Next Article in Journal
Low Hemoglobin Levels Are Associated with Reduced Psychomotor and Language Abilities in Young Ugandan Children
Next Article in Special Issue
Metabolic Effects of an Oral Glucose Tolerance Test Compared to the Mixed Meal Tolerance Tests: A Narrative Review
Previous Article in Journal
Leptin Levels of the Perinatal Period Shape Offspring’s Weight Trajectories through the First Year of Age
Previous Article in Special Issue
Dysmetabolism and Neurodegeneration: Trick or Treat?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

D-Pinitol—Active Natural Product from Carob with Notable Insulin Regulation

Carobway Ltd., Nes Ziona 7406520, Israel
Nutrients 2022, 14(7), 1453; https://doi.org/10.3390/nu14071453
Submission received: 3 March 2022 / Revised: 28 March 2022 / Accepted: 29 March 2022 / Published: 30 March 2022
(This article belongs to the Special Issue Impact of Diet Composition on Insulin Resistance)

Abstract

:
Carob is one of the major food trees for peoples of the Mediterranean basin, but it has also been traditionally used for medicinal purposes. Carob contains many nutrients and active natural products, and D-Pinitol is clearly one of the most important of these. D-Pinitol has been reported in dozens of scientific publications and its very diverse medicinal properties are still being studied. Presently, more than thirty medicinal activities of D-Pinitol have been reported. Among these, many publications have reported the strong activities of D-Pinitol as a natural antidiabetic and insulin regulator, but also as an active anti-Alzheimer, anticancer, antioxidant, and anti-inflammatory, and is also immune- and hepato-protective. In this review, we will present a brief introduction of the nutritional and medicinal importance of Carob, both traditionally and as found by modern research. In the introduction, we will present Carob’s major active natural products. The structures of inositols will be presented with a brief literature summary of their medicinal activities, with special attention to those inositols in Carob, as well as D-Pinitol’s chemical structure and its medicinal and other properties. D-Pinitol antidiabetic and insulin regulation activities will be extensively presented, including its proposed mechanism of action. Finally, a discussion followed by the conclusions and future vision will summarize this article.

Graphical Abstract

1. Introduction

1.1. Carob: The Faithful Companion of Humanity

Carob (Ceratonia siliqua L.) is one of the most important nutritional crops for peoples of the Middle East, North Africa, and Southern Europe [1]. Carob fruits (named pods or kibbles), contain a wide range of macro- and micronutrients, as well as many other natural products. A summary of Carob fruit composition is presented in Table 1.
However, since antiquity, humans have used different parts of the Carob tree for many and interesting purposes [3]. Among these, analgesic and anti-inflammatory activities are the most important [4,5]. Most of the traditional medicine uses have utilized different forms of fruits, including unripe pods, but these utilizations included extracts, decoctions and infusions of leaves and bark [6].
Modern research followed traditional knowledge and dozens of studies were published to date about dozens of medicinal activities of Carob’s various products, including its extracts and single natural products. Consequently, many review articles that summarize the research articles can also be found [7,8,9,10]. However, one of the best review articles about Carob’s composition has been published by K. Rtibi et al., where they focus on Carob-derived treatments of the gastrointestinal tract [11]. In Figure 1, major and new (red names) phenolic compounds are shown [12,13,14].
To conclude this section, it is important to indicate that in recent years there has emerged a rapidly growing interest in Carob seeds, their composition (protein rich), nutritional potential and medicinal activities [2,15,16].

1.2. Insulin Resistance in Type 2 Diabetes

Type 2 diabetes (T2D) is defined by the World Health Organization (WHO) as a “metabolic disorder of multiple etiology characterized by chronic hyperglycemia with disturbance of carbohydrate, fat, and protein metabolism resulting from defects in insulin secretion, insulin action, or both” [17]. The International Diabetes Federation reported that in 2018, there were 463 million people around the world affected by this disease, and the organization estimates that by 2045, there will be 700 million people affected by it [18]. It has also been reported that in 2017, the global healthcare expenditure associated with diabetes and its complications was USD 850 billion. The prevalence rate is estimated as 13.5% in low-income countries, compared with 10.4% in high-income nations. It is interesting to mention the fact that this trend is also found within different ethnicities in the same country. In the USA, the ethnic distribution of T2D follows the “rule” higher-income-lower-diabetes: Non-Hispanic Whites (highest income) 7.6%, Asians 9%, Hispanics 12.8%, and African Americans (lowest income) 13.2%, in 2017 [19]. In Israel, the author’s home country, there are 12% of diabetics among Arabs (lower income) and 6.2% among Jews (higher income) [20].
Therefore, in the abovementioned definition of T2D, insulin plays a critical role, and “insulin resistance” is the major cause of this disease. This health disorder is defined as: “a defect in insulin-mediated control of glucose metabolism in tissues—prominently in muscle, fat and liver” [17], but insulin has various functions in the human body, and they are presented in Table 2 [21].
The mechanism of action of insulin in healthy conditions can be found in many publications [22], and a simplified illustration of it is shown in Figure 2.
Insulin enters the cell through an insulin receptor. As a result, tyrosine (Tyr) phosphorylation occurs on the insulin receptor substrate (IRS) protein. The resulting adduct activates phosphoinositide 3-kinase (PI3K), resulting in activation of phosphoinositide-dependent kinase-1,2 (PDK1/2). Protein kinase (AKT) gets phosphorylated by PDK1/2 and promotes glucose transporter 4 (GLUT4) translocation to plasma membrane and facilitates glucose into cell. Thioredoxin interacting protein (TXNIP) inhibits is blocked.
Numerous research articles have been published about this key factor of T2D, and dozens of review articles that summarize these research publications. However, it is important to understand the possible mechanisms of insulin resistance that were also presented in most of these scientific publications. One of the most comprehensive and illustrated review articles was published by M.C. Petersen and G.I. Shulman [23]. Insulin resistance is discussed as major and sub-major types, where each section is illustrated with many figures and graphs.
The review article of H. Yaribeygi et al. follows the previous reference, though it is far less comprehensive [24]. However, one of its clearest advantages is the table that summarizes the molecular mechanisms that are involved in insulin resistance (page 6 in Ref. [24]), and it is partially cited here as shown in Table 3.
The review article of D.E. James has special importance for two major reasons [26]. First, it includes excellent figures that explain the putative factors that contribute to insulin resistance (Figure 4, page 12 in Ref. [26]). Second, it discusses the situation of fasting in insulin resistance conditions. This situation has great relevance for hundreds of millions of people around the world. Another review article with special importance about insulin resistance has been recently published by W.A. Banks and E.M. Rhea [27]. This article is important for three major reasons. First, it links insulin resistance with the brain–blood barrier (BBB); second, it discusses the relation of insulin signaling and oxidative stress manifestation in T2D and Alzheimer’s disease; and third, it contains excellent illustrations, especially the figure that shows the interactions between insulin and oxidative stress.

1.3. Treatment of Insulin Resistance with Natural Products

As mentioned in the previous section, T2D is a severe global health issue and a major cause of financial burden. Consequently, many methods have been developed to target this disorder. However, before presenting treatments that are based on natural products, we will briefly present some selected synthetic pharmaceuticals.
C.L. Reading et al. have reported the anti-inflammatory activity and improvement of the insulin-sensitivity activity of synthetic sterol (Figure 3) in insulin-resistant obese-impaired glucose tolerance [28].
A significantly different approach has been reported by S. Xue et al. who report a treatment for hepatogenous diabetes using Oleanolic acid, which triggered the expression of short-peptide genetic synthesis [30]. The synergistic activity of Oleanolic acid and the peptide (researchers have named it shGLP-1), proved to be more efficient than the activity of each component separately. To conclude this part, we indicate the review article of R. Vieira et al. which is very informative and comprehensive [31].
Many natural products have been tested and published for their insulin regulation activity. F.S. Saadeldeen et al. list in their excellent review article 98 naturally occurring compounds that regulate glucose metabolism and treat insulin resistance [32]. This article provides the structure of each compound, its botanical source, and its activity.
Following traditional Chinese medicine therapeutic methods, J. Li et al. list pure natural products and herbal formulations used to treat insulin resistance [33]. Formulations are listed with their Chinese names, and detailed information about methods and purposes of use.
In addition to D-Pinitol, which will be discussed in Section 3, numerous natural products have been published in research articles for having insulin regulation activity. We limit our presentation here to two of these compounds that have been mentioned in very recent publications. First, R. Alaaeldin et al. reported the amelioration of insulin resistance of Carpachromene (Figure 4), a natural product that can be found in Banyan (Ficus binghalensis L.) [34].
They found (in vitro model) that Carpachromene has significant insulin resistance amelioration compared with Metformin, a synthetic drug widely used for treating this disorder.
The second report was published by A. Deenadayalan et al. who tested the effect of Stevioside (Figure 5) on insulin resistance, in both in vivo (rats) and in silico models [35].
Their findings indicate that this compound has similar activity to metformin.
Finally, it is important to mention very recent research published by H. Sanz-Lamora et al. that found that treatment with pure polyphenol supplementation (D18060501) worsened insulin resistance in diet-induced obese mice [36].

2. Inositols—A Brief Presentation

Inositols are naturally occurring Cyclitols or Polyols, and they can be found in mammalian and plant kingdoms [37]. In terms of more specific chemical structure, these natural products are stereoisomers of hexahydroxy cyclohexane. In Figure 6, the structures of naturally occurring inositols are shown.
The biological properties of inositols have been extensively studied and published. Most of these activities have been summarized by O.C. Watkins et al. [38]. These properties include insulin regulation, antidiabetic, antioxidant, antibacterial, female fertility enhancer, metabolic syndrome treatment, antidepressant, gastroprotective, hepatoprotective, hypolipidemic and antiaging. However, in this review and in most published literature about the properties of these compounds, it is clear that most studies have focused on two activities: insulin regulation and treatment of female fertility disorders. In Table 4 we cite some of these notable publications, in chronological order.
From Carob, six inositols and their derivatives (methyl ethers) were isolated and characterized [61]. Their structures are shown in Figure 7.
myo-Inositol is the most abundant compound of this family in all life forms, followed by D-Pinitol and its precursor, D-chiro-Inositol, in the plant kingdom. D-Bornesitol and D-Sequoyitol are relatively rare, and their properties are almost unknown. D-Ononitol has been very limitedly studied [62,63].

3. D-Pinitol: Occurrence, Isolation, and Properties

D-Pinitol can be found in more than 20 plant sources, and its highest content is in Carob pods, at 5.5% [2,64]. To date, more than 40 publications have reported the quantification and/or isolation of this important natural product. One of the most notable works has been published by O. Negishi et al. [65]. They determined the content of methylated inositols in 43 edible plants by the HPAE-PAD analytical method. J. Qiu et al. reported the determination of D-Pinitol in rat plasma [66]. This study is highly important since it provides understanding of the pharmacokinetics and bioavailability of D-Pinitol in vivo.
The medicinal and other properties of D-Pinitol have been extensively studied and published. In Table 5, we list most of these reports, excluding publications that report no or low results.

4. D-Pinitol as Insulin Regulator

In Section 3, we cited eight important published studies about the activity of D-Pinitol as insulin regulator (Table 5). In fact, the number of publications about this topic is much higher, and many review articles have published about it and other medicinal properties of D-Pinitol. These review articles and the research publications that they cite, conclude that D-Pinitol has two mechanisms of action as an insulin regulator [152]: insulin sensitizing and insulin mimetic.
K. Srivastava et al. present the insulin-sensitizing effect of D-Pinitol in their review article about this natural product [153], and a simplified illustration of this effect is shown in Figure 8.
Interestingly, in a table that lists the botanical sources of D-Pinitol in Ref. [153] (page 3), the authors do not mention the three plants with the highest content of this natural product: Carob, Bougainvillea and Soybean [64].
T. Antonowski et al. present the insulin-like (insulin-mimetic) activity of D-Pinitol [154]. This publication, and others, demonstrates the simplified mechanism shown in Figure 9.
This minireview article is notably useful for understanding the structures of cyclitols and their role in ameliorating metabolic syndrome and diabetes.

5. Discussion

D-Pinitol is a naturally occurring inositol that can be found in many plant species. Carob has the highest content of D-Pinitol, which has a wide range of medicinal and other properties (Section 3). One of these, and probably the most important, is insulin regulation, which has two major mechanisms: insulin-sensitizing and insulin-mimetic [152].
Many natural products have one or both properties of insulin regulation, including plant extracts and other mixed compounds. For example, S.A. Kalekar et al. have reported on the in vitro insulin-sensitizing activity of hydroethanolic extracts of three plants: Phyllanthus emblica L., Tinospora cordifolia (Thunb.) Miers and Curcuma longa L. [155]. In a more recent study, V. Stadlbauer et al. tested more than 600 plant extracts and found three of them to have clear in vivo insulin-mimetic activity: Xysmalobium undulatum L., Sapindus mukorossi L., Chelidonium majus L. [156]. It is important to mention that in this study Carob is not included.
Despite the abovementioned, D-Pinitol, and D-Pinitol-containing products of Carob, have several advantages over other insulin-regulating plant products, due to the following reasons:
(A)
D-Pinitol content of Carob (pods) is the highest of all plants [64].
(B)
D-Pinitol-containing products of Carob such as molasses, have important health benefits [157].
(C)
Compared with most other natural products that have insulin-regulation activity, such as polyphenols, D-Pinitol is more stable in biological gastric conditions [48]. This property increases its bioavailability in the human body.
(D)
In addition to that which is mentioned in C, D-Pinitol is generally stable, but even if it undergoes methoxy group hydrolysis, the resulting compound is chiro-Inositol, which is an active insulin-regulator as well [158]. See Figure 6.
(E)
Even though there is a limited number of studies that indicate it, it is evident that D-Pinitol’s activities are significantly increased when it synergistically acts with other natural products [25,92,150,159].
(F)
D-Pinitol has wide range of medicinal activities (Table 5), so it is a multi-functional natural product. This property increases its potential as a drug.

6. Conclusions and Future Horizons

Most of the medicinal properties of D-Pinitol have been studied and published. Some of these have been extensively investigated, while others were limitedly or even not published. It is very important to conduct further studies of all activities of D-Pinitol, but activities such as insulin regulation, anti-Alzheimer, antiaging and possible anti-Parkinson activities must draw more attention.
The synergistic effect of D-Pinitol with other natural products of Carob and other plants is in its beginnings, so this topic must also be thoroughly studied.
Our group is currently investigating some known and unpublished activities of D-Pinitol, and we are examining possible clinical and other applications that will hopefully lead to healthy foods, food-additives, and other healthy products.

Funding

This research received no external funding.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Azab, A. Carob (Ceratonia siliqua): Super Food and Medicine. Literature Update. Eur. Chem. Bull. 2020, 9, 306–312. [Google Scholar] [CrossRef]
  2. Brassesco, M.E.; Brandao, T.; Silva, C.; Pintado, M. Carob bean (Ceratonia siliqua L.): A new perspective for functional food. Trends Food Sci. Technol. 2021, 114, 310–322. [Google Scholar] [CrossRef]
  3. Azab, A. Carob (Ceratonia siliqua): Health, medicine and chemistry. Eur. Chem. Bull. 2017, 6, 456–469. [Google Scholar] [CrossRef] [Green Version]
  4. Ben Ayache, S.; Behija Saafi, E.; Emhemmed, F.; Flamini, G.; Achour, L.; Muller, C.D. Biological activities of aqueous extracts from Carob plant (Ceratonia siliqua L.) by antioxidant, analgesic and proapoptotic properties evaluation. Molecules 2020, 25, 3120. [Google Scholar] [CrossRef]
  5. Khalifa, A.B. Herbs: Nature’s Pharmacy, 1st ed.; Arab Cultural Center: Casablanca, Morocco, 2004; pp. 286–288. [Google Scholar]
  6. Saad, B.; Said, O. Greco-Arab and Islamic Herbal Medicine; John Wiley & Sons, Inc.: Hoboken, NJ, USA, 2011; p. 308. [Google Scholar] [CrossRef]
  7. Nasar-Abbas, S.M.; E-Huma, Z.; Vu, T.H.; Khan, M.K.; Esbenshade, H.; Jayasena, V. Carob kibble: A bioactive-rich food ingredient. Compr. Rev. Food Sci. Food Saf. 2016, 15, 63–72. [Google Scholar] [CrossRef] [Green Version]
  8. Stavrou, I.J.; Christou, A.; Kapnissi-Christodoulou, C.P. Polyphenols in carobs: A review on their composition, antioxidant capacity and cytotoxic effects, and health impact. Food Chem. 2018, 269, 355–374. [Google Scholar] [CrossRef]
  9. Lakkab, I.; El Hajaji, H.; Lachkar, N.; El Bali, B.; Lachkar, M.; Ciobica, A. Phytochemistry, bioactivity: Suggestion of Ceratonia siliqua L. as neurodegenerative disease therapy. J. Complement. Integr. Med. 2018, 15, 20180013. [Google Scholar] [CrossRef]
  10. Zhu, B.J.; Zayed, M.Z.; Zhu, H.-X.; Zhao, J.; Li, S.-P. Functional polysaccharides of carob fruit: A review. Chin. Med. 2019, 14, 40. [Google Scholar] [CrossRef] [Green Version]
  11. Rtibi, K.; Selmi, S.; Grami, D.; Amri, M.; Eto, B.; El-Benna, J.; Sebai, H.; Marzouki, L. Chemical constituents and pharmacological actions of carob pods and leaves (Ceratonia siliqua L.) on the gastrointestinal tract: A review. Biomed. Pharmacother. 2017, 93, 522–528. [Google Scholar] [CrossRef]
  12. Papagiannopoulos, M.; Wollseifen, H.R.; Mellenthin, A.; Haber, B.; Galensa, R. Identification and quantification of polyphenols in carob fruits (Ceratonia siliqua L.) and derived products by HPLC-UV-ESI/MS. J. Agric. Food Chem. 2004, 52, 3784–3791. [Google Scholar] [CrossRef]
  13. Gohar, A.; Gedara, S.R.; Baraka, H.N. New acylated flavonol glycoside from Ceratonia siliqua L. seeds. J. Med. Plants Res. 2009, 3, 424–428. [Google Scholar] [CrossRef]
  14. Cavdarova, M.; Makris, D.P. Extraction Kinetics of Phenolics from Carob (Ceratonia siliqua L.) Kibbles Using Environmentally Benign Solvents. Waste Biomass Valori. 2014, 5, 773–779. [Google Scholar] [CrossRef]
  15. Benković, M.; Bosiljkov, T.; Semić, A.; Ježek, D.; Srečec, S. Influence of Carob Flour and Carob Bean Gum on Rheological Properties of Cocoa and Carob Pastry Fillings. Foods 2019, 8, 66. [Google Scholar] [CrossRef] [Green Version]
  16. Santonocito, D.; Granata, G.; Geraci, C.; Panico, A.; Siciliano, E.A.; Raciti, G.; Puglia, C. Carob Seeds: Food Waste or Source of Bioactive Compounds? Pharmaceutics 2020, 12, 1090. [Google Scholar] [CrossRef]
  17. Reed, J.; Bain, S.; Kanamarlapudi, V. A Review of Current Trends with Type 2 Diabetes Epidemiology, Aetiology, Pathogenesis, Treatments and Future Perspectives. Diabetes Metab. Syndr. Obes. 2021, 14, 3567–3602. [Google Scholar] [CrossRef]
  18. Ganasegeran, K.; Hor, C.P.; Jamil, M.F.; Loh, H.C.; Noor, J.M.; Hamid, N.A.; Suppiah, P.D.; Abdul Manaf, M.R.; Ch’ng, A.S.; Looi, I. A Systematic Review of the Economic Burden of Type 2 Diabetes in Malaysia. Int. J. Environ. Res. Public Health 2020, 17, 5723. [Google Scholar] [CrossRef]
  19. Rodríguez, J.E.; Campbell, K.M. Racial and Ethnic Disparities in Prevalence and Care of Patients with Type 2 Diabetes. Clin. Diabetes 2017, 35, 66–70. [Google Scholar] [CrossRef] [Green Version]
  20. Peleg, O. The Relationship between Type 2 Diabetes, Differentiation of Self, and Emotional Distress: Jews and Arabs in Israel. Nutrients 2022, 14, 39. [Google Scholar] [CrossRef]
  21. Kahn, C.R. The molecular mechanism of insulin action. Annu. Rev. Med. 1985, 36, 429–451. [Google Scholar] [CrossRef]
  22. Ingle, P.V.; Yin, S.B.; Ying, B.J.; Leong, B.K.; Xin, T.Z.; Hwa, L.T.; Mun, L.T. Current Trends in Pharmacological Treatment of Type II Diabetes Mellitus. Int. J. Pharm. Res. Rev. 2018, 7, 1–15. [Google Scholar]
  23. Petersen, M.C.; Shulman, G.I. Mechanisms of Insulin Action and Insulin Resistance. Physiol. Rev. 2018, 98, 2133–2223. [Google Scholar] [CrossRef] [Green Version]
  24. Yaribeygi, H.; Farrokhi, F.R.; Butler, A.E.; Sahebkar, A. Insulin resistance: Review of the underlying molecular mechanisms. J. Cell Physiol. 2019, 234, 8152–8161. [Google Scholar] [CrossRef]
  25. Ahmad, F.; Misra, L.; Gupta, V.K.; Darokar, M.P.; Prakash, O.; Khan, F.; Shukla, R. Synergistic effect of (+)-pinitol from Saraca asoca with β-lactam antibiotics and studies on the in silico possible mechanism. J. Kor. Diabetes Assoc. 2005, 29, 344–351, Reprinted in J. Assian Nat. Prod. Res. 2015, 18, 72–183. [Google Scholar] [CrossRef]
  26. James, D.E.; Stöckli, J.; Birnbaum, M.J. The aetiology and molecular landscape of insulin resistance. Nat. Rev. Mol. Cell Biol. 2021, 22, 751–771. [Google Scholar] [CrossRef]
  27. Banks, W.A.; Rhea, E.M. The Blood-Brain Barrier, Oxidative Stress, and Insulin Resistance. Antioxidants 2021, 10, 1695. [Google Scholar] [CrossRef]
  28. Reading, C.L.; Stickney, D.R.; Flores-Riveros, J.; Destiche, D.A.; Ahlem, C.N.; Cefalu, W.T.; Frincke, J.M. A synthetic anti-inflammatory sterol improves insulin sensitivity in insulin-resistant obese impaired glucose tolerance subjects. Obesity 2013, 21, E343–E349. [Google Scholar] [CrossRef]
  29. Ravindran, R.; Mitra, K.; Arumugam, S.K.; Doble, M. Preparation of Curdlan sulphate—Chitosan nanoparticles as a drug carrier to target Mycobacterium smegmatis infected macrophages. Carbohydr. Polym. 2021, 258, 117686. [Google Scholar] [CrossRef]
  30. Xue, S.; Yin, J.; Shao, J.; Yu, Y.; Yang, L.; Wang, Y.; Xie, M.; Fussenegger, M.; Ye, H. A Synthetic-Biology-Inspired Therapeutic Strategy for Targeting and Treating Hepatogenous Diabetes. Mol. Ther. 2017, 25, 443–455. [Google Scholar] [CrossRef] [Green Version]
  31. Vieira, R.; Souto, S.B.; Sánchez-López, E.; Machado, A.L.; Severino, P.; Jose, S.; Santini, A.; Fortuna, A.; García, M.L.; Silva, A.M.; et al. Sugar-Lowering Drugs for Type 2 Diabetes Mellitus and Metabolic Syndrome-Review of Classical and New Compounds: Part-I. Pharmaceuticals 2019, 12, 152. [Google Scholar] [CrossRef] [Green Version]
  32. Saadeldeen, F.S.; Niu, Y.; Wang, H.; Zhou, L.; Meng, L.; Chen, S.; Sun-Waterhouse, D.; Waterhouse, G.I.; Liu, Z.; Kang, W. Natural Products: Regulating Glucose Metabolism and Improving Insulin Resistance. Food Sci. Hum. Wellness 2020, 9, 214–228. [Google Scholar] [CrossRef]
  33. Li, J.; Bai, L.; Wei, F.; Zhao, J.; Wang, D.; Xiao, Y.; Yan, W.; Wei, J. Therapeutic Mechanisms of Herbal Medicines against Insulin Resistance: A Review. Front. Pharmacol. 2019, 10, 661. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Alaaeldin, R.; Abdel-Rahman, I.A.; Hassan, H.A.; Youssef, N.; Allam, A.E.; Abdelwahab, S.F.; Zhao, Q.L.; Fathy, M. Carpachromene Ameliorates Insulin Resistance in HepG2 Cells via Modulating IR/IRS1/PI3k/Akt/GSK3/FoxO1 Pathway. Molecules 2021, 26, 7629. [Google Scholar] [CrossRef] [PubMed]
  35. Deenadayalan, A.; Subramanian, V.; Paramasivan, V.; Veeraraghavan, V.P.; Rengasamy, G.; Sadagopan, J.C.; Rajagopal, P.; Jayaraman, S. Stevioside Attenuates Insulin Resistance in Skeletal Muscle by Facilitating IR/IRS-1/Akt/GLUT 4 Signaling Pathways: An In Vivo and In Silico Approach. Molecules 2021, 26, 7689. [Google Scholar] [CrossRef] [PubMed]
  36. Sanz-Lamora, H.; Marrero, P.F.; Haro, D.; Relat, J. A Mixture of Pure, Isolated Polyphenols Worsens the Insulin Resistance and Induces Kidney and Liver Fibrosis Markers in Diet-Induced Obese Mice. Antioxidants 2022, 11, 120. [Google Scholar] [CrossRef] [PubMed]
  37. Hoffmann-Ostenhof, O.; Pittner, F. The biosynthesis of myo-inositol and its isomers. Can. J. Chem. 1982, 60, 1863–1871. [Google Scholar] [CrossRef]
  38. Watkins, O.C.; Yong, H.E.; Sharma, N.; Chan, S.-Y. A review of the role of inositols in conditions of insulin dysregulation and in uncomplicated and pathological pregnancy. Crit. Rev. Food Sci. Nutr. 2022, 62, 1626–1673. [Google Scholar] [CrossRef]
  39. Kennington, A.S.; Hill, C.R.; Craig, J.; Bogardus, C.; Raz, I.; Ortmeyer, H.K.; Hansen, B.C.; Romero, G.; Larner, J. Low urinary chiro-inositol excretion in non-insulin-dependent diabetes mellitus. N. Engl. J. Med. 1990, 323, 373–378. [Google Scholar] [CrossRef]
  40. Hallman, M.; Bry, K.; Hoppu, K.; Lappi, M.; Pohjavuori, M. Inositol supplementation in premature infants with respiratory distress syndrome. N. Engl. J. Med. 1992, 326, 1233–1239. [Google Scholar] [CrossRef]
  41. Asplin, I.; Galasko, G.; Larner, J. Chiro-inositol deficiency and insulin resistance: A comparison of the chiro-inositol- and the myo-inositol-containing insulin mediators isolated from urine, hemodialysate, and muscle of control and type II diabetic subjects. Proc. Natl. Acad. Sci. USA 1993, 90, 5924–5928. [Google Scholar] [CrossRef] [Green Version]
  42. Levine, J. Controlled trials of inositol in psychiatry. Eur. Neuropsychopharmacol. 1997, 7, 147–155. [Google Scholar] [CrossRef]
  43. Nestler, J.E.; Jakubowicz, D.J.; Reamer, P.; Gunn, R.D.; Allan, G. Ovulatory and metabolic effects of D-chiro-inositol in the polycystic ovary syndrome. N. Engl. J. Med. 1999, 340, 1314–1320. [Google Scholar] [CrossRef] [Green Version]
  44. McLaurin, J.; Golomb, R.; Jurewicz, A.; Antel, J.P.; Fraser, P.E. Inositol stereoisomers stabilize an oligomeric aggregate of Alzheimer amyloid beta peptide and inhibit Aβ-induced toxicity. J. Biol. Chem. 2000, 275, 18495–18502. [Google Scholar] [CrossRef] [Green Version]
  45. Jung, T.S.; Hahm, J.R.; Kim, J.J.; Jung, J.H.; Kang, M.Y.; Moon, S.W.; Lee, K.W.; Kim, H.C.; Lee, J.D.; Kim, J.H.; et al. Determination of urinary Myo-/chiro-inositol ratios from Korean diabetes patients. Yonsei Med. J. 2005, 46, 532–538. [Google Scholar] [CrossRef] [Green Version]
  46. Nascimento, N.R.; Lessa, L.M.; Kerntopf, M.R.; Sousa, C.M.; Alves, R.S.; Queiroz, M.G.; Price, J.; Heimark, D.B.; Larner, J.; Du, X.; et al. Inositols prevent and reverse endothelial dysfunction in diabetic rat and rabbit vasculature metabolically and by scavenging superoxide. Proc. Natl. Acad. Sci. USA 2006, 103, 218–223. [Google Scholar] [CrossRef] [Green Version]
  47. Michell, R.H. Evolution of the diverse biological roles of inositols. Biochem. Soc. Symp. 2007, 74, 223–246. [Google Scholar] [CrossRef] [Green Version]
  48. Michell, R.H. Inositol derivatives: Evolution and functions. Nat. Rev. Mol. Cell Biol. 2008, 9, 151–161. [Google Scholar] [CrossRef]
  49. Bizzarri, M.; Carlomagno, G. Inositol: History of an effective therapy for Polycystic Ovary Syndrome. Eur. Rev. Med. Pharmacol. Sci. 2014, 18, 1896–1903. [Google Scholar] [PubMed]
  50. Mancini, M.; Andreassi, A.; Salvioni, M.; Pelliccione, F.; Mantellassi, G.; Banderali, G. Myoinositol and D-Chiro Inositol in Improving Insulin Resistance in Obese Male Children: Preliminary Data. Int. J. Endocrinol. 2016, 8720342. [Google Scholar] [CrossRef] [Green Version]
  51. Kalra, B.; Kalra, S.; Sharma, J.B. The inositols and polycystic ovary syndrome. Indian J. Endocrinol. Metab. 2016, 20, 720–724. [Google Scholar] [CrossRef]
  52. Hanna, R.; Wehbe, T.; Abou Jaoude, E. Metabolic Effects of D-Chiro-Inositol and Myo-Inositol in Polycystic Ovary Syndrome. Int. J. Clin. Endocrinol. Metab. 2017, 3, 029–033. [Google Scholar] [CrossRef]
  53. Orrù, B.; Circo, R.; Logoteta, P.; Petousis, S.; Carlomagno, G. Finding the best therapeutic approach for PCOS: The importance of inositol(s) bioavailability. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 83–88. [Google Scholar] [PubMed]
  54. Showell, M.G.; Mackenzie-Proctor, R.; Jordan, V.; Hodgson, R.; Farquhar, C. Inositol for subfertile women with polycystic ovary syndrome. Cochrane Database Syst. Rev. 2018, 12, CD012378. [Google Scholar] [CrossRef]
  55. Miñambres, I.; Cuixart, G.; Gonçalves, A.; Corcoy, R. Effects of inositol on glucose homeostasis: Systematic review and meta-analysis of randomized controlled trials. Clin. Nutr. 2019, 38, 1146–1152. [Google Scholar] [CrossRef]
  56. Chhetri, D.R. Myo-Inositol and Its Derivatives: Their Emerging Role in the Treatment of Human Diseases. Front Pharmacol. 2019, 10, 1172. [Google Scholar] [CrossRef] [Green Version]
  57. Roseff, S.; Montenegro, M. Inositol Treatment for PCOS Should Be Science-Based and Not Arbitrary. Int. J. Endocrinol. 2020, 2020, 6461254. [Google Scholar] [CrossRef] [Green Version]
  58. Merviel, P.; James, P.; Bouée, S.; Le Guillou, M.; Rince, C.; Nachtergaele, C.; Kerla, V. Impact of myo-inositol treatment in women with polycystic ovary syndrome in assisted reproductive technologies. Reprod. Health 2021, 18, 13. [Google Scholar] [CrossRef] [PubMed]
  59. Sharma, N.; Watkins, O.C.; Chu, A.H.; Cutfield, W.; Godfrey, K.M.; Yong, H.E.; Chan, S.Y. Myo-inositol: A potential prophylaxis against premature onset of labour and preterm birth. Nutr. Res. Rev. 2021, 34, 1–9. [Google Scholar] [CrossRef] [PubMed]
  60. Cantelmi, T.; Lambiase, E.; Unfer, V.R.; Gambioli, R.; Unfer, V. Inositol treatment for psychological symptoms in Polycystic Ovary Syndrome women. Eur. Rev. Med. Pharmacol Sci. 2021, 25, 2383–2389. [Google Scholar] [CrossRef] [PubMed]
  61. Goulas, V.; Stylos, E.; Chatziathanasiadou, M.V.; Mavromoustakos, T.; Tzakos, A.G. Functional Components of Carob Fruit: Linking the Chemical and Biological Space. Int. J. Mol. Sci. 2016, 17, 1875. [Google Scholar] [CrossRef] [PubMed]
  62. Skøt, L.; Egsgaard, H. Identification of ononitol and O-methyl-scyllo-inositol in pea root nodules. Planta 1984, 161, 32–36. [Google Scholar] [CrossRef]
  63. Sheveleva, E.; Chmara, W.; Bohnert, H.J.; Jensen, R.G. Increased Salt and Drought Tolerance by D-Ononitol Production in Transgenic Nicotiana tabacum L. Plant Physiol. 1997, 115, 1211–1219. [Google Scholar] [CrossRef] [Green Version]
  64. Kim, J.I.; Kim, J.C.; Joo, H.J.; Jung, S.H.; Kim, J.J. Determination of total chiro-inositol content in selected natural materials and evaluation of the antihyperglycemic effect of pinitol isolated from soybean and carob. Food Sci. Biotechnol. 2005, 14, 441–445. [Google Scholar]
  65. Negishi, O.; Mun’im, A.; Negishi, Y. Content of methylated inositols in familiar edible plants. J. Agric. Food Chem. 2015, 63, 2683–2688. [Google Scholar] [CrossRef]
  66. Qiu, J.; Yan, X.; Liao, Y.; Yu, D.; Wen, C.; Xiang, Z. An UPLC-MS/MS method for quantification of D-pinitol in rat plasma and its application to a pharmacokinetic and bioavailability study. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 2021, 1163, 122498. [Google Scholar] [CrossRef]
  67. Fenili, D.; Weng, Y.Q.; Aubert, I.; Nitz, M.; McLaurin, J. Sodium/myo-Inositol transporters: Substrate transport requirements and regional brain expression in the TgCRND8 mouse model of amyloid pathology. PLoS ONE 2011, 6, e24032. [Google Scholar] [CrossRef] [Green Version]
  68. Pitt, J.; Thorner, M.; Brautigan, D.; Larner, J.; Klein, W.L. Protection against the synaptic targeting and toxicity of Alzheimer’s-associated Aβ oligomers by insulin mimetic chiro-inositols. FASEB J. 2013, 27, 199–207. [Google Scholar] [CrossRef] [Green Version]
  69. Griñán-Ferré, C.; Bellver-Sanchis, A.; Olivares-Martín, M.; Bañuelos-Hortigüela, O.; Pallàs, M. Synergistic Neuroprotective Effects of a Natural Product Mixture against AD Hallmarks and Cognitive Decline in Caenorhabditis elegans and an SAMP8 Mice Model. Nutrients 2021, 13, 2411. [Google Scholar] [CrossRef]
  70. Hada, B.; Yoo, M.R.; Seong, K.M.; Jin, Y.W.; Myeong, H.K.; Min, K.J. D-chiro-inositol and pinitol extend the life span of Drosophila melanogaster. J. Gerontol. A Biol. Sci. Med. Sci. 2013, 68, 226–234. [Google Scholar] [CrossRef] [Green Version]
  71. Ravindran, R.; Chakrapani, G.; Mitra, K.; Doble, M. Inhibitory activity of traditional plants against Mycobacterium smegmatis and their action on Filamenting temperature sensitive mutant Z (FtsZ)-A cell division protein. PLoS ONE 2020, 15, e0232482. [Google Scholar] [CrossRef]
  72. Sethi, G.; Ahn, K.S.; Sung, B.; Aggarwal, B.B. Pinitol targets nuclear factor-kappaB activation pathway leading to inhibition of gene products associated with proliferation, apoptosis, invasion, and angiogenesis. Mol. Cancer Ther. 2008, 7, 1604–1614. [Google Scholar] [CrossRef] [Green Version]
  73. Lin, T.H.; Tan, T.W.; Tsai, T.H.; Chen, C.C.; Hsieh, T.F.; Lee, S.S.; Liu, H.H.; Chen, W.C.; Tang, C.H. D-pinitol inhibits prostate cancer metastasis through inhibition of αVβ3 integrin by modulating FAK, c-Src and NF-κB pathways. Int. J. Mol. Sci. 2013, 14, 9790–9802. [Google Scholar] [CrossRef] [PubMed]
  74. Rengarajan, T.; Nandakumar, N.; Rajendran, P.; Haribabu, L.; Nishigaki, I.; Balasubramanian, M.P. D-pinitol promotes apoptosis in MCF-7 cells via induction of p53 and Bax and inhibition of Bcl-2 and NF-κB. Asian Pac. J. Cancer Prev. 2014, 15, 1757–1762. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Jayasooriya, R.G.; Kang, G.-H.; Park, S.R.; Choi, Y.-H.; Kim, G.-Y. Pinitol Suppresses Tumor Necrosis Factor-α-Induced Invasion of Prostate Cancer LNCaP Cells by Inhibiting Nuclear Factor-κB-Mediated Matrix Metalloproteinase-9 Expression. Trop. J. Pharm. Res. 2015, 14, 1357–1364. [Google Scholar] [CrossRef] [Green Version]
  76. Shin, H.-C.; Bang, T.-H.; Kang, H.-M.; Park, B.-S.; Kim, I.-R. Anticancer effects of D-pinitol in human oral squamous carcinoma cells. Int. J. Oral Biol. 2020, 45, 152–161. [Google Scholar] [CrossRef]
  77. Yao, X.; Shi, K.; Yang, Y.; Gu, X.; Tan, W.; Wang, Q.; Gao, X.; Veeraraghavan, V.P.; Mohan, S.K.; Jin, S. D-Pinitol treatment induced the apoptosis in human leukemia MOLT-4 cells by improved apoptotic signaling pathway. Saudi J. Biol. Sci. 2020, 27, 2134–2138. [Google Scholar] [CrossRef]
  78. Rengarajan, T.; Jagadeesan, A.J.; Balamurugan, A.; Balasubramanian, M.P. Chemotherapeutic potential of D-Pinitol against 7, 12 dimethylbenz (a) (DMBA) induced mammary carcinoma in Sprague Dawley rats. Int. J. Pharm. BioSci. 2011, 2, 232–241. [Google Scholar]
  79. Rengarajan, T.; Nandakumar, N.; Balasubramanian, M.P. D-Pinitol a low-molecular cyclitol prevents 7,12-Dimethylbenz a anthracene induced experimental breast cancer through regulating anti-apoptotic protein Bcl-2, mitochondrial and carbohydrate key metabolizing enzymes. Biomed. Prevent. Nutr. 2012, 2, 25–30. [Google Scholar] [CrossRef]
  80. Rengarajan, T.; Nandakumar, N.; Balasubramanian, M.P. D-Pinitol attenuates 7, 12 dimethylbenz a anthracene induced hazards through modulating protein bound carbohydrates, adenosine triphosphatases and lysosomal enzymes during experimental mammary carcinogenesis. J. Exp. Ther. Oncol. 2012, 10, 39–49. [Google Scholar]
  81. Rengarajan, T.; Nandakumar, N.; Balasubramanian, M.P. D-Pinitol prevents rat breast carcinogenesis induced by 7, 12 -Dimethylbenz aanthracene through inhibition of Bcl-2 and induction of p53, caspase-3 proteins and modulation of hepatic biotransformation enzymes and antioxidants. Biomed. Prevent. Nutr. 2013, 3, 31–41. [Google Scholar] [CrossRef]
  82. Venkatachalam, S.; Boobathi, L.; Balasubramanian, B.M. D-Pinitol Prevents Rat Colon Carcinogenesis Induced by Azoxymethane through Free Radical Formation Induced Cell Damage and Affects Enzymes and Antioxidants. Res. J. Pharm. Technol. 2014, 7, 845–849. [Google Scholar]
  83. Rengarajan, T.; Nandakumar, N.; Rajendran, P.; Ganesh, M.K.; Balasubramanian, M.P.; Nishigaki, I. D-pinitol mitigates tumor growth by modulating interleukins and hormones and induces apoptosis in rat breast carcinogenesis through inhibition of NF-κB. J. Physiol. Biochem. 2015, 71, 191–204. [Google Scholar] [CrossRef]
  84. Lin, Y.; Wu, Y.; Su, J.; Wang, M.; Wu, X.; Su, Z.; Yi, X.; Wei, L.; Jian Cai, J.; Sun, Z. Therapeutic role of D-pinitol on experimental colitis via activating Nrf2/ARE and PPAR-γ/NF-κB signaling pathways. Food Funct. 2021, 12, 2554–2568. [Google Scholar] [CrossRef]
  85. Alonso-Castro, A.J.; Alba-Betancourt, C.; Rocha-González, E.; Ruiz-Arredondo, A.; Zapata-Morales, J.R.; Gasca-Martínez, D.; Pérez-Gutiérrez, S. Neuropharmacological effects of d-pinitol and its possible mechanisms of action. J. Food Biochem. 2019, 43, e13070. [Google Scholar] [CrossRef]
  86. Narayanan, C.R.; Joshi, D.D.; Muhumdar, A.M.; Dhekne, V.V. Pinitol—A new anti-diabetic compound from the leaves of Bougainvillea spectabilis. Curr. Sci. 1987, 56, 139–141. [Google Scholar]
  87. Sivakumar, S.; Subramanian, S.P. D-pinitol attenuates the impaired activities of hepatic key enzymes in carbohydrate metabolism of streptozotocin-induced diabetic rats. Gen. Physiol. Biophys. 2009, 28, 233–241. [Google Scholar] [CrossRef] [Green Version]
  88. Sivakumar, S.; Subramanian, S.P. Pancreatic tissue protective nature of D-Pinitol studied in streptozotocin-mediated oxidative stress in experimental diabetic rats. Eur. J. Pharmacol. 2009, 622, 65–70. [Google Scholar] [CrossRef]
  89. Dang, N.T.; Mukai, R.; Yoshida, K.; Ashida, H. D-pinitol and myo-inositol stimulate translocation of glucose transporter 4 in skeletal muscle of C57BL/6 mice. Biosci. Biotechnol. Biochem. 2010, 74, 1062–1067. [Google Scholar] [CrossRef] [Green Version]
  90. Lee, B.H.; Lee, C.C.; Wu, S.C. Ice plant (Mesembryanthemum crystallinum) improves hyperglycaemia and memory impairments in a Wistar rat model of streptozotocin-induced diabetes. J. Sci Food Agric. 2014, 94, 2266–2273. [Google Scholar] [CrossRef]
  91. Huang, B.; Wang, Z.; Park, J.H.; Ryu, O.H.; Choi, M.K.; Lee, J.Y.; Kang, Y.H.; Lim, S.S. Anti-diabetic effect of purple corn extract on C57BL/KsJ db/db mice. Nutr. Res. Pract. 2015, 9, 22–29. [Google Scholar] [CrossRef]
  92. Srivastava, K.; Dubey, A.; Tiwari, M.; Dubey, A. To evaluate the synergistic effect of Pinitol with Glimepride in diabetic Wistar rats. J. Crit. Rev. 2020, 7, 2058–2062. [Google Scholar]
  93. Kim, J.I.; Kim, J.C.; Kang, M.J.; Lee, M.S.; Kim, J.J.; Cha, I.J. Effects of pinitol isolated from soybeans on glycaemic control and cardiovascular risk factors in Korean patients with type II diabetes mellitus: A randomized controlled study. Eur. J. Clin. Nutr. 2005, 59, 456–458. [Google Scholar] [CrossRef]
  94. Kang, M.J.; Kim, J.I.; Yoon, S.Y.; Kim, J.C.; Cha, I.J. Pinitol from soybeans reduces postprandial blood glucose in patients with type 2 diabetes mellitus. J. Med. Food 2006, 9, 182–186. [Google Scholar] [CrossRef]
  95. Kim, M.J.; Yoo, K.H.; Kim, J.H.; Seo, Y.T.; Ha, B.W.; Kho, J.H.; Shin, Y.G.; Chung, C.H. Effect of pinitol on glucose metabolism and adipocytokines in uncontrolled type 2 diabetes. Diabetes Res. Clin. Pract. 2007, 77, S247–S251. [Google Scholar] [CrossRef]
  96. Hernández-Mijares, A.; Bañuls, C.; Peris, J.E.; Monzó, N.; Jover, A.; Bellod, L.; Victor, V.M.; Rocha, M. A single acute dose of pinitol from a naturally-occurring food ingredient decreases hyperglycaemia and circulating insulin levels in healthy subjects. Food Chem. 2013, 141, 1267–1272. [Google Scholar] [CrossRef]
  97. Lambert, C.; Cubedo, J.; Padró, T.; Vilahur, G.; López-Bernal, S.; Rocha, M.; Hernández-Mijares, A.; Badimon, L. Effects of a Carob-Pod-Derived Sweetener on Glucose Metabolism. Nutrients 2018, 10, 271. [Google Scholar] [CrossRef] [Green Version]
  98. Suzuki, Y.; Sakuraba, K.; Wada, T.; Watabane, N.; Wada, S.; Kitabayashi, Y.; Sunohara, M. Single-Dose Pinitol Ingestion Suppresses Post-Prandial Glucose Levels: A Randomized, Double-Blind, Placebo-Controlled, Crossover Trial. Nat. Prod. Commun. 2019, 14, 1–5. [Google Scholar] [CrossRef] [Green Version]
  99. Mishra, A.K.; Tewari, S.P. Theoretical evaluation of the bioactivity of plant-derived natural molecule D-Pinitol and other derived structure. AIP Conf. Proc. 2019, 2142, 150019. [Google Scholar] [CrossRef]
  100. Alonso-Castro, A.J.; Zapata-Morales, J.R.; Arana-Argáez, V.; Torres-Romero, J.C.; Ramírez-Villanueva, E.; Pérez-Medina, S.E.; Ramírez-Morales, M.A.; Juárez-Méndez, M.A.; Infante-Barrios, Y.P.; Martínez-Gutiérrez, F.; et al. Pharmacological and toxicological study of a chemical-standardized ethanol extract of the branches and leaves from Eysenhardtia polystachya (Ortega) Sarg. (Fabaceae). J. Ethnopharmacol. 2018, 224, 314–322. [Google Scholar] [CrossRef]
  101. Koh, E.S.; Kim, S.; Kim, M.; Hong, Y.A.; Shin, S.J.; Park, C.W.; Chang, Y.S.; Chung, S.; Kim, H.S. D-Pinitol alleviates cyclosporine A-induced renal tubulointerstitial fibrosis via activating Sirt1 and Nrf2 antioxidant pathways. Int. J. Mol. Med. 2018, 41, 1826–1834. [Google Scholar] [CrossRef]
  102. Geethan, P.K.; Prince, P.S. Antihyperlipidemic effect of D-pinitol on streptozotocin-induced diabetic Wistar rats. J. Biochem. Mol. Toxicol. 2008, 22, 220–224. [Google Scholar] [CrossRef]
  103. Singh, R.K.; Pandey, B.L.; Tripathi, M.; Pandey, V.B. Anti-inflammatory effect of (+)-pinitol. Fitoterapia 2001, 72, 168–170. [Google Scholar] [CrossRef]
  104. Kim, J.C.; Shin, J.Y.; Shin, D.H.; Kim, S.H.; Park, S.H.; Park, R.D.; Park, S.C.; Kim, Y.B.; Shin, Y.C. Synergistic antiinflammatory effects of pinitol and glucosamine in rats. Phytother. Res. 2005, 19, 1048–1051. [Google Scholar] [CrossRef] [PubMed]
  105. Sivakumar, S.; Palsamy, P.; Subramanian, S.P. Impact of D-pinitol on the attenuation of proinflammatory cytokines, hyperglycemia-mediated oxidative stress and protection of kidney tissue ultrastructure in streptozotocin-induced diabetic rats. Chem. Biol. Interact. 2010, 188, 237–245. [Google Scholar] [CrossRef]
  106. Zheng, K.; Zhao, Z.; Lin, N.; Wu, Y.; Xu, Y.; Zhang, W. Protective Effect of Pinitol Against Inflammatory Mediators of Rheumatoid Arthritis via Inhibition of Protein Tyrosine Phosphatase Non-Receptor Type 22 (PTPN22). Med. Sci. Monit. 2017, 23, 1923–1932. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Choi, M.S.; Lee, W.H.; Kwon, E.Y.; Kang, M.A.; Lee, M.K.; Park, Y.B.; Jeon, S.M. Effects of soy pinitol on the pro-inflammatory cytokines and scavenger receptors in oxidized low-density lipoprotein-treated THP-1 macrophages. J. Med. Food. 2007, 10, 594–601. [Google Scholar] [CrossRef] [PubMed]
  108. Eser, F.; Altundag, E.M.; Gedik, G.; Demirtas, I.; Onal, A.; Selvi, B. Anti-inflammatory effect of D-pinitol isolated from the leaves of Colutea cilicica Boiss et Bal. on K562 cells. Turk. J. Biochem. 2017, 42, 445–450. [Google Scholar] [CrossRef]
  109. Kong, J.; Du, Z.; Dong, L. Pinitol Prevents Lipopolysaccharide (LPS)-Induced Inflammatory Responses in BV2 Microglia Mediated by TREM2. Neurotox. Res. 2020, 38, 96–104. [Google Scholar] [CrossRef]
  110. López-Domènech, S.; Bañuls, C.; de Marañón, A.M.; Abab-Jiménez, Z.; Morillas, C.; Gómez-Abril, S.Á.; Rovira-Llopis, S.; Víctor, V.M.; Hernández-Mijares, A.; Rocha, M. Pinitol alleviates systemic inflammatory cytokines in human obesity by a mechanism involving unfolded protein response and sirtuin 1. Clin. Nutr. 2018, 37, 2036–2044. [Google Scholar] [CrossRef]
  111. Navarro, J.A.; Decara, J.; Medina-Vera, D.; Tovar, R.; Suarez, J.; Pavón, J.; Serrano, A.; Vida, M.; Gutierrez-Adan, A.; Sanjuan, C.; et al. D-Pinitol from Ceratonia siliqua Is an Orally Active Natural Inositol That Reduces Pancreas Insulin Secretion and Increases Circulating Ghrelin Levels in Wistar Rats. Nutrients 2020, 12, 2030. [Google Scholar] [CrossRef]
  112. Yu, J.; Choi, S.; Park, E.S.; Shin, B.; Yu, J.; Lee, S.H.; Takami, M.; Kang, J.S.; Meong, H.; Rho, J. D-chiro-inositol negatively regulates the formation of multinucleated osteoclasts by down-regulating NFATc1. J. Clin. Immunol. 2012, 32, 1360–1371. [Google Scholar] [CrossRef]
  113. Rengarajan, T.; Rajendran, P.; Nandakumar, N.; Balasubramanian, M.P.; Nishigaki, I. Free radical scavenging and antioxidant activity of D-pinitol against 7, 12 dimethylbenz(a) anthracene induced breast cancer in sprague dawley rats. Asian Pac. J. Trop. Dis. 2014, 4, 384–390. [Google Scholar] [CrossRef]
  114. Ma, J.; Feng, S.; Ai, D.; Liu, Y.; Yang, X. D-Pinitol Ameliorates Imiquimod-Induced PsoriasisLike Skin Inflammation in a Mouse Model via the NF-κB Pathway. J. Environ. Pathol. Toxicol. Oncol. 2019, 38, 285–295. [Google Scholar] [CrossRef]
  115. Suresh, K.G.; Manivannan, R.; Nivetha, B. In-silico docking analysis of phytochemicals from mimosa pudica l. Leaves as an antiviral agent against herpes simplex virus type I. Int. J. Biomed. NanoLet. 2021, 1, 1–9. [Google Scholar]
  116. Lee, J.S.; Lee, C.M.; Jeong, Y.I.; Jung, I.D.; Kim, B.H.; Seong, E.Y.; Kim, J.I.; Choi, I.W.; Chung, H.Y.; Park, Y.M. D-pinitol regulates Th1/Th2 balance via suppressing Th2 immune response in ovalbumin-induced asthma. FEBS Lett. 2007, 581, 57–64. [Google Scholar] [CrossRef] [Green Version]
  117. Liu, S.C.; Chuang, S.M.; Tang, C.H. D-pinitol inhibits RANKL-induced osteoclastogenesis. Int. Immunopharmacol. 2012, 12, 494–500. [Google Scholar] [CrossRef]
  118. Sudha, M.; Vetrichelvan, T. Protective effect of D-Pinitol isolated from aerial parts of soybean plants on haematological profile against Doxorubicin-induced cyto-toxicity in mice. Int. J. Pharm. Sci. Res. 2021, 12, 2926–2932. [Google Scholar] [CrossRef]
  119. Li, X.L.; Xu, M.; Yu, F.; Fu, C.L.; Yu, X.; Cheng, M.; Gao, H.Q. Effects of D-pinitol on myocardial apoptosis and fibrosis in streptozocin-induced aging-accelerated mice. J. Food Biochem. 2021, 45, e13669. [Google Scholar] [CrossRef]
  120. Hu, X.; Zhu, Y.; LV, X.; Feng, Z. Elucidation of the mechanism of action of pinitol against pressure overload-induced cardiac hypertrophy and fibrosis in an animal model of aortic stenosis. Biosci. Biotechnol. Biochem. 2021, 85, 643–655. [Google Scholar] [CrossRef]
  121. Cordero, C.P.; Pinzon, R.; Aristizabal, F.A. Cytotoxicity of bixin, rutin, pinitol B and ent-16-kauren-19-oic acid isolated from Colombian plants. Rev. Col. Cienc. Quím. Farm. 2003, 32, 137–140. [Google Scholar]
  122. Alonso-Castro, A.J.; Alba-Betancourt, C.; Yáñez-Barrientos, E.; Luna-Rocha, C.; Páramo-Castillo, A.S.; Aragón-Martínez, O.H.; Zapata-Morales, J.R.; Cruz-Jiménez, G.; Gasca-Martínez, D.; González-Ibarra, A.A.; et al. Diuretic activity and neuropharmacological effects of an ethanol extract from Senna septemtrionalis (Viv.) H.S. Irwin & Barneby (Fabaceae). J. Ethnopharmacol. 2019, 239, 111923. [Google Scholar] [CrossRef]
  123. Sudha, M.; Vetrichelvan, T. Genoprotective effect of D-Pinitol isolated from aerial parts of Soybean plants against Doxorubicin-induced genotoxicity evaluated by in vitro comet assay in Vero cell lines. Int. J. Res. Pharm. Sci. 2021, 12, 1379–1384. [Google Scholar] [CrossRef]
  124. Lee, E.; Lim, Y.; Kwon, S.W.; Kwon, O. Pinitol consumption improves liver health status by reducing oxidative stress and fatty acid accumulation in subjects with non-alcoholic fatty liver disease: A randomized, double-blind, placebo-controlled trial. J. Nutr. Biochem. 2019, 68, 33–41. [Google Scholar] [CrossRef]
  125. Ostlund, R.E.; Seemayer, R.; Gupta, S.; Kimmel, R.; Ostlund, E.L.; Sherman, W.R. A stereospecific myo-inositol/D-chiro-inositol transporter in HepG2 liver cells. Identification with D-chiro-3-3Hinositol. J. Biol. Chem. 1996, 271, 10073–10078. [Google Scholar] [CrossRef] [Green Version]
  126. Zhou, Y.; Park, C.M.; Cho, C.W.; Song, Y.S. Protective effect of pinitol against D-galactosamine-induced hepatotoxicity in rats fed on a high-fat diet. Biosci. Biotechnol. Biochem. 2008, 72, 1657–1666. [Google Scholar] [CrossRef] [Green Version]
  127. Sivakumar, S.; Palsamy, P.; Subramanian, S.P. Attenuation of oxidative stress and alteration of hepatic tissue ultrastructure by D-pinitol in streptozotocin-induced diabetic rats. Free Radic. Res. 2010, 44, 668–678. [Google Scholar] [CrossRef]
  128. Magielse, J.; Arcoraci, T.; Breynaert, A.; van Dooren, I.; Kanyanga, C.; Fransen, E.; Van Hoof, V.; Vlietinck, A.; Apers, S.; Pieters, L.; et al. Antihepatotoxic activity of a quantified Desmodium adscendens decoction and D-pinitol against chemically-induced liver damage in rats. J. Ethnopharmacol. 2013, 146, 250–256. [Google Scholar] [CrossRef]
  129. Rengarajan, T.; Rajendran, P.; Nandakumar, N.; Lokeshkumar, B.; Balasubramanian, M.P. D-Pinitol Protects Against Carbon Tetrachloride-Induced Hepatotoxicity in Rats. J. Environ. Pathol. Toxicol. Oncol. 2015, 34, 287–298. [Google Scholar] [CrossRef]
  130. Yan, L.; Luo, H.; Li, X.; Li, Y. d-Pinitol protects against endoplasmic reticulum stress and apoptosis in hepatic ischemia-reperfusion injury via modulation of AFT4-CHOP/GRP78 and caspase-3 signaling pathways. Int. J. Immunopathol. Pharmacol. 2021, 35, 20587384211032098. [Google Scholar] [CrossRef]
  131. Da Silva, J.A.; Da Silva, A.C.; Figueiredo, L.S.; Araujo, T.R.; Freitas, I.N.; Carneiro, E.M.; Ribeiro, E.S.; Ribeiro, R.A. D-Pinitol Increases Insulin Secretion and Regulates Hepatic Lipid Metabolism in Msg-Obese Mice. An. Acad. Bras. Cienc. 2020, 92, e20201382. [Google Scholar] [CrossRef]
  132. Muñoz, C.X.; Johnson, E.C.; Kunces, L.J.; McKenzie, A.L.; Wininger, M.; Butts, C.L.; Caldwell, A.; Seal, A.; McDermott, B.P.; Vingren, J.; et al. Impact of Nutrient Intake on Hydration Biomarkers Following Exercise and Rehydration Using a Clustering-Based Approach. Nutrients 2020, 12, 1276. [Google Scholar] [CrossRef]
  133. Adams, W.M.; Wininger, M.; Zaplatosch, M.E.; Hevel, D.J.; Maher, J.P.; McGuirt, J.T. Influence of Nutrient Intake on 24 Hour Urinary Hydration Biomarkers Using a Clustering-Based Approach. Nutrients 2020, 12, 2933. [Google Scholar] [CrossRef] [PubMed]
  134. Moreira, L.N.; Silva, J.F.; Silva, G.C.; Lemos, V.S.; Cortes, S.F. Activation of eNOS by D-pinitol Induces an Endothelium-Dependent Vasodilatation in Mouse Mesenteric Artery. Front. Pharmacol. 2018, 9, 528. [Google Scholar] [CrossRef] [PubMed]
  135. Lee, J.S.; Jung, I.D.; Jeong, Y.I.; Lee, C.M.; Shin, Y.K.; Lee, S.Y.; Suh, D.S.; Yoon, M.S.; Lee, K.S.; Choi, Y.H.; et al. D-pinitol inhibits Th1 polarization via the suppression of dendritic cells. Int. Immunopharmacol. 2007, 7, 791–804. [Google Scholar] [CrossRef] [PubMed]
  136. Bae, C.J.; Lee, J.W.; Shim, S.B.; Jee, S.W.; Lee, S.H.; Woo, J.M.; Lee, C.K.; Hwang, D.Y. GATA binding protein 3 overexpression and suppression significantly contribute to the regulation of allergic skin inflammation. Int. J. Mol. Med. 2011, 28, 171–179. [Google Scholar] [CrossRef] [PubMed]
  137. Chauhan, P.S.; Gupta, K.K.; Bani, S. The immunosuppressive effects of Agyrolobium roseum and pinitol in experimental animals. Int. Immunopharmacol. 2011, 11, 286–291. [Google Scholar] [CrossRef] [PubMed]
  138. Brautigan, D.L.; Brown, M.; Grindrod, S.; Chinigo, G.; Kruszewski, A.; Lukasik, S.M.; Bushweller, J.H.; Horal, M.; Keller, S.; Tamura, S.; et al. Allosteric activation of protein phosphatase 2C by D-chiro-inositol-galactosamine, a putative mediator mimetic of insulin action. Biochemistry 2005, 44, 11067–11173. [Google Scholar] [CrossRef] [PubMed]
  139. Kim, U.H.; Yoon, J.H.; Li, H.; Kang, J.H.; Ji, H.S.; Park, K.H.; Shin, D.H.; Park, H.Y.; Jeong, T.S. Pterocarpan-enriched soy leaf extract ameliorates insulin sensitivity and pancreatic β-cell proliferation in type 2 diabetic mice. Molecules 2014, 19, 18493–18510. [Google Scholar] [CrossRef]
  140. Gao, Y.; Zhang, M.; Wu, T.; Xu, M.; Cai, H.; Zhang, Z. Effects of D-Pinitol on Insulin Resistance through the PI3K/Akt Signaling Pathway in Type 2 Diabetes Mellitus Rats. J. Agric. Food Chem. 2015, 63, 6019–6026. [Google Scholar] [CrossRef]
  141. Medina-Vera, D.; Navarro, J.A.; Tovar, R.; Rosell-Valle, C.; Gutiérrez-Adan, A.; Ledesma, J.C.; Sanjuan, C.; Pavón, F.J.; Baixeras, E.; Rodríguez de Fonseca, F.; et al. Activation of PI3K/Akt Signaling Pathway in Rat Hypothalamus Induced by an Acute Oral Administration of D-Pinitol. Nutrients 2021, 13, 2268. [Google Scholar] [CrossRef]
  142. Kim, H.J.; Park, K.S.; Lee, S.K.; Min, K.W.; Han, K.A.; Kim, Y.K.; Ku, B.J. Effects of pinitol on glycemic control, insulin resistance and adipocytokine levels in patients with type 2 diabetes mellitus. Ann. Nutr. Metab. 2012, 60, 1–5. [Google Scholar] [CrossRef]
  143. Do, G.M.; Choi, M.S.; Kim, H.J.; Woo, M.N.; Lee, M.K.; Jeon, S.M. Soy pinitol acts partly as an insulin sensitizer or insulin mediator in 3T3-L1 preadipocytes. Genes Nutr. 2008, 2, 359–364. [Google Scholar] [CrossRef] [Green Version]
  144. Siracusa, L.; Occhiuto, C.; Molonia, M.S.; Cimino, F.; Palumbo, M.; Saija, A.; Speciale, A.; Rocco, C.; Ruberto, G.; Cristani, M. A pinitol-rich Glycyrrhiza glabra L. leaf extract as functional supplement with potential in the prevention of endothelial dysfunction through improving insulin signalling. Arch. Physiol. Biochem. 2020, 1–10. [Google Scholar] [CrossRef]
  145. Vasaikar, N.; Mahajan, U.; Patil, K.R.; Suchal, K.; Patil, C.R.; Ojha, S.; Goyal, S.N. D-pinitol attenuates cisplatin-induced nephrotoxicity in rats: Impact on pro-inflammatory cytokines. Chem. Biol. Interact. 2018, 290, 6–11. [Google Scholar] [CrossRef]
  146. Farias, V.X.; Macêdo, F.H.; Oquendo, M.B.; Tomé, A.R.; Báo, S.N.; Cintra, D.O.; Santos, C.F.; Albuquerque, A.A.; Heimark, D.B.; Larner, J.; et al. Chronic treatment with D-chiro-inositol prevents autonomic and somatic neuropathy in STZ-induced diabetic mice. Diabetes Obes. Metab. 2011, 13, 243–250. [Google Scholar] [CrossRef]
  147. Dong, W.; Zhao, S.; Wen, S.; Dong, C.; Chen, Q.; Gong, T.; Chen, W.; Liu, W.; Mu, L.; Shan, H.; et al. A preclinical randomized controlled study of ischemia treated with Ginkgo biloba extracts: Are complex components beneficial for treating acute stroke? Curr. Res. Transl. Med. 2020, 68, 197–203. [Google Scholar] [CrossRef]
  148. An, Y.; Li, J.; Liu, Y.; Fan, M.; Tian, W. Protective effect of D-pinitol on the experimental spinal cord injury in rats. Metab. Brain Dis. 2020, 35, 473–482. [Google Scholar] [CrossRef]
  149. Sakata, K.; Kawasaki, H.; Suzuki, T.; Ito, K.; Negishi, O.; Tsuno, T.; Tsuno, H.; Yamazaki, Y.; Ishida, N. Inositols affect the mating circadian rhythm of Drosophila melanogaster. Front. Pharmacol. 2005, 6, 111. [Google Scholar] [CrossRef] [Green Version]
  150. Rahaman, M.S.; Yamasaki, S.; Binte Hossain, K.F.; Hosokawa, T.; Saito, T.; Kurasaki, M. Effects of curcumin, D-pinitol alone or in combination in cytotoxicity induced by arsenic in PC12 Cells. Food Chem. Toxicol. 2020, 144, 111577. [Google Scholar] [CrossRef]
  151. Juneja, K.; Mishra, R.; Chauhan, S.; Gupta, S.; Roy, P.; Sircar, D. Metabolite profiling and wound-healing activity of Boerhavia diffusa leaf extracts using in vitro and in vivo models. J. Trad. Complement. Med. 2020, 10, 52–59. [Google Scholar] [CrossRef]
  152. López-Gambero, A.J.; Sanjuan, C.; Serrano-Castro, P.J.; Suárez, J.; Rodríguez de Fonseca, F. The Biomedical Uses of Inositols: A Nutraceutical Approach to Metabolic Dysfunction in Aging and Neurodegenerative Diseases. Biomedicines 2020, 8, 295. [Google Scholar] [CrossRef]
  153. Srivastava, K.; Tiwari, M.; Dubey, A.; Dubey, A. D-Pinitol—A Natural Phytomolecule and its Pharmacological effect. Int. J. Pharm. Life Sci. 2020, 11, 6609–6623. [Google Scholar]
  154. Antonowski, T.; Osowski, A.; Lahuta, L.; Górecki, R.; Rynkiewicz, A.; Wojtkiewicz, J. Health-Promoting Properties of Selected Cyclitols for Metabolic Syndrome and Diabetes. Nutrients 2019, 11, 2314. [Google Scholar] [CrossRef] [Green Version]
  155. Kalekar, S.A.; Munshi, R.P.; Bhalerao, S.S.; Thatte, U.M. Insulin sensitizing effect of 3 Indian medicinal plants: An in vitro study. Indian J. Pharmacol. 2013, 45, 30–33. [Google Scholar] [CrossRef]
  156. Stadlbauer, V.; Neuhauser, C.; Aumiller, T.; Stallinger, A.; Iken, M.; Weghuber, J. Identification of Insulin-Mimetic Plant Extracts: From an In Vitro High-Content Screen to Blood Glucose Reduction in Live Animals. Molecules 2021, 26, 4346. [Google Scholar] [CrossRef]
  157. Papaefstathiou, E.; Agapiou, A.; Giannopoulos, S.; Kokkinofta, R. Nutritional characterization of carobs and traditional carob products. Food Sci. Nutr. 2018, 6, 2151–2161. [Google Scholar] [CrossRef]
  158. Kim, I.-S.; Kim, C.-H.; Yang, W.-S. Physiologically Active Molecules and Functional Properties of Soybeans in Human Health—A Current Perspective. Int. J. Mol. Sci. 2021, 22, 4054. [Google Scholar] [CrossRef]
  159. Tewari, R.; Gupta, M.; Ahmad, F.; Rout, P.K.; Misra, L.; Patwardhan, A.; Vasudeva, R. Extraction, quantification and antioxidant activities of flavonoids, polyphenols and pinitol from wild and cultivated Saraca asoca bark using RP-HPLC-PDA-RI method. Ind. Crops Prod. 2017, 103, 73–80. [Google Scholar] [CrossRef]
Figure 1. Major and new phenolic compounds found in Carob pods and leaves [12,13,14].
Figure 1. Major and new phenolic compounds found in Carob pods and leaves [12,13,14].
Nutrients 14 01453 g001
Figure 2. Insulin mechanism of action in healthy conditions.
Figure 2. Insulin mechanism of action in healthy conditions.
Nutrients 14 01453 g002
Figure 3. Synthetic sterol with insulin-sensitivity improvement activity [29].
Figure 3. Synthetic sterol with insulin-sensitivity improvement activity [29].
Nutrients 14 01453 g003
Figure 4. Carpachromene and Metformin.
Figure 4. Carpachromene and Metformin.
Nutrients 14 01453 g004
Figure 5. Stevioside (Stevia rebaudiana Bertoni).
Figure 5. Stevioside (Stevia rebaudiana Bertoni).
Nutrients 14 01453 g005
Figure 6. Structures of naturally occurring inositols.
Figure 6. Structures of naturally occurring inositols.
Nutrients 14 01453 g006
Figure 7. Inositols and their methyl ethers isolated from Carob.
Figure 7. Inositols and their methyl ethers isolated from Carob.
Nutrients 14 01453 g007
Figure 8. Insulin-sensitizing mechanism of D-Pinitol.
Figure 8. Insulin-sensitizing mechanism of D-Pinitol.
Nutrients 14 01453 g008
Figure 9. Insulin-mimetic mechanism of D-Pinitol.
Figure 9. Insulin-mimetic mechanism of D-Pinitol.
Nutrients 14 01453 g009
Table 1. General composition of Carob fruits [2].
Table 1. General composition of Carob fruits [2].
ComponentProportion (%)
Moisture6.3–7.6
Protein1.7–5.9
Ash2.3–3.2
Fat0.2–4.4
Total dietary fiber11.7–47
Starch0.1
Total carbohydrates42–86
Fructose2–7.4
Glucose3–7.3
Sucrose15–34
D-Pinitol5.5
Table 2. Functions of insulin in human body [21].
Table 2. Functions of insulin in human body [21].
Effect TypeRole of Insulin
MetabolicStimulation of glucose transport and metabolism
Stimulation of glycogen synthesis
Stimulation of lipogenesis
Inhibition of lipolysis
Stimulation of ion flux
Growth-promotingStimulation of DNA synthesis
Stimulation of cell growth and differentiation
Metabolic & Growth-promotingStimulation of amino acid influx
Stimulation of protein synthesis
Inhibition of protein degradation
Stimulation of RNA synthesis
Table 3. Molecular mechanisms of insulin resistance [24].
Table 3. Molecular mechanisms of insulin resistance [24].
Molecular MechanismRoles in Insulin Resistance
Upregulation of PTP1B [25]Reverses insulin-induced phosphorylation in tyrosine residues of IRS-1 and so impairs insulin signal transduction
Inflammatory mediators and adipokinesActivation of IKKβ/NF-κB and JNK pathways, serine phosphorylation of IRS-1 in the site of 307, declines GLUT-4 expression, reduces IRS-1 expression via ERK1/2, induce IRS degradation through SOCS1- and SOCS3-dependent mechanisms
Free radical overloadActivates several serine–threonine kinase pathways, i.e., IKKβ/NF-κB and JNK, IRS degradation, suppresses GLUT-4 expression and localization in cell membrane,
decreases insulin-induced IRS-1 and PIP-kinase relocation between cytoplasm and microsomes, decreases PKB phosphorylation, serine phosphorylation at site of serine 307 of IRS-1, activates inflammatory responses
Defects in serine phosphorylation of IRS-1Decrease in insulin receptor phosphorylation, phosphorylation in serine 307 which blocks signaling
Obesity and adipocytes importanceDecrease in insulin receptor phosphorylation, phosphorylation in serine 307 which blocks signaling
Accelerated insulin degradationAutoimmune antibodies against insulin or abnormal
insulin structure due to mutation
Mitochondrial dysfunctionInduces oxidative stress, impairs insulin signaling
Reduced the capacity of
receptors to binding to insulin
Decrease in number of insulin receptors, reduction in
functional receptors due to mutation, autoimmune
antibodies against insulin receptors
Mutations of GLUT-4Point mutation changes normal modification of GLUT-4, inhibits glucose entering into dependent cells and
impairs subsequent signaling pathways
ER stressDisrupts proper protein folding leading to accumulation of misfolded proteins
PTP1B [25], protein tyrosine phosphatase 1B; IRS-1, insulin receptor substrates-1; IKKβ/NF-κB, central regulator of NF-κB; GLUT-4, type 4 glucose transporter; ERK, extracellular signal-regulated kinase SOCS1/3, suppressor of cytokine signaling; JNK, c-Jun N-terminal kinase; ER, endoplasmic reticulum.
Table 4. Selected publications of insulin regulation and women fertility disorders treatment of inositols.
Table 4. Selected publications of insulin regulation and women fertility disorders treatment of inositols.
Property Short DescriptionType of PublicationRef., Year
Insulin regulation in human diabeticsresearch[39], 1990
Treatment respiratory disorders in infantsresearch[40], 1992
Insulin regulation in human diabeticsresearch[41], 1993
Treatments of psychiatric disordersreview[42], 1997
Treatment of polycystic ovary syndrome (PCOS)research[43], 1999
Treatment of Alzheimer disease, in vitroresearch[44], 2000
Insulin regulation in human diabeticsresearch[45], 2005
Treatment of endothelial dysfunction, antioxidant, animal modelresearch[46], 2006
Biological rolesreview[47], 2007
Derivatives and their functionsreview[48], 2008
Treatment of PCOSreview[49], 2014
Insulin regulation in obese male childrenresearch[50], 2016
Treatment of PCOSreview[51], 2016
Treatment of PCOSresearch[52], 2017
Bioavailability for treatment of PCOSreview[53], 2017
Treatment of PCOS in subfertile womenreview[54], 2018
Effects on glucose homeostasisreview[55], 2019
General presentation of medicinal activitiesreview[56], 2019
Treatment of PCOSreview[57], 2020
Treatment of PCOS, with other technologiesreview[58], 2021
Treatment of preterm birthreview[59], 2021
Treatment of psychological symptoms in PCOSreview[60], 2021
Insulin regulation in pregnancyreview[38], 2022
Table 5. Published properties of D-Pinitol.
Table 5. Published properties of D-Pinitol.
Activity/PropertyTesting MethodRef.
Anti-AlzheimerIn vivo, mice[67]
Anti-AlzheimerIn vitro, hippocampal cultures[68]
Anti-AlzheimerIn vivo, C. elegans, mice[69]
AntiagingIn vivo, D. Melanogaster[70]
AntibacterialM. smegmatis[71]
AnticancerIn vitro, human cancer cells[72,73,74,75,76,77]
AnticancerIn vivo, rats[78,79,80,81,82,83]
Anti-colitisIn vivo, rats[84]
AntidepressantIn vivo, mice[85]
AntidiabeticIn vivo, mice/rats[86,87,88,89,90,91,92]
AntidiabeticIn vivo, humans[93,94,95,96,97,98]
AntidiabeticTheoretical evaluation[99]
AntidiarrhealIn vivo, mice[100]
AntifibroticIn vivo, mice[101]
AntihyperlipidemicIn vivo, rats[64,102]
Anti-inflammatoryIn vivo, mice/rats[103,104,105,106]
Anti-inflammatoryIn vitro, Human cells[72,107,108]
Anti-inflammatoryIn vitro, BV2 microglial cells[109]
AntinociceptiveIn vivo, mice[100]
Anti-obesityIn vivo, humans[110]
Anti-obesityIn vivo, rats[111]
Anti-osteoclasticIn vitro, UAMS32 cells[112]
AntioxidantIn vivo, rats[78,81,82,88,113]
Anti-psoriaticIn vivo, mice[114]
AntiviralTheoretical evaluation[115]
Asthma treatmentIn vivo, mice[116]
Bone protectionIn vitro, Bone marrow cell lines, rats[117]
Bone protectionIn vivo, rats[118]
CardioprotectiveIn vivo, humans[93]
CardioprotectiveIn vivo, mice/rats[119,120]
CytotoxicIn vitro, human cancer cell lines[121]
DiureticIn vivo, mice[122]
Geno-protectiveIn vitro, monkey liver cell lines[123]
HepatoprotectiveIn vivo, humans[124]
HepatoprotectiveIn vivo, mice/rats[125,126,127,128,129,130,131]
Hydration biomarkerIn vivo, humans[132,133]
HypotensiveIn vivo, mice[134]
Immuno-protectiveTheoretical evaluation[99]
Immuno-protectiveIn vivo, mice[116,135,136]
ImmunosuppressiveIn vivo, mice[137]
Insulin regulationIn vivo, mice/rats[111,131,138,139,140,141]
Insulin regulationIn vivo, humans[96,142]
Insulin regulationIn vitro, 3T3-L1, HUVEC cells[143,144]
Memory enhancementIn vivo, rats[90]
Nanoparticles loadedIn vitro, against M. smegmatis[29]
NephroprotectiveIn vivo, mice/rats[105,145]
NeuroprotectiveIn vivo, mice/rats[85,122,146,147,148]
Sleep enhancerIn vivo, D. melanogaster, in vitro PC12 cells[149]
Synergism w/ curcuminIn vitro, PC12 cells, against As+3 toxicity[150]
Wound healingIn vivo, rats, in vitro, HaCaT cells[151]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Azab, A. D-Pinitol—Active Natural Product from Carob with Notable Insulin Regulation. Nutrients 2022, 14, 1453. https://doi.org/10.3390/nu14071453

AMA Style

Azab A. D-Pinitol—Active Natural Product from Carob with Notable Insulin Regulation. Nutrients. 2022; 14(7):1453. https://doi.org/10.3390/nu14071453

Chicago/Turabian Style

Azab, Abdullatif. 2022. "D-Pinitol—Active Natural Product from Carob with Notable Insulin Regulation" Nutrients 14, no. 7: 1453. https://doi.org/10.3390/nu14071453

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