose of 500 mg/kg [80]. Another study using silver nanoparticles of the leaf extract of R. ellipticus also showed an antimalarial effect against the malarial vector Anophelese stephensi [116]. They found that the aqueous-extract-loaded silver nanoparticles showed greater larvicidal, ovicidal, oviposition deterrent, and adulticidal activities against Culex quinquefasciatus, Anophelese stephensi, and Aedes aegypti as compared to the leaf extract alone [116].

7.3. Antidiabetic Activity

α-Amylase and α-glucosidase are the carbohydrate-hydrolyzing enzymes that catalyze the breakdown of starch and disaccharides into glucose and are thus important in regulating blood glucose level [117,118]. Subba et al. reported a moderate α-amylase inhibitory activity of the methanolic leaf extract with an IC50 value of 269.94 ± 0.11 µg/mL [37]. Li et al. isolated 31 triterpenoids present in R. ellipticus and performed their α-glucosidase inhibition assay. They found that, among all the extracted compounds, euscaphic acid was found to be the most potent with an IC50 value of 0.65 ± 0.09 mM, which was comparable with the positive control, acarbose (0.82 ± 0.11 mM) [42]. Sharma et al. performed an antidiabetic activity assay of R. ellipticus fruit extract using the glucose tolerance test and alloxan-induced diabetes assay using two different experimental diabetic models (Swiss albino mice and Wistar albino rats). It was reported that the ethanolic extract of the fruit was more effective in reducing the blood glucose level in alloxan-induced diabetic Wistar albino rats and Swiss albino mice as compared with its ether and aqueous extract [119]. Since the ethanolic extract was found to contain triterpenoids, flavonoids, as well as saponins, these compounds might contribute to the higher antidiabetic activity of the ethanolic extract compared to others [42,118,120,121].

7.4. Antiproliferative and Anticancer Activity

Uncontrolled proliferation or production of the cells results in cancer. Recently, most researchers have been focused on either synthesizing new compounds or searching potent natural compounds that can be used for the treatment of cancer. A study conducted by George et al. showed an increased survival in tumor-enriched Swiss albino rats with the administration of 250 mg/kg of methanolic leaf extract from R. ellipticus. A similar finding was observed in another study showing a reduced solid tumor volume of Dalton’s Lymphoma Ascites-induced Swiss albino mice when treated with 100 mg/kg of R. ellipticus methanolic leaf extract [115]. An in vitro study by Saini et al., against two human cervical cancer cell lines (C33A and HeLa), showed reduced cell proliferation upon treatment with fruit extract. They found a 60% inhibition in proliferation of the C33A cell line by methanol and acetone extract and the complete inhibition of proliferation in the HeLa cell, with no toxicity to the normal cells [41,122]. The antiproliferative activity of R. ellipticus extract might be due to the presence of the higher concentration of gallic acid and ellagic acid in its extract [122,123]. Different extracts of R. ellipticus, R. niveus, and R. fairholmianus also showed a potent antiproliferative activity of human colon cancer cells (Caco-2) in the MTT ((3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide) assay [38]. This might be due to the presence of diverse phytochemicals in the fruit extract such as kaempferol [38,124].

7.5. Anti-Inflammatory Activity

Parimelazhagan et al. reported the anti-inflammatory activity of R. ellipticus methanolic leaf extract using carrageenan-induced inflammation and croton-oil-induced ear inflammation models in rats. The supplementation of methanolic leaf extract at doses of 400 mg/kg and 200 mg/kg to carrageenan-induced inflamed rats showed a reduction in inflammation. The extract showed a reduction in rat paw with edema by about 66.47% and 45.78%, respectively, at those doses, which were comparable to the standard drug, indomethacin [46]. Hydroalcoholic (90% ethanol) extract of the root of R. ellipticus significantly inhibited the inflammation induced by the carrageenan in rats. The extract was able to reduce the vascular permeability induced by carrageenan, resulting from reduced accumulation of the fluid in the vascular tissues, lowering inflammation [50].

7.6. Antifertility Activity

Prakash et al. performed a series of screening in order to identify the ability of the ethanolic extract of the R. ellipticus plant for its anti-fertility properties. They observed that the extract of R. ellipticus possessed a potent antifertility effect with estrogenic action, as evidenced from screening using rats, mice, and hamsters [125]. Likewise, Dhanabal et al. reported 91.43% of anti-fertility activity, 37.10% of early abortifacient activity, and 54.33% of anti-implantation activity, when 200 mg/mL of R. ellipticus leaf extract was administered to female Wistar albino rats. Additionally, an anti-implantation effect was also observed in female albino rats due to the significantly increased reabsorption site and decreased implantation sites upon treatment with the extract [126]. Similarly, the anti-implantation activities of R. ellipticus root extract and the whole plant without the root extract were reported as 60% and almost 100%, respectively, in albino rats during 7 days of pregnancy [127].

7.7. Nephroprotective Activity

Sharma et al. studied the nephroprotective activity of various extracts of R. ellipticus (petroleum ether, ethanolic, aqueous). These extracts were able to normalize increased blood urea nitrogen, serum uric acid, creatinine, and serum urea levels induced by cisplatin and gentamicin [128]. Such a potent nephroprotective activity might be due to the presence of certain phytochemicals like phenols, tannins, flavonoids, and triterpenoids, which act as an antioxidant, reducing the risk of renal dysfunction [129,130,131]. Likewise, the ethanolic extract of R. ellipticus was found to be an effective nephroprotective agent compared to its petroleum ether and aqueous extract. The extract also improved histological disturbance in acetaminophen-induced nephrotoxicity in animal models [110].

7.8. Antiviral Activity

According to Panda et al., both aqueous as well as ethanol extracts of R. ellipticus showed potent antiviral as well as cytotoxic activities against enterovirus 71 strains revealed by various parameters like the selectivity index, selectivity surface, as well as therapeutic index [132].

7.9. Antipyretic Activity

The antipyretic activity of the leaf methanolic extract of R. ellipticus was studied using yeast-induced hyperpyrexia in rats. Rectal temperature was continuously monitored after the treatment of 200 and 400 mg/kg doses of the extract. A reduction in elevated rectal temperature was observed in the 3rd to 7th hour after treatment, which was comparable to that of the standard drug, paracetamol. This suggested the ability of R. ellipticus extracts to inhibit various inflammatory cytokines produced during hyperpyrexia [46].

7.10. Effect on Central Nervous System (CNS)

The anticonvulsant property of R. ellipticus ethanolic leaf extract was reported using maximum electroshock-induced convulsion in an experimental animal model [133]. In such studies, the severity of convulsion is assessed based on the duration of the flexion, extension, clonus, stupor, and recovery phase [134]. Administration of the ethanolic extract (100 mg/kg per oral) was found to inhibit the extensor phase of convulsion [32,133].

7.11. Antimicrobial Activity

Panda et al. studied the antimicrobial effect of acetone, water, and ethanolic extracts of the leaf of R. ellipticus against Escherichia coli, Staphylococcus aureus, and Candida albicans. More than 50% of growth inhibition of all these microorganisms (bacteria and fungi) was shown by most of the extracts (acetone, water, ethanol) [132]. Likewise, Khanal et al. used disc diffusion and the Resazurin microtiter assay technique to test the methanolic extract of R. ellipticus root bark for antibacterial activity against Gram-positive S. aureus, Gram-negative Klebsiella pneumoniae, and Salmonella typhi. They found that the methanolic extract of R. ellipticus root bark has significant antibacterial activity against S. aureus with a zone of inhibition of 17 mm, but no impact was observed on Gram-negative organisms [135]. Singh et al. reported that the ethanolic leaf extract was capable of inhibiting Pseudomonas aeruginosa and Enterococcus faecalis at an extract concentration of 62.5 µg/mL, whereas S. aureus and S. typhi were sensitive at 125 µg/mL of crude extract [136]. R. ellipticus ethanolic fruit extract was also reported to have significant inhibition against E. coli and Streptococcus pyogenes [39]. Dhatwalia et al. studied the antimicrobial effect of cuprous-oxide-mediated nanoparticles of the fruit extract of R. ellipticus. They found that the synthesized nanoparticle can significantly inhibit Gram-positive bacteria (Bacillus subtilis and S. aureus) with minimum inhibitory concentrations (MICs) of 7.81 µg/mL and 15.62 µg/mL, respectively. Those nanoparticles also inhibited the growth of Gram-negative P. aeruginosa and E. coli with MICs of 31.25 µg/ mL and 31.25, µg/mL, respectively [114].

7.12. Wound Healing Activity

George et al. studied the percentage of contraction of the wound via an excision and infection model with 1% and 2% w/w ointments of R. ellipticus methanolic leaf extract. In the excision model, a 2% acetone-extract-treated Wistar male rat demonstrated 94.23% of contraction on the 12th day of excision, which was comparable with the percentage contraction of betadine, a standard drug. Meanwhile, in the case of an S. aureus-infected wound, complete epithelization was observed in the 2% acetone-extract-treated group on the 12th day with a percentage wound contraction of 79.25% [46].

7.13. Photocatalytic Activity

Methylene blue is a toxic cationic dye used to color paper, leather, and textiles. The dye in industrial effluent is mixed with river as well as other water sources, preventing solar radiation from penetrating and eventually affecting water-based photosynthesis, causing harm to the water ecology, environmental pollution, and poisoning the food chain [137]. Zinc-oxide nanoparticles obtained by green synthesis using fruit aqueous extract of R. ellipticus were found to degrade 17.5% of methylene blue within 1 h, with 72.7% of methylene dye degradation at the end of the experiment [138]. Likewise, Khalil et al. reported 98% of degradation of methylene blue dye after 150 minutes of exposure of R. ellipticus-leaf-mediated silver particles [139]. Thus, R. ellipticus is not only useful for its pharmacological properties but also showed a significant role (photocatalytic) in eliminating water pollution via dye degradation.

8. Conclusions and Future Prospective

Rubus ellipticus is a highly nutritious wild fruit with many health benefits including antioxidant, antidiabetic, anticancer, anti-inflammatory, nephroprotective, antipyretic, anticonvulsant, and anti-infective activities. Its fruits, leaves, and roots are rich in secondary metabolites like polyphenols, flavonoids, anthocyanins, tannins, and terpenoids. These findings in the literature validated the widespread use of this fruit in the folk medicinal system and the consumption of this wild fruit by local communities. Based on this evidence, this wild fruit and extract from different part of this medicinal plant hold strong potential for developing as a nutraceutical. Indeed, as this fruit is readily available in rural villages, it can also help to improve the nutritional status of people living in those areas and also uplift their economy, if properly utilized and marketed. Moreover, detailed research including focused bioactivity studies, standardizations of fruits from different climatic conditions, and advanced toxicological, pharmacological, and pharmacokinetic evaluations is necessary to determine additional uses and the impact of this fruit on the molecular level.
As the drug discovery research using herbal medicine and the demand of nutraceuticals continue growing, the future of wild nutritious fruits like R. ellipticus seems promising. This can play an important role as a source of lead compounds, herbal therapeutic agents, and functional foods. Moreover, this wildly growing medicinal plant can become a good source of nutrition and a means of economic upliftment for the people living in villages. So, future research should focus on confirming the scientific relevance of ethnomedicinal values, the mechanism of action on animal and human subjects, and the commercialization of this wild fruit. Such studies will help highlight this wild fruit and also establish good market value. Furthermore, as this fruit is merely collected from the wild, research on the cultivation and optimization of cultivation to obtain higher yield is also necessary to conserve this plant. This will ensure the sustainable use of this wild fruit and protect against overexploitation.

Author Contributions

Conceptualization and original draft preparation: A.L. and G.L.; reviewing and editing: A.L., G.L. and H.P.D.; supervision: G.L. and H.P.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Authors would like to thank Dipa Paneru and Sujata Lamichhane for providing permission to use their photographs in this manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Not applicable.

References

  1. Van Wyk, B.-E.; Wink, M. Phytomedicines, Herbal Drugs, and Poisons; University of Chicago Press: Chicago, IL, USA, 2015. [Google Scholar]
  2. Gurib-Fakim, A. Medicinal plants: Traditions of yesterday and drugs of tomorrow. Mol. Asp. Med. 2006, 27, 1–93. [Google Scholar] [CrossRef] [PubMed]
  3. Newman, D.J.; Cragg, G.M. Natural products as sources of new drugs over the nearly four decades from 01/1981 to 09/2019. J. Nat. Prod. 2020, 83, 770–803. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Atanasov, A.G.; Waltenberger, B.; Pferschy-Wenzig, E.-M.; Linder, T.; Wawrosch, C.; Uhrin, P.; Temml, V.; Wang, L.; Schwaiger, S.; Heiss, E.H. Discovery and resupply of pharmacologically active plant-derived natural products: A review. Biotechnol. Adv. 2015, 33, 1582–1614. [Google Scholar] [CrossRef] [Green Version]
  5. Singh, Y.D.; Panda, M.K.; Satapathy, K.B. Ethnomedicine for drug discovery. In Advances in Pharmaceutical Biotechnology: Recent Progress and Future Applications; Springer: Singapore, 2020; pp. 15–28. [Google Scholar]
  6. Kunwar, R.M.; Bussmann, R.W. Ethnobotany in the nepal himalaya. J. Ethnobiol. Ethnomed. 2008, 4, 24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Steinmann, D.; Ganzera, M. Recent advances on HPLC/MS in medicinal plant analysis. J. Pharm. Biomed. Anal. 2011, 55, 744–757. [Google Scholar] [CrossRef] [PubMed]
  8. Lamichhane, G.; Sharma, G.; Sapkota, B.; Adhikari, M.; Ghimire, S.; Poudel, P.; Jung, H.-J. Screening of Antioxidant, Antibacterial, Anti-Adipogenic, and Anti-Inflammatory Activities of Five Selected Medicinal Plants of Nepal. J. Exp. Pharmacol. 2023, 15, 93–106. [Google Scholar] [CrossRef]
  9. Bagale, R.; Acharya, S.; Gupta, A.; Chaudhary, P.; Chaudhary, G.P.; Pandey, J. Antibacterial and antioxidant activities of Prinsepia utilis royle leaf and seed extracts. J. Trop. Med. 2022, 2022, 3898939. [Google Scholar] [CrossRef]
  10. Pandeya, P.R.; Lamichhane, R.; Lamichhane, G.; Lee, K.-H.; Lee, H.K.; Rhee, S.-j.; Jung, H.-J. 18KHT01, a potent anti-obesity polyherbal formulation. Front. Pharmacol. 2021, 12, 807081. [Google Scholar] [CrossRef]
  11. Pandeya, P.R.; Lamichhane, G.; Lamichhane, R.; Luo, J.; Li, X.-J.; Rhee, S.-j.; Xiangqian, L.; Jung, H.-J. Antiobesity activity of two polyherbal formulations in high-fat diet-induced obese C57BL/6J mice. BioMed Res. Int. 2022, 2022, 9120259. [Google Scholar] [CrossRef]
  12. Giri, B.R.; Baral, R.; Bhatt, H.; Khadka, A.; Tamrakar, R.; Timalsina, G.; Gyawali, R. Phytochemical Screening, Free-Radical Scavenging Activity, in vitro Alpha-Amylase Inhibitory Activity, and in vivo Hypoglycemic Activity Studies of Several Crude Drug Formulations Based on Selected Medicinal Plants of Nepal. Pharm. Chem. J. 2023, 56, 1369–1378. [Google Scholar] [CrossRef]
  13. Jugran, A.K.; Rawat, S.; Devkota, H.P.; Bhatt, I.D.; Rawal, R.S. Diabetes and plant-derived natural products: From ethnopharmacological approaches to their potential for modern drug discovery and development. Phytother. Res. 2021, 35, 223–245. [Google Scholar] [CrossRef]
  14. Fahed, A.C.; Wang, M.; Patel, A.P.; Ajufo, E.; Maamari, D.J.; Aragam, K.G.; Brockman, D.G.; Vosburg, T.; Ellinor, P.T.; Ng, K. Association of the interaction between familial hypercholesterolemia variants and adherence to a healthy lifestyle with risk of coronary artery disease. JAMA Netw. Open 2022, 5, e222687. [Google Scholar] [CrossRef] [PubMed]
  15. Civeira, F.; International Panel on Management of Familial Hypercholesterolemia. Guidelines for the diagnosis and management of heterozygous familial hypercholesterolemia. Atherosclerosis 2004, 173, 55–68. [Google Scholar] [CrossRef] [PubMed]
  16. Appel, L.J.; Champagne, C.M.; Harsha, D.W.; Cooper, L.S.; Obarzanek, E.; Elmer, P.J.; Stevens, V.J.; Vollmer, W.M.; Lin, P.H.; Svetkey, L.P.; et al. Effects of comprehensive lifestyle modification on blood pressure control: Main results of the PREMIER clinical trial. JAMA 2003, 289, 2083–2093. [Google Scholar] [CrossRef] [PubMed]
  17. Thoma, C.; Day, C.P.; Trenell, M.I. Lifestyle interventions for the treatment of non-alcoholic fatty liver disease in adults: A systematic review. J. Hepatol. 2012, 56, 255–266. [Google Scholar] [CrossRef]
  18. Suzuki, A.; Lindor, K.; St Saver, J.; Lymp, J.; Mendes, F.; Muto, A.; Okada, T.; Angulo, P. Effect of changes on body weight and lifestyle in nonalcoholic fatty liver disease. J. Hepatol. 2005, 43, 1060–1066. [Google Scholar] [CrossRef]
  19. Woo, J. Relationships among diet, physical activity and other lifestyle factors and debilitating diseases in the elderly. Eur. J. Clin. Nutr. 2000, 54, S143–S147. [Google Scholar] [CrossRef] [Green Version]
  20. Bourbeau, J.; Nault, D.; Dang-Tan, T. Self-management and behaviour modification in COPD. Patient Educ. Couns. 2004, 52, 271–277. [Google Scholar] [CrossRef]
  21. Anand, P.; Kunnumakara, A.B.; Sundaram, C.; Harikumar, K.B.; Tharakan, S.T.; Lai, O.S.; Sung, B.; Aggarwal, B.B. Cancer is a preventable disease that requires major lifestyle changes. Pharm. Res. 2008, 25, 2097–2116. [Google Scholar] [CrossRef]
  22. Khan, H.; Sureda, A.; Belwal, T.; Çetinkaya, S.; Süntar, İ.; Tejada, S.; Devkota, H.P.; Ullah, H.; Aschner, M. Polyphenols in the treatment of autoimmune diseases. Autoimmun. Rev. 2019, 18, 647–657. [Google Scholar] [CrossRef]
  23. Ahmad, M.; Masood, S.; Sultana, S.; Hadda, T.B.; Bader, A.; Zafar, M. Antioxidant and nutraceutical value of wild medicinal Rubus berries. Pak. J. Pharm. Sci. 2015, 28, 241–247. [Google Scholar] [PubMed]
  24. Laura, A.; Alvarez-Parrilla, E.; González-Aguilar, G.A. Fruit and Vegetable Phytochemicals: Chemistry, Nutritional Value and Stability; John Wiley & Sons: Hoboken, NJ, USA, 2009. [Google Scholar]
  25. Vivarelli, S.; Costa, C.; Teodoro, M.; Giambò, F.; Tsatsakis, A.M.; Fenga, C. Polyphenols: A route from bioavailability to bioactivity addressing potential health benefits to tackle human chronic diseases. Arch. Toxicol. 2023, 97, 3–38. [Google Scholar] [CrossRef] [PubMed]
  26. Gul, K.; Singh, A.; Jabeen, R. Nutraceuticals and functional foods: The foods for the future world. Crit. Rev. Food Sci. Nutr. 2016, 56, 2617–2627. [Google Scholar] [CrossRef] [PubMed]
  27. Tseng, P.S.; Ande, C.; Moremen, K.W.; Crich, D. Influence of side chain conformation on the activity of glycosidase inhibitors. Angew. Chem. 2023, 135, e202217809. [Google Scholar] [CrossRef]
  28. Chennaiah, A.; Bhowmick, S.; Vankar, Y.D. Conversion of glycals into vicinal-1, 2-diazides and 1, 2-(or 2, 1)-azidoacetates using hypervalent iodine reagents and Me 3 SiN 3. Application in the synthesis of N-glycopeptides, pseudo-trisaccharides and an iminosugar. RSC Adv. 2017, 7, 41755–41762. [Google Scholar] [CrossRef] [Green Version]
  29. Devkota, H.P.; Paudel, K.R.; Khanal, S.; Baral, A.; Panth, N.; Adhikari-Devkota, A.; Jha, N.K.; Das, N.; Singh, S.K.; Chellappan, D.K. Stinging Nettle (Urtica dioica L.): Nutritional Composition, Bioactive Compounds, and Food Functional Properties. Molecules 2022, 27, 5219. [Google Scholar] [CrossRef]
  30. Chaudhary, D.; Lamichhane, G. Overview of natural products in nutraceutical industries. In Herbs, Spices and Their Roles in Nutraceuticals and Functional Foods; Elsevier: Amsterdam, The Netherlands, 2023; pp. 1–13. [Google Scholar]
  31. Singh, B.; Singh, L.; Kewlani, P.; Joshi, V.C.; Bhatt, I.D. Rubus spp.(Rubus armeniacus, Rubus ellipticus, Rubus fruticosus, Rubus nepalensis, Rubus niveus, Rubus occidentalis). In Himalayan Fruits and Berries; Elsevier: Amsterdam, The Netherlands, 2023; pp. 381–394. [Google Scholar]
  32. Pandey, Y.; Bhatt, S.S. Overview of Himalayan yellow raspberry (Rubus ellipticus Smith.): A nutraceutical plant. J. Appl. Nat. Sci. 2016, 8, 6. [Google Scholar] [CrossRef] [Green Version]
  33. Badhani, A.; Rawat, S.; Bhatt, I.D.; Rawal, R.S. Variation in Chemical Constituents and Antioxidant Activity in Yellow Himalayan (Rubus ellipticus Smith) and Hill Raspberry (Rubus niveus Thunb.). J. Food Biochem. 2015, 39, 663–672. [Google Scholar] [CrossRef]
  34. Sharma, S.; Kaur, R.; Kumar, K.; Kumar, D.; Solanke, A.K.U. Genetic variability in Rubus ellipticus collections assessed by morphological traits and EST-SSR markers. J. Plant Biochem. Biotechnol. 2021, 30, 37–55. [Google Scholar] [CrossRef]
  35. Lamichhane, A.; Khatri, S.; Dhungana, M.; Tripathi, B.; Bhattrai, N.; Baral, R.; Jamarkattel, N. Qualitative and quantitative phytochemical screening and free radical scavenging activity of different parts of Rubus ellipticus Sm. Curr. Perspect. Med. Aromat. Plants. 2023, 5, 106–117. [Google Scholar] [CrossRef]
  36. Rubus ellipticus Sm; Plants of the World Online; Royal Botanic Gardens Kew: Richmond, UK. Available online: https://powo.science.kew.org/taxon/urn:lsid:ipni.org:names:736506-1 (accessed on 3 April 2023).
  37. Subba, B.; Gaire, S.; Sharma, K.R. Analysis of phyto-constituents, antioxidant, and alpha amylase inhibitory activities of persea americana Mill., Rhododendron arboretum Sm. Rubus ellipticus Sm. from Arghakhanchi district Nepal. Asian J. Pharm. Clin. Res. 2019, 12, 301. [Google Scholar] [CrossRef]
  38. Muniyandi, K.; George, E.; Sathyanarayanan, S.; George, B.P.; Abrahamse, H.; Thamburaj, S.; Thangaraj, P. Phenolics, tannins, flavonoids and anthocyanins contents influenced antioxidant and anticancer activities of Rubus fruits from Western Ghats, India. Food Sci. Hum. Wellness 2019, 8, 73–81. [Google Scholar] [CrossRef]
  39. Saklani, S.; Chandra, S.; Badoni, P.; Dogra, S. Antimicrobial activity, nutritional profile and phytochemical screening of wild edible fruit of Rubus ellipticus. Int. J. Med. Aromat. Plants 2012, 2, 269–274. [Google Scholar]
  40. Bhardwaj, P.; Hallan, V. Occurrence of Apple stem grooving virus on Rubus ellipticus, a perennial weed in India. Eur. J. Plant Pathol. 2019, 153, 311–319. [Google Scholar] [CrossRef]
  41. Saini, R.; Dangwal, K.; Singh, H.; Garg, V. Antioxidant and antiproliferative activities of phenolics isolated from fruits of Himalayan yellow raspberry (Rubus ellipticus). J. Food Sci. Technol. 2014, 51, 3369–3375. [Google Scholar] [CrossRef] [Green Version]
  42. Li, W.; Fu, H.; Bai, H.; Sasaki, T.; Kato, H.; Koike, K. Triterpenoid saponins from Rubus ellipticus var. obcordatus. J. Nat. Prod. 2009, 72, 1755–1760. [Google Scholar] [CrossRef] [PubMed]
  43. Khaniya, L.; Bhattarai, R.; Jan, H.A.; Hussain, W.; Abbasi, A.M.; Bussmann, R.W.; Paniagua-Zambrana, N.Y. Rubus ellipticus Sm. Rubus foliolosus Weihe & Nees Rubus fruticosus L. Rubus irritans Focke Rosaceae. In Ethnobotany of the Himalayas; Kunwar, R.M., Sher, H., Bussmann, R.W., Eds.; Springer International Publishing: Cham, Switzerland, 2020; pp. 1–17. [Google Scholar]
  44. Singh, R.K.N. Yellow Himalayan Raspberry; Flowers of India, India. Available online: http://www.flowersofindia.net/ (accessed on 5 April 2023).
  45. Shikha, D.; Kashyap, P. Yellow Himalayan Berry. In Antioxidants in Fruits: Properties and Health Benefits; Springer: Singapore, 2020; pp. 67–81. [Google Scholar]
  46. George, B.P.; Parimelazhagan, T.; Saravanan, S. Anti-inflammatory, analgesic and antipyretic activities of Rubus ellipticus Smith. Leaf Methanol Extract. Int. J. Pharm. Pharm. Sci. 2013, 5, 220–224. [Google Scholar]
  47. Wu, K.; Zhang, J.; Zhang, G.; Ding, J. Epiblema tetragonana and Epinotia ustulana (Lepidoptera: Tortricidae), two potential biological control agents for the invasive plant, Rubus ellipticus. Biol. Control 2014, 77, 51–58. [Google Scholar] [CrossRef]
  48. Lalla, R.; Cheek, M.; Nxumalo, M.; Renteria, J. First assessment of naturalised Rubus ellipticus Sm. populations in South Africa-A potential invasion risk? S. Afr. J. Bot. 2018, 114, 111–116. [Google Scholar] [CrossRef]
  49. Sharma, U.S.; Kumar, A. In vitro antioxidant activity of Rubus ellipticus fruits. J. Adv. Pharm. Technol. Res. 2011, 2, 47. [Google Scholar] [CrossRef]
  50. Vadivelan, R.; Bhadra, S.; Ravi, A.; Shanish, K.S.A.; Elango, K.; Suresh, B. Evaluation of anti-inflammatory and membrane stabilizing property of ethanol root extract of Rubus ellipticus Smith in Albino rats. J. Nat. Remedies 2009, 9, 74–78. [Google Scholar]
  51. Shan, S.; Huang, X.; Shah, M.H.; Abbasi, A.M. Evaluation of polyphenolics content and antioxidant activity in edible wild fruits. BioMed Res. Int. 2019, 2019, 1381989. [Google Scholar] [CrossRef] [Green Version]
  52. Kirtikar, K.; Basu, B.; CS, I. Indian medicinal plants, oriental enterprises. Dehradun 2001, 6, 2029–2035. [Google Scholar]
  53. Bhakuni, R.; Shukla, Y.; Thakur, R. Chemical examination of the roots of Rubus ellipticus. Indian Drugs 1987, 24, 272. [Google Scholar]
  54. Yadav, R.; Agarwala, M. Phytochemical analysis of some medicinal plants. J. Phytol. 2011, 3, 10–14. [Google Scholar]
  55. Pfoze, N.L.; Kumar, Y.; Myrboh, B. Survey and assessment of ethnomedicinal plants used in Senapati District of Manipur State, Northeast India. Phytopharmacology 2012, 2, 285–311. [Google Scholar]
  56. Kewlani, P.; Singh, L.; Belwal, T.; Bhatt, I.D. Optimization of ultrasonic-assisted extraction for bioactive compounds in Rubus ellipticus fruits: An important source for nutraceutical and functional foods. Sustain. Chem. Pharm. 2022, 25, 100603. [Google Scholar] [CrossRef]
  57. Wangchuk, P.; Pyne, S.G.; Keller, P.A. Ethnobotanical authentication and identification of Khrog-sman (lower elevation medicinal plants) of Bhutan. J. Ethnopharmacol. 2011, 134, 813–823. [Google Scholar] [CrossRef] [PubMed]
  58. Yeshi, K.; Kashyap, S.; Yangdon, P.; Wangchuk, P. Taxonomical identification of Himalayan edible medicinal plants in Bhutan and the phenolic contents and antioxidant activity of selected plants. J. Biol. Act. Prod. Nat. 2017, 7, 89–106. [Google Scholar] [CrossRef]
  59. Khan, M.P.Z.; Ahmad, M. Traditional preference of Wild Edible Fruits (WEFs) for digestive disorders (DDs) among the indigenous communities of Swat Valley-Pakistan. J. Ethnopharmacol. 2015, 174, 339–354. [Google Scholar] [CrossRef]
  60. Shrestha, N.; Prasai, D.; Shrestha, K.K.; Shrestha, S.; Zhang, X.-C. Ethnomedicinal practices in the highlands of central Nepal: A case study of Syaphru and Langtang village in Rasuwa district. J. Ethnopharmacol. 2014, 155, 1204–1213. [Google Scholar] [CrossRef] [PubMed]
  61. Abbasi, A.M.; Khan, M.A.; Khan, N.; Shah, M.H. Ethnobotanical survey of medicinally important wild edible fruits species used by tribal communities of Lesser Himalayas-Pakistan. J. Ethnopharmacol. 2013, 148, 528–536. [Google Scholar] [CrossRef] [PubMed]
  62. Khan, M.P.Z.; Ahmad, M.; Zafar, M.; Sultana, S.; Ali, M.I.; Sun, H. Ethnomedicinal uses of edible wild fruits (EWFs) in Swat Valley, Northern Pakistan. J. Ethnopharmacol. 2015, 173, 191–203. [Google Scholar] [CrossRef] [PubMed]
  63. Ibrar, M.; Rauf, A.; Hadda, T.B.; Mubarak, M.S.; Patel, S. Quantitative ethnobotanical survey of medicinal flora thriving in Malakand Pass Hills, Khyber Pakhtunkhwa, Pakistan. J. Ethnopharmacol. 2015, 169, 335–346. [Google Scholar] [CrossRef]
  64. Patel, A.; Rojas-Vera, J.; Dacke, C. Therapeutic constituents and actions of Rubus species. Curr. Med. Chem. 2004, 11, 1501–1512. [Google Scholar] [CrossRef] [PubMed]
  65. Maity, D.; Pradhan, N.; Chauhan, A.S. Folk uses of some medicinal plants from North Sikkim. Indian J. Tradit. Knowl. 2004, 3, 66–71. [Google Scholar]
  66. Rana, D.; Bhatt, A.; Lal, B. Ethnobotanical knowledge among the semi-pastoral Gujjar tribe in the high altitude (Adhwari’s) of Churah subdivision, district Chamba, Western Himalaya. J. Ethnobiol. Ethnomed. 2019, 15, 10. [Google Scholar] [CrossRef] [Green Version]
  67. Bhattarai, N. Traditional phytotherapy among the sherpas of Helambu, central Nepal. J. Ethnopharmacol. 1989, 27, 45–54. [Google Scholar] [CrossRef]
  68. Joshi, A.; Edington, J. The use of medicinal plants by two village communities in the central development region of Nepal. Econ. Bot. 1990, 44, 71–83. [Google Scholar] [CrossRef]
  69. Bhatia, H.; Sharma, Y.P.; Manhas, R.; Kumar, K. Ethnomedicinal plants used by the villagers of district Udhampur, J&K, India. J. Ethnopharmacol. 2014, 151, 1005–1018. [Google Scholar] [CrossRef]
  70. Rao, P.; Hasan, S.; Bhellum, B.; Manhas, R. Ethnomedicinal plants of Kathua district, J&K, India. J. Ethnopharmacol. 2015, 171, 12–27. [Google Scholar] [CrossRef] [PubMed]
  71. Singh, B.; Singh, B.; Kishor, A.; Singh, S.; Bhat, M.N.; Surmal, O.; Musarella, C.M. Exploring plant-based ethnomedicine and quantitative ethnopharmacology: Medicinal plants utilized by the population of Jasrota Hill in Western Himalaya. Sustainability 2020, 12, 7526. [Google Scholar] [CrossRef]
  72. Dubey, A.K.; Singh, S. A new whitefly of quarantine importance infesting a native and invasive plant, Rubus ellipticus Sm.(Rosaceae) in the Western Himalaya, India. J. Asia-Pac. Entomol. 2021, 24, 1239–1243. [Google Scholar] [CrossRef]
  73. Rokaya, M.B.; Münzbergová, Z.; Timsina, B. Ethnobotanical study of medicinal plants from the Humla district of western Nepal. J. Ethnopharmacol. 2010, 130, 485–504. [Google Scholar] [CrossRef]
  74. Lata, S.; Yadav, A.; Kumar, P.; Yadav, M. Biodiversity of underutilized fruits of the Himalayas. Genet. Resour. Crop Evol. 2023, 70, 71–94. [Google Scholar] [CrossRef]
  75. De Rus Jacquet, A.; Subedi, R.; Ghimire, S.K.; Rochet, J.C. Nepalese traditional medicine and symptoms related to Parkinson’s disease and other disorders: Patterns of the usage of plant resources along the Himalayan altitudinal range. J. Ethnopharmacol. 2014, 153, 178–189. [Google Scholar] [CrossRef]
  76. Subedi, R. Ethnobotanical Study of Panchase Protected Forest, Kaski District, Central Nepal; Central Department of Botany Tribhuvan University Kirtipur: Kathmandu, Nepal, 2017. [Google Scholar]
  77. Ambu, G.; Chaudhary, R.P.; Mariotti, M.; Cornara, L. Traditional uses of medicinal plants by ethnic people in the Kavrepalanchok district, Central Nepal. Plants 2020, 9, 759. [Google Scholar] [CrossRef]
  78. Bhattarai, K.R. Ethnobotanical study of plants used by Thami community in Ilam District, eastern Nepal. Our Nat. 2018, 16, 55–67. [Google Scholar] [CrossRef]
  79. Khadka, B.; Panthi, M.; Rimal, S. Folklore medicinal plants used against typhoid and fever in Lwangghalel, Kaski District, Central Nepal. J. Plant Resour. 2020, 18, 258–266. [Google Scholar]
  80. Sachdeva, C.; Mohanakrishnan, D.; Kumar, S.; Kaushik, N.K. Assessment of in vitro and in vivo antimalarial efficacy and GC-fingerprints of selected medicinal plant extracts. Exp. Parasitol. 2020, 219, 108011. [Google Scholar] [CrossRef]
  81. Rojas-Vera, J.; Patel, A.V.; Dacke, C.G. Relaxant activity of raspberry (Rubus idaeus) leaf extract in guinea-pig ileum in vitro. Phytother. Res. 2002, 16, 665–668. [Google Scholar] [CrossRef] [PubMed]
  82. Singh, M.; Malla, S.; Rajbhandari, S.; Manandhar, A. Medicinal plants of Nepal—Retrospects and prospects. Econ. Bot. 1979, 33, 185–198. [Google Scholar] [CrossRef]
  83. Sharma, K.; Verma, R.; Kumar, D.; Nepovimova, E.; Kuča, K.; Kumar, A.; Raghuvanshi, D.; Dhalaria, R.; Puri, S. Ethnomedicinal plants used for the treatment of neurodegenerative diseases in Himachal Pradesh, India in Western Himalaya. J. Ethnopharmacol. 2022, 293, 115318. [Google Scholar] [CrossRef] [PubMed]
  84. Liu, W.; Yin, D.; Li, N.; Hou, X.; Wang, D.; Li, D.; Liu, J. Influence of environmental factors on the active substance production and antioxidant activity in Potentilla fruticosa L. and its quality assessment. Sci. Rep. 2016, 6, 28591. [Google Scholar] [CrossRef] [PubMed]
  85. Belwal, T.; Pandey, A.; Bhatt, I.D.; Rawal, R.S.; Luo, Z. Trends of polyphenolics and anthocyanins accumulation along ripening stages of wild edible fruits of Indian Himalayan region. Sci. Rep. 2019, 9, 1–11. [Google Scholar] [CrossRef] [Green Version]
  86. Dirar, A.; Alsaadi, D.; Wada, M.; Mohamed, M.; Watanabe, T.; Devkota, H. Effects of extraction solvents on total phenolic and flavonoid contents and biological activities of extracts from Sudanese medicinal plants. S. Afr. J. Bot. 2019, 120, 261–267. [Google Scholar] [CrossRef]
  87. Belwal, T.; Ezzat, S.M.; Rastrelli, L.; Bhatt, I.D.; Daglia, M.; Baldi, A.; Devkota, H.P.; Orhan, I.E.; Patra, J.K.; Das, G. A critical analysis of extraction techniques used for botanicals: Trends, priorities, industrial uses and optimization strategies. TrAC Trends Anal. Chem. 2018, 100, 82–102. [Google Scholar] [CrossRef]
  88. Belwal, T.; Cravotto, C.; Prieto, M.; Venskutonis, P.R.; Daglia, M.; Devkota, H.P.; Baldi, A.; Ezzat, S.M.; Gómez-Gómez, L.; Salama, M.M. Effects of different drying techniques on the quality and bioactive compounds of plant-based products: A critical review on current trends. Dry. Technol. 2022, 40, 1539–1561. [Google Scholar] [CrossRef]
  89. Pandey, J.; Singh, L.; Saxena, G.; Devkota, H.P. Environmental challenges for himalayan medicinal plants. In Environmental Challenges and Medicinal Plants: Sustainable Production Solutions under Adverse Conditions; Springer: Berlin, Germany, 2022; pp. 29–47. [Google Scholar]
  90. Zia-Ul-Haq, M.; Riaz, M.; De Feo, V.; Jaafar, H.Z.; Moga, M. Rubus fruticosus L.: Constituents, biological activities and health related uses. Molecules 2014, 19, 10998–11029. [Google Scholar] [CrossRef] [Green Version]
  91. Zafra-Rojas, Q.; Cruz-Cansino, N.; Delgadillo-Ramírez, A.; Alanís-García, E.; Añorve-Morga, J.; Quintero-Lira, A.; Castañeda-Ovando, A.; Ramírez-Moreno, E. Organic acids, antioxidants, and dietary fiber of Mexican blackberry (Rubus fruticosus) residues cv. Tupy. J. Food Qual. 2018, 2018, 5950761. [Google Scholar] [CrossRef] [Green Version]
  92. Stajčić, S.M.; Tepić, A.N.; Đilas, S.M.; Šumić, Z.M.; Čanadanović-Brunet, J.M.; Ćetković, G.S.; Vulić, J.J.; Tumbas, V.T. Chemical composition and antioxidant activity of berry fruits. Acta Period. Technol. 2012, 43, 93–105. [Google Scholar] [CrossRef]
  93. Plessi, M.; Bertelli, D.; Albasini, A. Distribution of metals and phenolic compounds as a criterion to evaluate variety of berries and related jams. Food Chem. 2007, 100, 419–427. [Google Scholar] [CrossRef]
  94. Schulz, M.; Chim, J.F. Nutritional and bioactive value of Rubus berries. Food Biosci. 2019, 31, 100438. [Google Scholar] [CrossRef]
  95. Van de Velde, F.; Grace, M.H.; Esposito, D.; Pirovani, M.É.; Lila, M.A. Quantitative comparison of phytochemical profile, antioxidant, and anti-inflammatory properties of blackberry fruits adapted to Argentina. J. Food Compos. Anal. 2016, 47, 82–91. [Google Scholar] [CrossRef]
  96. Ono, M.; Yasuda, S.; Nishi, K.; Yamamoto, K.; Fuchizaki, S.; Higuchi, S.; Komatsu, H.; Okawa, M.; Kinjo, J.; Yoshimitsu, H. Two new triterpenoids from the seeds of blackberry (Rubus fructicosus). Nat. Prod. Res. 2016, 30, 904–911. [Google Scholar] [CrossRef]
  97. Cosmulescu, S.; Trandafir, I.; Nour, V. Phenolic acids and flavonoids profiles of extracts from edible wild fruits and their antioxidant properties. Int. J. Food Prop. 2017, 20, 3124–3134. [Google Scholar] [CrossRef] [Green Version]
  98. Karuppusamy, S.; Muthuraja, G.; Rajasekaran, K. Antioxidant Activity of Selected Lesser Known Edible Fruits from Western Ghats of India; NISCAIR-CSIR: New Delhi, India, 2011. [Google Scholar]
  99. Gudej, J.; Tomczyk, M. Determination of flavonoids, tannins and ellagic acid in leaves from Rubus L. species. Arch. Pharmacal Res. 2004, 27, 1114–1119. [Google Scholar] [CrossRef]
  100. Sasikumar, J.; Poulin, R.; Meseret, C.; Selvakumar, P. In vitro analysis of antioxidant capacity of Indian yellow raspberry (Rubus ellipticus Smith.). Int. Food Res. J. 2015, 22, 1338–1346. [Google Scholar]
  101. Burlando, B.; Cornara, L.; Boggia, R. Nutraceutical Potential of High-latitude and High-altitude Berries Rich in Ellagitannins. Curr. Med. Chem. 2023, 30, 2121–2140. [Google Scholar] [CrossRef]
  102. Kewlani, P.; Tiwari, D.; Rawat, S.; Bhatt, I.D. Pharmacological and phytochemical potential of Rubus ellipticus: A wild edible with multiple health benefits. J. Pharm. Pharmacol. 2023, 75, 143–161. [Google Scholar] [CrossRef]
  103. Joshi, R.K.; Laurindo, L.F.; Rawat, P.; de Alvares Goulart, R.; Barbalho, S.M. Himalayan Yellow Raspberry (Rubus ellipticus Smith.): A Plant with Multiple Medicinal Purposes. Rec. Agric. Food. Chem. 2022, 2, 75–83. [Google Scholar]
  104. Panche, A.N.; Diwan, A.D.; Chandra, S.R. Flavonoids: An overview. J. Nutr. Sci. 2016, 5, e47. [Google Scholar] [CrossRef] [Green Version]
  105. Agrawal, A. Pharmacological activities of flavonoids: A review. Int. J. Pharm. Sci. Nanotechnol. 2011, 4, 1394–1398. [Google Scholar] [CrossRef]
  106. Bachheti, A.; Deepti; Bachheti, R.K.; Singh, A.; Zebeaman, M.; Hunde, Y.; Husen, A. Bioactive constituents and health promoting compounds of underutilized fruits of the northern Himalayas of India: A review. Food Prod. Process. Nutr. 2023, 5, 24. [Google Scholar] [CrossRef]
  107. Dias, M.C.; Pinto, D.C.; Silva, A.M. Plant flavonoids: Chemical characteristics and biological activity. Molecules 2021, 26, 5377. [Google Scholar] [CrossRef] [PubMed]
  108. Talapatra, S.; Shrestha, K.; Talapatra, B. Chemical Investigation of Some Medicinal-Plants of Nepal 2; Council Scientific Industrial Research Publication and Information Directorate: New Delhi, India, 1989; Volume 28, pp. 880–881. [Google Scholar]
  109. Aswal, B.; Goel, A.; Kulshrestha, D.; Mehrotra, B.; Patnaik, G. Screening of Indian Plants for Biological Activity: Part XV; CSIR-CDRI: Lucknow, India, 1996. [Google Scholar]
  110. Karn, A.; Quasim, M.A.; Paul, S.; Sharma, H.K. An updated review of Rubus ellipticus (an edible shrub), its bioactive constituents and functional properties. Sci. Phytochem. 2022, 1, 22–33. [Google Scholar] [CrossRef]
  111. Xiao-Hong, Z.; Kasai, R.; Ohtani, K.; Tanaka, O.; Rui-Lin, N.; Chong-Ren, Y.; Jun, Z.; Yamasaki, K. Oleanane and ursane glucosides from Rubus species. Phytochemistry 1992, 31, 3642–3644. [Google Scholar] [CrossRef]
  112. Trivedi, A.; Verma, S.; Tyagi, R. Variability in morpho-physiological traits and antioxidant potential of Rubus species in Central Himalayan Region. Ind. Crops Prod. 2016, 82, 1–8. [Google Scholar] [CrossRef]
  113. Santos-Sánchez, N.F.; Salas-Coronado, R.; Villanueva-Cañongo, C.; Hernández-Carlos, B. Antioxidant compounds and their antioxidant mechanism. In Antioxidants; IntechOpen Limited: London, UK, 2019; Volume 10, pp. 1–29. [Google Scholar]
  114. Dhatwalia, J.; Kumari, A.; Chauhan, A.; Batoo, K.M.; Banerjee, A.; Radhakrishnan, A.; Thakur, S.; Guleria, I.; Lal, S.; Ghotekar, S. Rubus ellipticus fruits extract-mediated cuprous oxide nanoparticles: In vitro antioxidant, antimicrobial, and toxicity study. Chem. Pap. 2023, 77, 1377–1393. [Google Scholar]
  115. George, B.P.; Parimelazhagan, T.; Kumar, Y.T.; Sajeesh, T. Antitumor and wound healing properties of Rubus ellipticus Smith. J. Acupunct. Meridian Stud. 2015, 8, 134–141. [Google Scholar] [CrossRef] [Green Version]
  116. Khanal, L.N.; Sharma, K.R.; Paudyal, H.; Parajuli, K.; Dahal, B.; Ganga, G.; Pokharel, Y.R.; Kalauni, S.K. Green synthesis of silver nanoparticles from root extracts of Rubus ellipticus Sm. and comparison of antioxidant and antibacterial activity. J. Nanomater. 2022, 2022, 1832587. [Google Scholar] [CrossRef]
  117. Benmohamed, M.; Guenane, H.; Messaoudi, M.; Zahnit, W.; Egbuna, C.; Sharifi-Rad, M.; Chouh, A.; Seghir, B.B.; Rebiai, A.; Boubekeur, S. Mineral Profile, Antioxidant, Anti-Inflammatory, Antibacterial, Anti-Urease and Anti-α-Amylase Activities of the Unripe Fruit Extracts of Pistacia atlantica. Molecules 2023, 28, 349. [Google Scholar] [CrossRef] [PubMed]
  118. Papoutsis, K.; Zhang, J.; Bowyer, M.C.; Brunton, N.; Gibney, E.R.; Lyng, J. Fruit, vegetables, and mushrooms for the preparation of extracts with α-amylase and α-glucosidase inhibition properties: A review. Food Chem. 2021, 338, 128119. [Google Scholar] [CrossRef]
  119. Sharma, U.; Kumar, A. Anti-diabetic effect of Rubus ellipticus fruit extracts in alloxan induced diabetic rats. J. Diabetol. 2011, 2, 4. [Google Scholar]
  120. Elekofehinti, O.O. Saponins: Anti-diabetic principles from medicinal plants–A review. Pathophysiology 2015, 22, 95–103. [Google Scholar] [CrossRef] [PubMed]
  121. Shamsudin, N.F.; Ahmed, Q.U.; Mahmood, S.; Shah, S.A.A.; Sarian, M.N.; Khattak, M.M.A.K.; Khatib, A.; Sabere, A.S.M.; Yusoff, Y.M.; Latip, J. Flavonoids as antidiabetic and anti-inflammatory agents: A review on structural activity relationship-based studies and meta-analysis. Int. J. Mol. Sci. 2022, 23, 12605. [Google Scholar] [CrossRef]
  122. You, B.R.; Moon, H.J.; Han, Y.H.; Park, W.H. Gallic acid inhibits the growth of HeLa cervical cancer cells via apoptosis and/or necrosis. Food Chem. Toxicol. 2010, 48, 1334–1340. [Google Scholar] [CrossRef]
  123. Losso, J.N.; Bansode, R.R.; Trappey, A., II; Bawadi, H.A.; Truax, R. In vitro anti-proliferative activities of ellagic acid. J. Nutr. Biochem. 2004, 15, 672–678. [Google Scholar] [CrossRef]
  124. Zhong, R.; Guo, Q.; Zhou, G.; Fu, H.; Wan, K. Three new labdane-type diterpene glycosides from fruits of Rubus chingii and their cytotoxic activities against five humor cell lines. Fitoterapia 2015, 102, 23–26. [Google Scholar] [CrossRef] [PubMed]
  125. Prakash, A. Biological evaluation of some medicinal plant extracts for contraceptive efficacy in females. In Future Aspects in Contraception; Springer: Dordrecht, The Netherlands, 1985; pp. 115–128. [Google Scholar]
  126. Dhanabal, S.; Prasanth, S.; Ramanathan, M.; Elango, K.; Suresh, B. Validation of antifertility activity of various Rubus species in female albino rats. Indian J. Pharm. Sci. 2000, 62, 58. [Google Scholar]
  127. Sharma, B.; Varshney, M.; Gupta, D.; Prakash, A.O. Antifertility screening of plants. Part I. Effect of ten indigenous plants on early pregnancy in albino rats. Int. J. Crude Drug Res. 1983, 21, 183–187. [Google Scholar] [CrossRef]
  128. Sharma, U.S.; Kumar, A. Nephroprotective evaluation of Rubus ellipticus (smith) fruits extracts against cisplatin and gentamicin induced renal-toxicity in rats. J. Pharm. Res. 2011, 1, 285–287. [Google Scholar]
  129. Sharma, I.; Khan, M.U.; Zahiruddin, S.; Basist, P.; Ahmad, S. Multi-Mechanistic and Therapeutic Exploration of Nephroprotective Effect of Traditional Ayurvedic Polyherbal Formulation Using In Silico, In Vitro and In Vivo Approaches. Biomedicines 2023, 11, 168. [Google Scholar] [CrossRef]
  130. Ma, Q.; Wang, L.-H.; Jiang, J.-G. Hepatoprotective effect of flavonoids from Cirsium japonicum DC on hepatotoxicity in comparison with silymarin. Food Funct. 2016, 7, 2179–2184. [Google Scholar] [CrossRef]
  131. Santos, R.S.; Chaves-Filho, A.B.; Silva, L.A.; Garcia, C.A.; Silva, A.R.; Dolabella, S.S.; da Costa, S.S.; Miyamoto, S.; Matos, H.R. Bioactive compounds and hepatoprotective effect of Hancornia speciosa gomes fruit juice on acetaminophen-induced hepatotoxicity in vivo. Nat. Prod. Res. 2022, 36, 2565–2569. [Google Scholar] [CrossRef]
  132. Panda, S.K.; Padhi, L.; Leyssen, P.; Liu, M.; Neyts, J.; Luyten, W. Antimicrobial, anthelmintic, and antiviral activity of plants traditionally used for treating infectious disease in the Similipal Biosphere Reserve, Odisha, India. Front. Pharmacol. 2017, 8, 658. [Google Scholar] [CrossRef] [Green Version]
  133. Kumar, S.; Madaan, R.; Bansal, G.; Jamwal, A.; Sharma, A. Plants and plant products with potential anticonvulsant activity–a review. Phcog Commn. 2012, 2, 3–99. [Google Scholar] [CrossRef] [Green Version]
  134. Mishra, A.; Mishra, A.K.; Jha, S. Effect of traditional medicine brahmi vati and bacoside A-rich fraction of Bacopa monnieri on acute pentylenetetrzole-induced seizures, amphetamine-induced model of schizophrenia, and scopolamine-induced memory loss in laboratory animals. Epilepsy Behav. 2018, 80, 144–151. [Google Scholar] [CrossRef]
  135. Khanal, L.N.; Sharma, K.R.; Pokharel, Y.R.; Kalauni, S.K. Assessment of phytochemical, antioxidant and antimicrobial activities of some medicinal plants from Kaski district of Nepal. Am. J. Plant Sci. 2020, 11, 1383. [Google Scholar] [CrossRef]
  136. Singh, M.; Purohit, R. Study on phytochemical characterization and antibacterial activity of fruit trees of Chamoli District, Uttarakhand, India. Int. J. Pharm. Biol. Sci. 2019, 9, 226–231. [Google Scholar]
  137. Rambabu, K.; Bharath, G.; Monash, P.; Velu, S.; Banat, F.; Naushad, M.; Arthanareeswaran, G.; Show, P.L. Effective treatment of dye polluted wastewater using nanoporous CaCl2 modified polyethersulfone membrane. Process Saf. Environ. Prot. 2019, 124, 266–278. [Google Scholar] [CrossRef]
  138. Dhatwalia, J.; Kumari, A.; Chauhan, A.; Mansi, K.; Thakur, S.; Saini, R.V.; Guleria, I.; Lal, S.; Kumar, A.; Batoo, K.M. Rubus ellipticus Sm. Fruit Extract Mediated Zinc Oxide Nanoparticles: A Green Approach for Dye Degradation and Biomedical Applications. Materials 2022, 15, 3470. [Google Scholar] [CrossRef] [PubMed]
  139. Khalil, S.; Mehmood, A.; Khan, M.A.R.; Ahmad, K.S.; Abasi, F.; Raffi, M.; Ali, K.; Khan, M.E.H.; Jones, D.A.; Abdelkarim, M. Antibacterial, antioxidant and photocatalytic activity of novel Rubus ellipticus leaf mediated silver nanoparticles. J. Saudi Chem. Soc. 2023, 27, 101576. [Google Scholar] [CrossRef]
Figure 1. Photographs of different plant parts of Rubus ellipticus Sm. (A) Shrub, (B) flowers, and (C) fruits. (Photo credits: Dipa Paneru (A,C), Sujata Lamichhane (B).)
Figure 1. Photographs of different plant parts of Rubus ellipticus Sm. (A) Shrub, (B) flowers, and (C) fruits. (Photo credits: Dipa Paneru (A,C), Sujata Lamichhane (B).)
Molecules 28 06071 g001
Table 2. Macronutrient composition of fruit of R. ellipticus and R. fruticosus.
Table 2. Macronutrient composition of fruit of R. ellipticus and R. fruticosus.
Nutrient CompositionValues for R. ellipticus Values for R. fruticosus
Protein (%)4.37 ± 0.52% [23]1.39 g/100 g [90]
Fiber (%)2.35 ± 0.05% [39]44.2 % of dry matter [91]
Fat (%) 0.96 ± 0.20% [39]0.49 g/100 g [90]
Carbohydrates (%)86.4 ± 0.38% [23]9.07 ± 0.80% [91]
Ash Value2.97 g/100 g of dry matter [23]3.0 g/100 g of fresh weight [92]
Table 3. Micronutrient composition of fruit of R. ellipticus and R. fruticosus.
Table 3. Micronutrient composition of fruit of R. ellipticus and R. fruticosus.
Nutrient CompositionQuantity on R. ellipticusQuantity on R. fruticosus
Sodium89.43 ± 0.01 mg/100 g DW [23]5.91 mg/100 g [93]
Potassium1.82 ± 0.25 mg/100 g DW [39]8.9 mg/g [31]
Calcium0.95 ± 0.10 mg/100 g DW [39]193mg/100 g [93]
Magnesium118.72 ± 0.48 mg/100 g DW [23]151 mg/100 g [93]
Copper0.020 ± 0.01 mg/100 g DW [23]165 µg/100 g [90]
Zinc12.77 ± 0.05 mg/100 g DW [23]2.3 mg/100 g [93]
Iron4.249 ± 0.15 mg/100 g DW [23]5.9 mg/100 g [93]
Manganese1.948 ± 0.03 mg/100 g DW [23]0.646 mg/100 g [90]
Ascorbic acid (Vitamin C) 19.8 mg/100 g Fresh weight [94]7.1–9.6 mg/100 g [95]
DW: dry weight
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

Lamichhane, A.; Lamichhane, G.; Devkota, H.P. Yellow Himalayan Raspberry (Rubus ellipticus Sm.): Ethnomedicinal, Nutraceutical, and Pharmacological Aspects. Molecules 2023, 28, 6071. https://doi.org/10.3390/molecules28166071

AMA Style

Lamichhane A, Lamichhane G, Devkota HP. Yellow Himalayan Raspberry (Rubus ellipticus Sm.): Ethnomedicinal, Nutraceutical, and Pharmacological Aspects. Molecules. 2023; 28(16):6071. https://doi.org/10.3390/molecules28166071

Chicago/Turabian Style

Lamichhane, Ananda, Gopal Lamichhane, and Hari Prasad Devkota. 2023. "Yellow Himalayan Raspberry (Rubus ellipticus Sm.): Ethnomedicinal, Nutraceutical, and Pharmacological Aspects" Molecules 28, no. 16: 6071. https://doi.org/10.3390/molecules28166071

Article Metrics

Back to TopTop