Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
EXTRACTS FROM PLANTS OF THE MORINGACEAE FAMILY AND METHODS OF MAKING
Document Type and Number:
WIPO Patent Application WO/2016/164224
Kind Code:
A1
Abstract:
The present application is directed to materials and methods for producing from a plant of the Moringaceae family having high concentrations of moringa isothiocyanates.

Inventors:
RASKIN ILYA (US)
WATERMAN CARRIE (US)
Application Number:
PCT/US2016/024924
Publication Date:
October 13, 2016
Filing Date:
March 30, 2016
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV RUTGERS (US)
International Classes:
A61K8/97; A61K36/185; A61Q19/00
Domestic Patent References:
WO2014053944A12014-04-10
Foreign References:
US20110245526A12011-10-06
US20070264366A12007-11-15
US20060127996A12006-06-15
US20030022838A12003-01-30
US5744161A1998-04-28
Other References:
WATERMAN ET AL.: "Stable, water extractable isothiocyanates from Moringa oleifera leaves attenuate inflammation in vitro", PHYTOCHEMISTRY, vol. 103, July 2014 (2014-07-01), pages 114 - 122, XP055321973
BAIS ET AL.: "Antiobesity and Hypolipidemic activity of Moringa oleifera leaves against high fat diet-induced obesity in rats", ADVANCES IN BIOLOGY., vol. 2014, 10 July 2014 (2014-07-10), pages 1 - 9, XP055321974
KINZIG ET AL., J. NEUROSCI., vol. 23, 2003, pages 6163 - 6170
MBIKAY M: "Therapeutic potential of Moringa oleifera leaves in chronic hyperglycemia and dyslipidemia: a review", FRONT PHARMACOL, vol. 3, 2012, pages 1 - 12
WATERMAN CCHENG DMROJAS-SILVA PPOULEV ADREIFUS JLILA MA ET AL.: "Stable, water extractable isothiocyanates from Moringa oleifera leaves attenuate inflammation in vitro", PHYTOCHEM, vol. 103, 2014, pages 114 - 22, XP055321973, DOI: 10.1016/j.phytochem.2014.03.028
CHEENPRACHA SPARK E-JYOSHIDA WYBARIT CWALL MPEZZUTO JM ET AL.: "Potential anti-inflammatory phenolic glycosides from the medicinal plant Moringa oleifera fruits", BIOORGAN MED CHEM, vol. 18, no. 17, 2010, pages 6598 - 602, XP027218846
BAE JYLIM SSKIM SJCHOI JSPARK JJU SM ET AL.: "Bog blueberry anthocyanins alleviate photoaging in ultraviolet-B irradiation-induced human dermal fibroblasts", MOL NUTR FOOD RES, vol. 53, no. 6, 2009, pages 726 - 38, XP055612763, DOI: 10.1002/mnfr.200800245
BRUNELLI DTAVECCHIO MFALCIONI CFRAPOLLI RERBA EIORI R ET AL.: "The isothiocyanate produced from glucomoringin inhibits NF-kB and reduces myeloma growth in nude mice in vivo", BIOCHEM PHARMACOL, vol. 79, no. 8, 2010, pages 1141 - 8
FAIZI SSIDDIQUI BSSALEEM RSIDDIQUI SAFTAB KGILANI AH: "Isolation and structure elucidation of new nitrile and mustard oil glycosides from Moringa oleifera and their effect on blood pressure", J NAT PROD, vol. 57, no. 9, 1994, pages 1256 - 61, XP009024216, DOI: 10.1021/np50111a011
SHAPIRO TAFAHEY JWWADE KLSTEPHENSON KKTALALAY P: "Chemoprotective glucosinolates and isothiocyanates of broccoli sprouts metabolism and excretion in humans", CANCER EPIDEM BIOMAR, vol. 10, no. 5, 2001, pages 501 - 8
HIGDON JVDELAGE BWILLIAMS DEDASHWOOD RH: "Cruciferous vegetables and human cancer risk: epidemiologic evidence and mechanistic basis", PHARMACOL RES, vol. 55, no. 3, 2007, pages 224 - 36, XP022014513, DOI: 10.1016/j.phrs.2007.01.009
VERHOEVEN DTGOLDBOHM RAVAN POPPEL GVERHAGEN HVAN DEN BRANDT PA: "Epidemiological studies on brassica vegetables and cancer risk", CANCER EPIDEM BIOMAR, vol. 5, no. 9, 1996, pages 733 - 48, XP002669900
TRAKA MMITHEN R: "Glucosinolates, isothiocyanates and human health", PHYTOCHEM REV, vol. 8, no. 1, 2009, pages 269 - 82, XP019686429
MIRMIRAN PBAHADORAN ZHOSSEINPANAH FKEYZAD AAZIZI F: "Effects of broccoli sprout with high sulforaphane concentration on inflammatory markers in type 2 diabetic patients: A randomized double-blind placebo-controlled clinical trial", J FUNCT FOODS, vol. 4, no. 4, 2012, pages 837 - 41, XP055100429, DOI: 10.1016/j.jff.2012.05.012
BAHADORAN ZTOHIDI MNAZERI PMEHRAN MAZIZI FMIRMIRAN P: "Effect of broccoli sprouts on insulin resistance in type 2 diabetic patients: a randomized double-blind clinical trial", INT J FOOD SCI NUTR, vol. 63, no. 7, 2012, pages 767 - 71, XP009176001, DOI: 10.3109/09637486.2012.665043
BAHADORAN ZMIRMIRAN PAZIZI F: "Potential Efficacy of Broccoli Sprouts as a Unique Supplement for Management of Type 2 Diabetes and Its Complications", J MED FOOD, 2013
WU HLIANG HYUAN QWANG TYAN X: "Preparation and stability investigation of the inclusion complex of sulforaphane with hydroxypropyl-P-cyclodextrin", CARBOHYD POLYM, vol. 82, no. 3, 2010, pages 613 - 7, XP027206323
PARK E-JCHEENPRACHA SCHANG LCKONDRATYUK TPPEZZUTO JM: "Inhibition of lipopolysaccharide-induced cyclooxygenase-2 and inducible nitric oxide synthase expression by 4-[(2'-O-acetyl-a-L-rhamnosyloxy)benzyl]isothiocyanate from Moringa oleifera", NUTR CANCER, vol. 63, no. 6, 2011, pages 971 - 82
SHETTY P: "Public health: India's diabetes time bomb", NATURE, vol. 485, no. 7398, 2012, pages S14 - S6
MBANYA JCNMOTALA AASOBNGWI EASSAH FKENORU ST: "Diabetes in sub-Saharan Africa", LANCET, vol. 375, no. 9733, 2010, pages 2254 - 66, XP027291392, DOI: 10.1016/S0140-6736(10)60550-8
NG MFLEMING TROBINSON MTHOMSON BGRAETZ NMARGONO C ET AL.: "Global, regional, and national prevalence of overweight and obesity in children and adults during 1980-2013: a systematic analysis for the Global Burden of Disease Study 2013", LANCET, 2014
FOLCH JLEES MSLOANE-STANLEY G: "A simple method for the isolation and purification of total lipids from animal tissues", J BIOL CHEM, vol. 226, no. 1, 1957, pages 497 - 509
SCHMITTGEN TDLIVAK KJ: "Analyzing real-time PCR data by the comparative CT method", NATURE PROTOCOLS, vol. 3, no. 6, 2008, pages 1101 - 8, XP055137608, DOI: 10.1038/nprot.2008.73
CHENG DMKUHN PPOULEV AROJO LELILA MARASKIN I: "In vivo and in vitro antidiabetic effects of aqueous cinnamon extract and cinnamon polyphenol-enhanced food matrix", FOOD CHEM, vol. 135, no. 4, 2012, pages 2994 - 3002
MOSMANN T: "Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays", J IMMUNOL METHODS, vol. 65, no. 1, 1983, pages 55 - 63, XP002455867, DOI: 10.1016/0022-1759(83)90303-4
JAISWAL DKUMAR RAI PKUMAR AMEHTA SWATAL G: "Effect of Moringa oleifera Lam. leaves aqueous extract therapy on hyperglycemic rats", J ETHNOPHARMACOL, vol. 123, no. 3, 2009, pages 392 - 6, XP026150433, DOI: 10.1016/j.jep.2009.03.036
NDONG MOUSSA UMKATSUMATA SHIN-ICHI: "Suzuki Kazuharu Effects of Oral Administration of Moringa oleifera Lam on Glucose Tolerance in Goto-Kakizaki and Wistar Rats", J CLIN BIOCHEM NUTR, vol. 40, no. 3, 2007, pages 229 - 33
MILLER RSBECKER KGPRABHU VCOOKE DW: "Adipocyte gene expression is altered in formerly obese mice and as a function of diet composition", J NUTR, vol. 138, no. 6, 2008, pages 1033 - 8
KORDA MKUBANT RPATTON SMALINSKI T: "Leptin-induced endothelial dysfunction in obesity", AM J PHYSIOL-HEART C, vol. 295, no. 4, 2008, pages H1514 - H21
MOLLER DE: "Potential role of TNF-alpha in the pathogenesis of insulin resistance and type 2 diabetes", TRENDS ENDOCRIN MET, vol. 11, no. 6, 2000, pages 212 - 7
UYSAL KTWIESBROCK, S.M.MARINO, M.W.HOTAMISLIGIL, G.S.: "Protection from obesity-induced insulin resistance in mice lacking TNFalpha function", NATURE, vol. 389, 1997, pages 610 - 4
SCHREYER SACHUA JR SCLEBOEUF RC: "Obesity and diabetes in TNF-alpha receptor-deficient mice", J CLINL INVEST, vol. 102, no. 2, 1998, pages 402
HOTAMISLIGIL GSMURRAY DLCHOY LNSPIEGELMAN BM: "Tumor necrosis factor alpha inhibits signaling from the insulin receptor", PNAS, vol. 91, no. 11, 1994, pages 4854 - 8
HUNDAL RSKRSSAK MDUFOUR SLAURENT DLEBON VCHANDRAMOULI V ET AL.: "Mechanism by which metformin reduces glucose production in type 2 diabetes", DIABETES, vol. 49, no. 12, 2000, pages 2063 - 9
KNOWLER WCBARRETT-CONNOR EFOWLER SEHAMMAN RFLACHIN JMWALKER EA ET AL.: "Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin", NEW ENGL J MED, vol. 346, no. 6, 2002, pages 393 - 403, XP055111414, DOI: 10.1056/NEJMoa012512
RENA GPEARSON ERSAKAMOTO K: "Molecular mechanism of action of metformin: old or new insights?", DIABETOLOGIA, vol. 56, no. 9, 2013, pages 1898 - 906
GEERLING JJBOON MRVAN DER ZON GCVAN DEN BERG SAVAN DEN HOEK AMLOMBES M ET AL.: "Metformin lowers plasma triglycerides by promoting VLDL-triglyceride clearance by brown adipose tissue in mice", DIABETES, vol. 63, no. 3, 2013, pages 880 - 91
MADIRAJU AKERION DMRAHIMI YZHANG X-MBRADDOCK DTALBRIGHT RA ET AL.: "Metformin suppresses gluconeogenesis by inhibiting mitochondrial glycerophosphate dehydrogenase", NATURE, vol. 510, 2014, pages 542 - 6
WIDJAJA ASTRATTON IMHORN RHOLMAN RRTURNER RBRABANT G: "UKPDS 20: plasma leptin, obesity, and plasma insulin in type 2 diabetic subjects", J CLIN ENDOCRINOL METAB, vol. 82, no. 2, 1997, pages 654 - 7
STEPPAN CMBAILEY STBHAT SBROWN EJBANERJEE RRWRIGHT CM ET AL.: "The hormone resistin links obesity to diabetes", NATURE, vol. 409, no. 6818, 2001, pages 307 - 12, XP002970710, DOI: 10.1038/35053000
SRINIVASAN KVISWANAD BASRAT LKAUL CRAMARAO P: "Combination of high-fat diet-fed and low-dose streptozotocin-treated rat: a model for type 2 diabetes and pharmacological screening", PHARMACOL RES, vol. 52, no. 4, 2005, pages 313 - 20, XP005031012, DOI: 10.1016/j.phrs.2005.05.004
EL MESSAOUDI SRONGEN GADE BOER RARIKSEN NP: "The cardioprotective effects of metformin", CURRENT OPIN LIPIDOL, vol. 22, no. 6, 2011, pages 445 - 53
ALBERTI ET AL., CIRCULATION, vol. 120, no. 16, 2009, pages 1640 - 1645
AMAGLO ET AL., FOOD CHEMISTRY, vol. 122, no. 4, 2010, pages 1047 - 1054
BAO ET AL., BIOCHIMICA ET BIOPHYSICA ACTA - REVIEWS ON CANCER, vol. 1815, no. 2, 2011, pages 135 - 146
SIDDHURAJU, P.BECKER, K., JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY, vol. 51, no. 11, 2003, pages 2144 - 2155
BHARGAVA, P.LEE, C., BIOCHEMICAL JOURNAL, vol. 442, 2012, pages 253 - 262
BRUNELLI ET AL., BIOCHEMICAL PHARMACOLOGY, vol. 79, no. 8, 2010, pages 1141 - 1148
CHEENPRACHA ET AL., BIOORGANIC & MEDICINAL CHEMISTRY, vol. 18, no. 17, 2010, pages 6598 - 6602
DILLARD, C. J.GERMAN, J. B., JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE, vol. 80, no. 12, 2000, pages 1744 - 1756
EYLEN ET AL., JOURNAL OF FOOD ENGINEERING, vol. 89, no. 2, 2008, pages 178 - 186
FAHEY, J. W., TREES FOR LIFE JOURNAL, vol. 1, 2005, pages 5
FERRANTE, A. W., JOURNAL OF INTERNAL MEDICINE, vol. 262, no. 4, 2007, pages 408 - 414
FORCE ET AL., POSTHARVEST BIOLOGY AND TECHNOLOGY, vol. 44, no. 2, 2007, pages 175 - 178
FRANKLIN ET AL., DRUG DEVELOPMENT AND INDUSTRIAL PHARMACY, vol. 0, 2013, pages 1 - 9
GALLAHER ET AL., JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY, vol. 60, no. 6, 2012, pages 1358 - 1362
GERONIKAKI, A. A.GAVALAS, A. M., COMBINATORIAL CHEMISTRY & HIGH THROUGHPUT SCREENING, vol. 9, no. 6, 2006, pages 425 - 442
GIUSTI, M. M.WROLSTAD, R. E., BIOCHEMICAL ENGINEERING JOURNAL, vol. 14, no. 3, 2003, pages 217 - 225
HOBBS ET AL., ANNUAL REVIEW OF PHARMACOLOGY AND TOXICOLOGY, vol. 39, no. 1, 1999, pages 191 - 220
HOTAMISLIGIL ET AL., PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, vol. 91, no. 11, 1994, pages 4854 - 4858
MARIAPPAN ET AL., CARDIOVASCULAR RESEARCH, vol. 85, no. 3, 2010, pages 473 - 483
MBIKAY, M., FRONTIERS IN PHARMACOLOGY, vol. 3, 2012, pages 1 - 12
MIRZA ET AL., CYTOKINE, vol. 57, no. 1, 2012, pages 136 - 142
MOCELLIN, S.NITTI, D., FRONTIERS IN BIOSCIENCE, vol. 13, 2008, pages 2774 - 2783
MOSMANN, T., JOURNAL OF IMMUNOLOGICAL METHODS, vol. 65, no. 1, 1983, pages 55 - 63
MOYO ET AL., MEAT SCIENCE, vol. 91, no. 4, 2012, pages 441 - 447
PANDEY ET AL., MEDICINAL & AROMATIC PLANTS: OPEN ACCESS, vol. 1, 2012, pages 1 - 8
PARK ET AL., NUTRITION AND CANCER, vol. 63, no. 6, 2011, pages 971 - 982
PEREIRA ET AL., JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY, vol. 50, no. 21, 2002, pages 6239 - 6244
SINGLETON, V.ROSSI, J. A., AMERICAN JOURNAL OF ENOLOGY AND VITICULTURE, vol. 16, no. 3, 1965, pages 144 - 158
SONG, L.THORNALLEY, P. J., FOOD AND CHEMICAL TOXICOLOGY, vol. 45, no. 2, 2007, pages 216 - 224
SREELATHA, S.PADMA, P. R., PLANT FOODS FOR HUMAN NUTRITION, vol. 64, no. 4, 2009, pages 303 - 311
SRIVASTAVA, S. K.SINGH, S. V., CARCINOGENESIS, vol. 25, no. 9, 2004, pages 1701 - 1709
TRAKA, M.MITHEN, R.: "Glucosinolates, isothiocyanates and human health", PHYTOCHEMISTRY REVIEWS, vol. 8, no. 1, 2009, pages 269 - 282, XP019686429
VONGSAK ET AL., INDUSTRIAL CROPS AND PRODUCTS, vol. 44, 2012, pages 566 - 571
WADSWORTH, T. L.KOOP, D. R., BIOCHEMICAL PHARMACOLOGY, vol. 57, no. 8, 1999, pages 941 - 949
WANG, Y.BEYDOUN, M. A., EPIDEMIOLOGIC REVIEWS, vol. 29, no. 1, 2007, pages 6 - 28
WU ET AL.: "Lipophilic and hydrophilic antioxidant capacities of common foods in the United States", JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY, vol. 52, no. 12, 2004, pages 4026 - 4037
XU CET, ONCOGENE, vol. 24, 2005, pages 4486 - 4495
XU ET AL., THE JOURNAL OF CLINICAL INVESTIGATION, vol. 112, no. 12, 2003, pages 1821 - 1830
Attorney, Agent or Firm:
BRASHEAR, Jeanne M. et al. (GERSTEIN & BORUN LLP233 S. Wacker Drive,6300 Willis Towe, Chicago IL, US)
Download PDF:
Claims:
What is claimed is:

1. A method of activating myrosinase present in a plant of the Moringaceae family comprising incubating injured seeds of the plant in a solution comprising water at a temperature of less than 100°C for a time sufficient to activate myrosinase present in the plant.

2. The method of claim 1, wherein the seeds are injured in a solution comprising water at room temperature.

3. The method of claim 2, wherein the injuring comprises pressing, slicing, blending, rolling, juicing, pulverizing or grinding the seeds of the plant.

4. The method of claim 1, wherein the plant of the Moringaceae family is a M. oleifera plant.

5. A method of producing a plant composition comprising contacting injured seeds of a plant of the Moringaceae family with a solution comprising water at a temperature of less than 100°C to produce the plant composition, wherein the plant composition comprises at least 0.1 wt% moringa isothiocyanates.

6. The method of claim 5, further comprising separating solid seed material from the plant composition.

7. A plant composition produced by the method of claim 5.

8. A method of producing an extract from a plant of the Moringaceae family comprising: contacting injured seeds from a plant of the Moringaceae family with an extraction fluid to produce the extract, wherein the injured seeds are incubated in a solution comprising water at a temperature of less than 100°C for a time sufficient to activate myrosinase present in the seeds prior to the contacting step.

9. The method of claim 8, the seeds are injured by a process selected from the group consisting of pressing, slicing, blending, juicing, rolling, pulverizing and grinding.

10. The method of claim 8, wherein the injured seeds are incubated in a solution comprising an amount of water at a ratio ranging from 1: 1 (w/v) to about 1:4 (w/v).

11. The method of claim 8, wherein the contacting step is performed at room temperature.

12. The method of claim 8, further comprising separating solid seed material from the extract.

13. The method of claim 12, wherein the separating step is performed using a method selected from the group consisting of filtration, sedimentation, centrifugation, evaporation, reduced-pressure distillation, precipitation and adsorption.

14. The method of claim 8, further comprising the step of drying the extract.

15. The method of claim 14, wherein the drying is performed by one or more methods selected from the group consisting of air drying, spray drying, speed vacuum, rotoevaporation and lyophilization.

16. The method of claim 8, wherein the contacting step comprises contacting the injured seeds with an extraction fluid in an amount ranging from about 1:5 to about 1:20 (w/w).

17. The method of claim 8, wherein the extraction fluid is 95% ethanol.

18. An extract produced by the method of claim 8.

19. The extract of claim 18, comprising at least 1% moringa isothiocyanates (MICs) per gram of injured seeds.

20. The extract of claim 18, that comprises at least 1% MIC-1 per gram of injured seeds.

21. A food product comprising the extract of claim 18.

22. The food product of claim 21, that is a baked good, a beverage, a dietary supplement or a medical food.

23. A cosmetic product comprising the extract of claim 18.

24. A method for maintaining healthy body weight in a mammalian subject in need thereof comprising administering the extract of claim 18 to the subject in need thereof in an amount sufficient to maintain a healthy body weight in the subject.

25. A method for promoting a healthy metabolism in a mammalian subject in need thereof comprising administering the extract of claim 18 to the subject in an amount sufficient to promote a healthy metabolism in the subject.

26. A method for treating a mammalian subject suffering from a metabolic disorder comprising administering to the subject in need thereof the extract according to claim 15 in an amount sufficient to treat the metabolic disorder.

27. The method of claim 26, wherein the metabolic disorder is selected from the group consisting of obesity, diabetes, satiety, endocrine deficiencies of aging, diabetes as a consequence of obesity, hyperglycemia, dyslipidemia, hypertriglyceridemia, metabolic syndrome, insulin resistance, impaired glucose tolerance (IGT), diabetic dyslipidemia, hyperlipidemia, a cardiovascular disease, and hypertension.

28. The method of claim 26, wherein the metabolic disorder is type II diabetes or obesity.

Description:
EXTRACTS FROM PLANTS OF THE MORINGACEAE FAMILY AND METHODS

OF MAKING

CROSS-REFERENCE TO RELATED APPLICATIONS

[0001] This application is a continuation-in-part of PCT/US2014/063178, filed October 30, 2014, which claims the benefit of priority to U.S. Provisional Application No. 61/898,795, filed November 1, 2013 and U.S. Provisional Application No. 62/032,496, filed August 1, 2014, the disclosures of which are incorporated herein by reference in their entireties.

BACKGROUND OF THE INVENTION

[0002] Moringa {Moringa oleifera L.) is a fast growing tropical tree known as the

"drumstick or horse radish tree." M. oleifera belongs to the monogenic family Moringaceae which contains only one genus and 13 species. The family is in the order Brassicales, to which broccoli and other cruciferous vegetables belong as members of Brassicaceae.

Moringa leaves are historically used as nutritious foods and traditional medicine in Asia and Africa. Moringa leaves contain approximately 27% protein by dry weight, and all essential amino acids. In addition, moringa leaves contain high levels of vitamins, and beneficial phytoactives (Pandey et al., 2012). These include polyphenols and four unique sugar- modified aromatic glucosinolates (GLSs; Bennett et al., 2003).

[0003] Moringaceae isothiocyanates (ITCs) are formed from their glycosylated precursors, glucosinolates, via a reaction carried out by myrosinase (thioglucoside glucohydrolase).

Myrosinase cleaves the thio-linked glucose in GLS, leaving the aglycone which rearranges quickly to form the active ITC. Despite well-documented health benefits of ITCs from crucifers, such as sulforaphane (SF) from broccoli and phenethylisothiocyanate from winter cress in treating inflammation and cancer, their clinical and dietary use is somewhat restricted because of their inherent chemical instability. For example SF, formed from broccoli glucoraphanin, its GLS precursor, is rapidly converted to several degradation products, mainly dimethyl disulfide and S-methyl methylthiosulfinate, making it difficult to formulate and deliver by means other than eating fresh vegetables (Franklin et al., 2013). Consuming ITCs from crucifers in their non-active, but more stable, GLS precursor form remains an option. However, GLSs undergo an uncertain and variable degree of enzymatic conversion to ITCs by host gut microbiota (Traka & Mithen, 2009) resulting in low yields and reduced or non-existent health benefits. SUMMARY OF THE INVENTION

[0004] The present disclosure is based in part on the discovery that the enzyme necessary to convert moringa glucosinolates (MGLs) into moringa isothiocyanates (MICs) (i.e., myrosinase) can be activated by simply injuring plant material (e.g., seeds, or fresh leaves or sprouts) of a plant of the Moringaceae family, without first subjecting the plant material to harsh conditions, such as harsh temperatures and drying conditions. Once the seeds, fresh leaves or sprouts of the plant are injured, the plant material can then be dried, stored and subject to extraction at a later time to retrieve the isothiocyanates.

[0005] In one aspect, described herein is a method of activating myrosinase present in a plant of the Moringaceae family comprising injuring fresh leaves or sprouts of the plant at a temperature of less than 100°C for a time sufficient to activate myrosinase present in the plant. In some embodiments, the injuring step is optionally performed at room temperature. In some embodiments, the injuring comprises pressing, slicing, blending, juicing, rolling, pulverizing or grinding fresh leaves or sprouts of the plant. In some embodiments, the plant of the Moringaceae family is a M. oleifera plant. In some embodiments, the method comprises injuring both leaves and sprouts of a plant of the Moringaceae family.

[0006] In another aspect, described herein is a method of activating myrosinase present in a plant of the Moringaceae family comprising incubating injured seeds of the plant in a solution comprising water at a temperature of less than 100°C for a time sufficient to activate myrosinase present in the plant. In some embodiments, the seeds are injured in a solution comprising water at room temperature. In some embodiments, the injuring comprises pressing, slicing, blending, juicing, rolling, pulverizing or grinding the seeds of the plant. In some embodiments, the plant of the Moringaceae family is a M. oleifera plant.

[0007] In another aspect, described herein is a method of producing a plant composition comprising injuring fresh leaves or sprouts of a plant of the Moringaceae family at a temperature of less than 100°C to produce the plant composition, wherein the plant composition comprises at least 0.1wt% moringa isothiocyanates. In some embodiments, the method comprises injuring both leaves and sprouts of a plant of the Moringaceae family. In some embodiments, the injuring is optionally performed at room temperature. In some embodiments, the injuring is performed in the presence of water. In some embodiments, the injuring is performed in the absence of water. [0008] In another aspect, described herein is a method of producing a plant composition comprising contacting injured seeds of a plant of the Moringaceae family with a solution comprising water at a temperature of less than 100°C to produce the plant composition, wherein the plant composition comprises at least 0.1 wt% moringa isothiocyanates. In some embodiments, the injuring is optionally performed at room temperature.

[0009] In some embodiments, the methods of producing a plant composition as described herein optionally comprises the step of separating solid plant material (i.e., seeds, leaves or sprouts) from the plant composition. The separating step can be performed using any method known in the art including, but not limited to, filtration, sedimentation, centrifugation, evaporation, including reduced-pressure evaporation (e.g., rotavap), reduced-pressure distillation (less than 100°C), precipitation, and adsorption.

[0010] In yet another aspect, described herein is a method of producing an extract from a plant of the Moringaceae family comprising contacting fresh injured leaves or sprouts of the plant with an extraction fluid comprising water at a temperature of less than 100°C to produce an extraction mixture; and separating solid leaves or sprouts from the extraction mixture to produce the extract.

[0011] In yet another aspect, described herein is a method of producing an extract from a plant of the Moringaceae family comprising contacting injured seeds from the plant with an extraction fluid to produce the extract, wherein the injured seeds are incubated in a solution comprising water at a temperature of less than 100°C for a time sufficient to activate myrosinase present in the seeds prior to the contacting step. In some embodiments, the injured seeds are incubated in a solution comprising an amount of water ranging from 1: 1 (w/v) to about 1:4 (w/v). In some embodiment, the seeds are injured in a solution comprising an amount of water ranging froml: l (w/v) to about 1:4 (w/v). The method optionally comprises separating solid seed material from the extract.

[0012] In some embodiments, the contacting step is optionally performed at room temperature. The separating step can be performed using any method known in the art including, but not limited to, filtration, sedimentation, centrifugation, evaporation, including reduced-pressure evaporation (e.g., rotavap), reduced-pressure distillation (less than 100°C), precipitation, and adsorption. In some embodiments, the methods further comprises injuring the seeds, leaves or sprouts by pressing, slicing, blending, juicing, rolling, pulverizing or grinding the seeds, fresh leaves or sprouts. In some embodiments, the methods optionally further comprises the step of drying the extract. Exemplary drying methods include, but are not limited to, air drying, spray drying, speed vacuum, rotoevaporation and lyophilization. In some embodiments, the method optionally comprises drying the injured fresh leaves or sprouts of the plant prior to the contacting step.

[0013] In yet another aspect, disclosed herein is a method of producing an extract from a plant of the Moringaceae family comprising injuring fresh leaves or sprouts of the plant; drying the injured fresh leaves or sprouts to produce dried injured fresh leaves or sprouts; contacting dried injured leaves or sprouts with an extraction fluid comprising water at a temperature of less than 100°C to produce an extraction mixture; and separating solid leaves or sprouts from the extraction mixture to produce the extract.

[0014] In any of the methods described herein, the methods optionally comprise contacting both leaves and sprouts of a plant of the Moringaceae family with the extraction fluid.

[0015] In some embodiments, the extraction fluid comprises at least 95% water. In some embodiments, the solvent mixture optionally comprises fresh leaves or sprouts to extraction fluid at a 1:5 (w/v) ratio.

[0016] In other embodiments, the extraction fluid comprises 95% ethanol. In such embodiments, the contacting step comprising contacting injured seeds with an extraction fluid in an amount ranging from 1:5 (w/w) to about 1:20 (w/w).

[0017] Plant compositions and extracts produced by the methods described herein are also provided. In some embodiments, the extracts produced by the methods disclosed herein comprise at least at least 0.5% moringa isothiocyanates per gram of fresh injured leaves or sprouts. In some embodiments, the extracts produced by the methods disclosed herein comprise at least 1.5% moringa isothiocyanates per gram of fresh injured leaves or sprouts. In some embodiments, the extracts produced by the methods described herein comprise at least 1% moringa isothiocyanates per gram of seeds In some embodiments, the moringa isothiocyanates are selected from the group consisting of 4-[(a-L-rhamnosyloxy)benzyl] isothiocyanate (MIC-1) and 4-[(4'-0-acetyl-a-L-rhamnosyloxy)benzyl] isothiocyanate (MIC- 4). In some embodiments, the moringa isothiocyanate is MIC-1.

[0018] In another aspect, the disclosure provides a method for maintaining healthy body weight in a mammalian subject in need thereof comprising administering a plant composition or extract prepared according to the methods described herein to the subject in an amount sufficient to maintain a healthy body weight in the subject. The phrase "healthy body weight" as used herein refers to a body weight that is within the normal range on the body mass index (BMI). BMI is a number calculated from a person's weight and height. A BMI of 19-24 is considered normal, while BMIs of 25-29 are defined as overweight. In some embodiments, the disclosure provides a method of promoting or maintaining a normal BMI comprising administering a plant composition or extract prepared according to the methods described herein to the subject in an amount sufficient to maintain or promote a normal BMI in the subject. In another aspect, the disclosure provides a method for promoting a healthy metabolism in a mammalian subject in need thereof comprising administering a plant composition or extract prepared according to the methods described herein to the subject in an amount sufficient to promote a healthy metabolism in the subject. In some embodiments, the subject is suffering from a metabolic disorder.

[0019] In another aspect, the disclosure provides a method for treating a mammalian subject suffering from a metabolic disorder comprising administering to the subject in need thereof a plant composition or extract produced by the methods described herein in an amount sufficient to treat the metabolic disorder. Exemplary metabolic disorders include, but are not limited to, diabetes (e.g., type I or type II diabetes), obesity, diabetes as a consequence of obesity, hyperglycemia, dyslipidemia, hypertriglyceridemia, syndrome X, insulin resistance, impaired glucose tolerance (IGT), diabetic dyslipidemia, hyperlipidemia, a cardiovascular disease, and hypertension. In some embodiments, the subject is suffering from type II diabetes. In some embodiments, the subject is suffering from obesity.

[0020] The subject may be, e.g., a human. In some embodiments, the plant composition or extract is administered to the subject over the course of, e.g., 1 year, 6 months, 3 months, 1 month, 2 weeks, 1 week, 3 days, or 1 day. In some embodiments, the subject may also be administered a second therapeutic for treating the metabolic disorder. Exemplary second therapeutics for treating the metabolic disorder include, but are not limited to, an antidiabetic agent, an antihyperuricemic agent, a lipid-lowering/lipid-modulating agent, or an anti-obesity agent, such as those described herein. In other embodiments, the second therapeutic is used for its known purpose and is selected from non- sulfonylurea secretagogues, glucagon-like peptides, exendin-4 polypeptides, PPAR agonists, dipeptidyl peptidase IV inhibitors, .alpha.- glucosidase inhibitors, immunomodulators, angiotensin converting enzyme inhibitors, adenosine Al receptor agonists, adenosine A2 receptor agonists, aldosterone antagonists, .alpha.1 adrenoceptor antagonists, .alpha.2 adrenoceptor agonists, angiotensin receptor antagonists, antioxidants, ATPase inhibitors, atrial peptide agonists, β adrenoceptor antagonists, calcium channel agonists, calcium channel antagonists, diuretics, dopamine Dl receptor agonists, endopeptidase inhibitors, endothelin receptor antagonists, guanylate cyclase stimulants, phosphodiesterase V inhibitors, protein kinase inhibitors, Cdc2 kinase inhibitors, renin inhibitors, thromboxane synthase inhibitors, vasopeptidase inhibitors, vasopressin 1 antagonists, vasopressin 2 antagonists, angiogenesis inhibitors, advanced glycation end product inhibitors, bile acid binding agents, bile acid transport inhibitors, bone formation stimulants, apolipoprotein Al agonists, DNA topoisomerase inhibitors, cholesterol absorption inhibitors, cholesterol antagonists, cholesteryl ester transfer protein antagonists, cytokine synthesis inhibitors, DNA polymerase inhibitors, dopamine D2 receptor agonists, endothelin receptor antagonists, growth hormone antagonists, lipase inhibitors, lipid peroxidation inhibitors, lipoprotein A antagonists, microsomal transport protein inhibitors, microsomal triglyceride transfer protein inhibitors, nitric oxide synthase inhibitors, oxidizing agents, phospholipase A2 inhibitors, radical formation agonists, platelet aggregation antagonists, prostaglandin synthase stimulants, reverse cholesterol transport activators, rho kinase inhibitors, selective estrogen receptor modulators, squalene epoxidase inhibitors, squalene synthase inhibitors, thromboxane A2 antagonists, cannabinoid receptor antagonists, cholecystokinin A agonists, corticotropin-releasing factor agonists, dopamine uptake inhibitors, G protein-coupled receptor modulators, glutamate antagonists, melanin- concentrating hormone receptor antagonists, nerve growth factor agonists, neuropeptide Y agonists, neuropeptide Y antagonists, serotonin-norepinephrine reuptake inhibitors (SNRIs), protein tyrosine phosphatase inhibitors, and serotonin 2C receptor agonists.

[0021] In any of the ranges described herein, the endpoints of the range are included in the range. Additional features and variations of the invention will be apparent to those skilled in the art from the entirety of this application and all such features are intended as aspects of the invention. Likewise, features of the invention described herein can be re-combined into additional embodiments that also are intended as aspects of the invention, irrespective of whether the combination of features is specifically mentioned above as an aspect or embodiment of the invention. Also, only such limitations which are described herein as critical to the invention should be viewed as such; variations of the invention lacking limitations which have not been described herein as critical are intended as aspects of the invention. BRIEF DESCRIPTION OF THE DRAWINGS

[0022] Figure 1 provides the chemical structures of moringa glucosinolates (MGLs) and moringa isothiocyanates (MICs) from M. oleifera and sulforaphane (SF) from broccoli.

[0023] Figure 2 shows the effect of dilution factor and temperature on isothiocyanate (MIC) content and percent yield in M. oleifera extract preparation. A. Effect of dilution ratio of fresh leaves (g):water (mL) on MIC concentration (mg of MIC/100 mg of extract). B. Effect of dilution ratio on extract percent yield (mg of extract/100 mg of fresh leaves). C. Effect of temperature on MIC concentrations (mg of MIC/ 100 mg of extract). D. Effect on temperature on extract percent yield (mg of extract/100 mg of fresh leaves).

[0024] Figure 3 provides a mass chromatogram of moringa glucosinolates (MGLs) and moringa isothiocyanates (MICs) at (A) 22°C and (B) 100°C.

[0025] Figure 4 shows the effect of storage of extract at 37°C on isothiocyanate (MIC) stability.

[0026] Figure 5 shows the anti-inflammatory effects of a moringa extract produced by the methods described herein, MIC-1, and MIC-4 on LPS-induced iNOS, IL-Ιβ, IL-6 and TNF-a gene expression in RAW 264.7 macrophage cells. Cells were pretreated for 2 hours with moringa extract or MICs and then induced with LPS for 6 hours. Values show relative gene expression compared to vehicle with LPS control, as determined by comparative AACt analysis. A: Effect of moringa extract on iNOS and IL-Ιβ. B: Effect of moringa extract on LPS-induced IL-6 and TNF-a. In A and B MIC a indicates the corresponding MIC

concentration present in moringa extract at the given doses. 1.6 μg/μL of MICs in the 100μg/uL moringa extract treatment corresponds to a MIC concentration of 5.5 μΜ (4 μΜ MIC-1 and 1.5 μΜ MIC-4). C: Effect of moringa extract on IL-6 and TNF-a. D: Effect of MICs on LPS-induced IL-6 and TNF-a.

[0027] Figure 6. Body weight gain (A), ratio of accumulated food intake to body weight (β), fat mass (C) and free fat mass (D) in VHFD and VHFD + 5% moringa extract-fed mice. n=12 mice per group, Data are means + SEM. Comparisons to controls were made by Welch's test. *P < 0.05; **P < 0.01; ***P < 0.001.

[0028] Figure 7. Oral glucose tolerance test performed at 4 (A), 8 (B) and 12 (C) weeks on mice fed VHFD, VHFD + 5% moringa extract, and on mice receiving VHDF gavaged with 300mg/kg metformin on the day of OGTT. Area Under the Curve of OGTT at 4, 8, and 12 weeks (D). n = 12 mice per group, except for metformin group where n=6 and only shown as a reference group. Data are means + SEM. Comparisons to controls were made by t-test. *P < 0.05; **P < 0.01 ; ***P < 0.001 in comparison of VHFD and VHFD + 5% moringa extract only.

[0029] Figure 8. Gross examination of liver samples from VHFD-fed mice (A) and VHFD + 5% moringa extract-fed mice (B). Liver weight in VHFD and VHFD + 5% moringa extract (n=12,) (C) Data are means + SEM. **: p<.01. Histological examination of liver samples from VHFD (D) and VHFD + 5% moringa extract (E). Fat content in liver from VHFD-fed mice and VHFD + 5% moringa extract-fed mice (n=12) (F). Comparisons to controls were made by Welch' s test. Data are means + SEM. **P < 0.01 ; ***P < 0.001.

[0030] Figure 9. Blood plasma expression of insulin, leptin, resistin (A), IL- Ιβ, TNFa (B), total cholesterol and triglycerides (C) in VHFD and VHFD + 5% moringa extract-fed mice. n=12 mice per group except for IL- Ιβ and TNFa where n=5, undetectable levels below 2.4 pg/mL were excluded. Comparisons to controls were made by Welch' s test. Data are means + SEM. * P < 0.05; ** P < 0.01.

[0031] Figure 10. Gene expression of inflammatory markers in liver (A) and ileum (B) of VHFD and VHFD + 5% moringa extract-fed mice (n = 12). Data are means + SEM.

Comparisons to controls were made by Welch's test for liver and ileum. *P < 0.05.

[0032] Figure 11. Effects of moringa extract, MIC-1, MIC-4 and sulforaphane (SF) on glucose production (A, B) and gene expression of G6P and PEPCK in HII4E liver cells; n=3 (C). Expression of G6P and PEPCK in hepatic tissue of VHFD and VHFD + 5% moringa extract-fed mice (D) n=12. Acute OGTT test in VHFD-fed mice gavaged with 2g/kg of moringa extract. (E) n = 6. Comparisons to controls were made by Dunnett' s test for A and C, t-test for D and Welch' s for E. Data are means + SEM. *: p<.05, **: p<.01, p<.001.

[0033] Figure 12. Effects of MICs, SF and moringa extract on glucose metabolism in vitro (A, B, C) and in vivo (D, E). Effects of MC, MIC- 1, MIC-4 and sulforaphane (SF) on glucose production (A, B) and gene expression of G6P and PEPCK in HII4E liver cells; n=3 (C). Expression of G6P and PEPCK in hepatic tissue of VHFD and VHFD + 5% MC-fed mice (P) n=12. Acute OGTT test in VHFD-fed mice gavaged with 2g/kg of MC (E) n = 6.

Comparisons to controls were made by Dunnett' s test for A and C, t-test for D and Welch' s for E. Data are means + SEM. *: p<.05, **: p<.01, p<.001. DETAILED DESCRIPTION

[0034] Moringa leaves contain considerable quantities of bioactive phytochemicals, including polyphenols and glucosinolates. While not biologically active, glucosinolates can be converted to isothiocyanates by the naturally occurring enzyme, myrosinase.

Isothiocyanates isolated from a plant of the Moringaceae family are structurally related to sulforaphane found in broccoli, but contain an unusual, if not a unique substitute rhamnose moiety which confers greatly enhanced stability and bioavailability compared to

sulforaphane.

Moringa glucosinolates (MGLs) and isothiocyanates (MICs)

MGLs MICs

[0035] As shown above, moringa glucosinolates (MGLs) contain an additional sugar moiety in the aglycone/isothiocyanate portion of the molecule. These MGLs can be converted in situ to four bioactive and relatively stable moringa isothiocyanates (MICs), referred to as MIC-lthrough MIC-4. MIC-1 (4-[(a-L-rhamnosyloxy) benzyl] isothiocyanate) and MIC-4 (4-[(4'-0-acetyl-a-L-rhamnosyloxy) benzyl] isothiocyanate) are the most abundant MICs, formed from MGL-1 and MGL-4. MICs are solid and relatively stable compounds at room temperature, in contrast to volatile isothiocyanates from crucifers that are mostly viscous liquids. The retained rhamnose sugar moiety found in MICs is extremely unique in nature and likely responsible for the high stability and solid appearance (Brunelli et al., 2010). [0036] The present disclosure is based in part on the discovery that the enzyme necessary to convert moringa glucosinolates (MGLs) into moringa isothiocyanates (MICs) (i.e., myrosinase) can be activated by simply injuring plant material (e.g., seeds, fresh leaves or sprouts) of a plant of the Moringaceae family, without first subjecting the plant material to harsh conditions, such as harsh temperatures and harsh drying conditions. This simple method can be used to effectively convert MGLs into MICs to produce a shelf-stable moringa plant extract containing more than 1.0% MICs. Harsh procedures used for the manufacture of moringa leaf powder, such as high temperatures or outdoor drying prior to injuring the plant material, usually lead to almost complete degradation of MGLs and MICs. As demonstrated in Example 2, analysis of several samples of moringa leaf powder from multiple commercial vendors confirmed the absence, or significant reduction, in levels of these desirable compounds. Similarly, the inventors have discovered that the use of a harsh solvent, such as 95% ethanol, in an extraction method prior to injuring seed material in a solution comprising water (or incubating injured seed material in a solution comprising water) inhibits formation of MICs. As demonstrated in Example 8, incubating injured seeds in a solution comprising water, or injuring seeds in a solution comprising water produced optimal conditions for activating myrosinase and forming MICs. Adding solvent (e.g., ethanol) to the injured seeds before water progressively inhibited formation of MICs. No detectable MICs were formed when 95% ethanol was directly added to the injured seeds or when the seeds were injured in 95% ethanol.

[0037] Thus, in one aspect, disclosed herein is a method of activating myrosinase present in a plant of the Moringaceae family comprising injuring fresh leaves or sprouts of the plant at a temperature of less than 100°C for a time sufficient to activate myrosinase present in the plant. In some embodiments, the plant of the Moringaceae family is a M. oleifera plant.

[0038] In another aspect, disclosed herein is a method of activating myrosinase present in a plant of the Moringaceae family comprising contacting injured seeds of the plant with a solution comprising water for a time sufficient to activate myrosinase present in the plant. In some embodiments, the contacting occurs at a temperature of less than 100°C.

[0039] The phrase "solution comprising water" as used herein refers to a solution comprising at least 50% water. In some embodiments, at least 55%, or at least 60%, or at least 65%, or at least 70%, or at least 75%, or at least 80%, or at least 90%, or at least 95%, or at least 99% or more of the solution comprises water. [0040] In some embodiments, the methods disclosed herein utilize fresh leaves or sprouts of a plant from the Moringaceae family. The term "fresh leaves or sprouts" of a plant of the Moringaceae family as used herein refers to leaves or sprouts of the plant that have not been dried or that have not been subjected to mechanical or chemical processing prior to their use in the methods disclosed herein.

[0041] In some embodiments, the methods disclosed herein utilize seeds of a plant from the Moringaceae family.

[0042] The term "injuring" as used herein refers to a method of processing seeds or the fresh leaves or sprouts of the plant such that the myrosinase present in the seeds, fresh leaves or sprouts of the plant is preserved and activated. In some embodiments, the "injuring" comprises pressing, slicing, blending, juicing, rolling, pulverizing or grinding the seeds, fresh leaves or sprouts of the plant.

[0043] Because the processing of moringa plant material at high temperatures is associated with the degradation of myrosinase in the plant material, the methods described herein are performed at a temperature of less than 100°C, optionally at a temperature ranging from 18°C to 100°C. In some embodiments, the methods described herein are performed at a

temperature of less than 90°C, or less than 85°C, or less than 80°C, or less than 75°C, or less than 70°C, or less than 65°C, or less than 60°C, or less than 55°C, or less than 50°C, or less than 45°C, or less than 40°C, or less than 35°C, or less than 30°C, or less than 25°C, or less than 20°C. In some embodiments, the methods described herein are performed at a temperature of aboutl8°C, about 19°C, about 20°C, about 21°C, about 22°C, about 23°C, about 24°C, about 25°C, about 26°C, about 27°C, about 28°C, about 29°C, about 30°C, about 31°C, about 32°C, about 33°C, about 34°C, about 35°C, about 36°C, about 37°C, about 38°C, about 39°C, about 40°C, about 41°C, about 42°C, about 43°C, about 44°C, about 45°C, about 46°C, about 47°C, about 48°C, about 49°C, about 50°C, about 51°C, about 52°C, about 53°C, about 54°C, about 55°C, about 56°C, about 57°C, about 58°C, about 59°C, about 60°C, about 61°C, about 62°C, about 63°C, about 64°C, about 65°C, about 66°C, about 67°C, about 68°C, about 69°C, about 70°C, about 71°C, about 72°C, about 73°C, about 74°C, about 75°C, about 76°C, about 77°C, about 78°C, about 79°C, about 80°C, about 81°C, about 82°C, about 83°C, about 84°C, about 85°C, about 86°C, about 87°C, about 88°C, about 89°C, about 90°C, about 91°C, about 92°C, about 93°C, about 94°C, about 95°C, about 96°C, about 97°C, about 98°C, or about 99°C. In some embodiments, the methods described herein are performed at room temperature. The term "room temperature" as used herein refers to a temperature generally ranging from 18°C to 25 °C.

[0044] Also described herein is a method of producing a plant composition comprising blending fresh leaves or sprouts of a plant of the Moringaceae family at a temperature of less than 100°C to produce the plant composition, wherein the plant composition comprises at least 0.05 wt% moringa isothiocyanates (MICs). A plant composition can also be prepared by a method comprising contacting injured seeds of a plant of a Moringaceae family with a solution comprising water at a temperature of less than 100°C to produce the plant composition, wherein the plant composition comprises at least 1 wt% MICs. In some embodiments, the plant of the Moringaceae family is a M. oleifera plant. The term "plant composition" as used herein refers to a composition obtainable from a plant of the

Moringaceae family without the use of an extraction fluid, as that term is defined below. In some embodiments, the plant composition comprises about 0.05 wt% MICs or about 0.1wt% MICs or about 0.2wt% MICs or about 0.3 wt% MICs. In some embodiments, the method of producing the plant composition is performed at a temperature of less than 100°C, optionally at a temperature ranging from 18°C to 100°C. In some embodiments, the method is performed at a temperature of less than 90°C, or less than 85°C, or less than 80°C, or less than 75°C, or less than 70°C, or less than 65°C, or less than 60°C, or less than 55°C, or less than 50°C, or less than 45°C, or less than 40°C, or less than 35°C, or less than 30°C, or less than 25°C, or less than 20°C. In some embodiments, the method is performed at a

temperature of aboutl8°C, about 19°C, about 20°C, about 21°C, about 22°C, about 23°C, about 24°C, about 25°C, about 26°C, about 27°C, about 28°C, about 29°C, about 30°C, about 31°C, about 32°C, about 33°C, about 34°C, about 35°C, about 36°C, about 37°C, about 38°C, about 39°C, about 40°C, about 41°C, about 42°C, about 43°C, about 44°C, about 45°C, about 46°C, about 47°C, about 48°C, about 49°C, about 50°C, about 51°C, about 52°C, about 53°C, about 54°C, about 55°C, about 56°C, about 57°C, about 58°C, about 59°C, about 60°C, about 61°C, about 62°C, about 63°C, about 64°C, about 65°C, about 66°C, about 67°C, about 68°C, about 69°C, about 70°C, about 71°C, about 72°C, about 73°C, about 74°C, about 75°C, about 76°C, about 77°C, about 78°C, about 79°C, about 80°C, about 81°C, about 82°C, about 83°C, about 84°C, about 85°C, about 86°C, about 87°C, about 88°C, about 89°C, about 90°C, about 91°C, about 92°C, about 93°C, about 94°C, about 95°C, about 96°C, about 97°C, about 98°C, or about 99°C. In some embodiments, the method is performed at room temperature, as that term is defined herein. [0045] The method optionally further comprises separating solid plant material (e.g., seeds, leaves or sprouts) from the plant composition. Exemplary methods of separation include, but are not limited to, filtration, sedimentation, centrifugation, evaporation, including reduced- pressure evaporation (e.g., rotavap), reduced-pressure distillation (less than 100°C), precipitation, and adsorption. In some embodiments, the resulting plant composition is dried, but the drying is performed post-injury, permitting endogenous myrosinase an opportunity to at least partially convert MGLs to MICs. Exemplary methods of drying the plant

composition include, but are not limited to, air drying, speed vacuum, rotoevaporation and lyophilization.

[0046] Also described herein are methods of producing an extract from a plant of the Moringaceae family. In some embodiments, the plant of the Moringaceae family is a M. oleifera plant. In some embodiments, the method comprises contacting fresh injured leaves or sprouts of the plant with an extraction fluid comprising water at a temperature of less than 100°C to produce an extraction mixture; and separating solid leaves and/or sprouts from the extraction mixture to produce the extract.

[0047] Also described herein are methods of producing an extract from seeds of a plant Moringaceae family. In some embodiments, the plant of the Moringaceae family is a M. oleifera plant. In some embodiments, the method comprises contacting injured seeds of the plant with an extraction fluid to produce the extract, wherein the injured seeds are incubated in a solution comprising water at a temperature of less than 100°C for a time sufficient to activate myrosinase present in the seeds prior to the contacting step. In some embodiments, the injured seeds are incubated in a solution comprising an amount of water ranging from 1: 1 (w/v) to about 1:4 (w/v). In some embodiment, the seeds are injured in a solution comprising an amount of water ranging froml: l (w/v) to about 1:4 (w/v).

[0048] The injured seeds are incubated in a solution comprising water (or injured in a solution comprising water) at a temperature of less than 100°C, optionally at a temperature ranging from 18°C to 100°C. In some embodiments, the method is performed at a

temperature of less than 90°C, or less than 85°C, or less than 80°C, or less than 75°C, or less than 70°C, or less than 65°C, or less than 60°C, or less than 55°C, or less than 50°C, or less than 45°C, or less than 40°C, or less than 35°C, or less than 30°C, or less than 25°C, or less than 20°C. In some embodiments, the method is performed at a temperature of aboutl8°C, about 19°C, about 20°C, about 21°C, about 22°C, about 23°C, about 24°C, about 25°C, about 26°C, about 27°C, about 28°C, about 29°C, about 30°C, about 31°C, about 32°C, about 33°C, about 34°C, about 35°C, about 36°C, about 37°C, about 38°C, about 39°C, about 40°C, about 41°C, about 42°C, about 43°C, about 44°C, about 45°C, about 46°C, about 47°C, about 48°C, about 49°C, about 50°C, about 51°C, about 52°C, about 53°C, about 54°C, about 55°C, about 56°C, about 57°C, about 58°C, about 59°C, about 60°C, about 61°C, about 62°C, about 63°C, about 64°C, about 65°C, about 66°C, about 67°C, about 68°C, about 69°C, about 70°C, about 71°C, about 72°C, about 73°C, about 74°C, about 75°C, about 76°C, about 77°C, about 78°C, about 79°C, about 80°C, about 81°C, about 82°C, about 83°C, about 84°C, about 85°C, about 86°C, about 87°C, about 88°C, about 89°C, about 90°C, about 91°C, about 92°C, about 93°C, about 94°C, about 95°C, about 96°C, about 97°C, about 98°C, or about 99°C. In some embodiments, the method is performed at room temperature, as that term is defined herein.

[0049] In some embodiments, the injured seeds are incubated with a solution comprising a volume of water at an exemplary ratio of 1: 1 (i.e., grams of seeds used to volume of water (mL)). In other embodiments, the injured seeds are incubated with a solution comprising a volume of water at an exemplary ratio of 1:2, or 1 :3, or 1:4, or 1:5. In some embodiments, the injured seeds are incubated with a solution comprising a volume of water at a ratio of 1:3.

[0050] In some embodiments, the method of producing the extract is performed at a temperature of less than 100°C, optionally at a temperature ranging from 18°C to 100°C. In some embodiments, the method is performed at a temperature of less than 90°C, or less than 85°C, or less than 80°C, or less than 75°C, or less than 70°C, or less than 65°C, or less than 60°C, or less than 55°C, or less than 50°C, or less than 45°C, or less than 40°C, or less than 35°C, or less than 30°C, or less than 25°C, or less than 20°C. In some embodiments, the method is performed at a temperature of aboutl8°C, about 19°C, about 20°C, about 21°C, about 22°C, about 23°C, about 24°C, about 25°C, about 26°C, about 27°C, about 28°C, about 29°C, about 30°C, about 31°C, about 32°C, about 33°C, about 34°C, about 35°C, about 36°C, about 37°C, about 38°C, about 39°C, about 40°C, about 41°C, about 42°C, about 43°C, about 44°C, about 45°C, about 46°C, about 47°C, about 48°C, about 49°C, about 50°C, about 51°C, about 52°C, about 53°C, about 54°C, about 55°C, about 56°C, about 57°C, about 58°C, about 59°C, about 60°C, about 61°C, about 62°C, about 63°C, about 64°C, about 65°C, about 66°C, about 67°C, about 68°C, about 69°C, about 70°C, about 71°C, about 72°C, about 73°C, about 74°C, about 75°C, about 76°C, about 77°C, about 78°C, about 79°C, about 80°C, about 81°C, about 82°C, about 83°C, about 84°C, about 85°C, about 86°C, about 87°C, about 88°C, about 89°C, about 90°C, about 91°C, about 92°C, about 93°C, about 94°C, about 95°C, about 96°C, about 97°C, about 98°C, or about 99°C. In some embodiments, the method is performed at room temperature, as that term is defined herein.

[0051] Solid plant material (e.g., seeds, leaves and/or sprouts) can be separated from the extraction mixture by any method known in the art including, but not limited to, filtration, sedimentation, centrifugation, evaporation, including reduced -pressure evaporation (e.g., rotavap), reduced-pressure distillation (less than 100°C), precipitation, and adsorption. In some embodiments, the separating step comprises filtering the extraction mixture to produce the extract. Any filter material and apparatus known in the art are contemplated for use in filtering the extraction mixture.

[0052] In other embodiments, the method of producing an extract from a plant of the Moringaceae family comprises injuring fresh leaves or sprouts of the plant, drying the injured leaves or sprouts, contacting dried injured leaves or sprouts with an extraction fluid comprising water at a temperature of less than 100°C to produce an extraction mixture, and separating solid leaves and/or sprouts from the extraction mixture to produce the extract.

[0053] The injured fresh leaves or sprouts of the plant are preferably dried at a temperature that permits endogenous myrosinase an opportunity to at least partially convert MGLs to MICs. In some embodiments, the injured fresh leaves or sprouts are dried using a method including, but not limited to, heat drying, air drying or microwaves. In some embodiments, the injured fresh leaves or sprouts are dried at a temperature of less than 100°C, optionally at a temperature ranging from 18°C to 100°C. In some embodiments, the injured fresh leaves or sprouts are dried at a temperature of less than 90°C, or less than 85°C, or less than 80°C, or less than 75°C, or less than 70°C, or less than 65°C, or less than 60°C, or less than 55°C, or less than 50°C, or less than 45°C, or less than 40°C, or less than 35°C, or less than 30°C, or less than 25°C, or less than 20°C. In some embodiments, the injured fresh leaves or sprouts are dried at a temperature of aboutl8°C, about 19°C, about 20°C, about 21°C, about 22°C, about 23°C, about 24°C, about 25°C, about 26°C, about 27°C, about 28°C, about 29°C, about 30°C, about 31°C, about 32°C, about 33°C, about 34°C, about 35°C, about 36°C, about 37°C, about 38°C, about 39°C, about 40°C, about 41°C, about 42°C, about 43°C, about 44°C, about 45°C, about 46°C, about 47°C, about 48°C, about 49°C, about 50°C, about 51°C, about 52°C, about 53°C, about 54°C, about 55°C, about 56°C, about 57°C, about 58°C, about 59°C, about 60°C, about 61°C, about 62°C, about 63°C, about 64°C, about 65°C, about 66°C, about 67°C, about 68°C, about 69°C, about 70°C, about 71°C, about 72°C, about 73°C, about 74°C, about 75°C, about 76°C, about 77°C, about 78°C, about 79°C, about 80°C, about 81°C, about 82°C, about 83°C, about 84°C, about 85°C, about 86°C, about 87°C, about 88°C, about 89°C, about 90°C, about 91°C, about 92°C, about 93°C, about 94°C, about 95°C, about 96°C, about 97°C, about 98°C, or about 99°C. In some embodiments, the injured fresh leaves or sprouts are dried at about 37°C. In some embodiments, the injured fresh leaves or sprouts are dried at room temperature, as that term is defined herein.

[0054] The term "extract from a plant of the Moringaceae family" as used herein means a substance or composition obtained from injured seeds and/or injured fresh leaves or sprouts of a plant of the Moringaceae family (or obtained from dried, injured fresh leaves or sprouts of a plant of the Moringaceae family, wherein the fresh leaves or sprouts were injured before being dried) through the use of an extraction fluid. Chemical and/or physical action, as would be understood in the art, may be required to obtain the substance or composition from the seeds, fresh leaves or sprouts of the plant (or obtained from dried, injured fresh leaves or sprouts of a plant of the Moringaceae family).

[0055] An "extraction fluid" for use in extraction methods includes water and well-known organic solvents such as, but not limited to, alcohols, alkanes, halocarbons, ethers, aromatic solvents, ketones, aqueous solvents, esters, and supercritical fluids. In some embodiments, ethanol is used to practice an extraction method described herein. In some embodiments, the extraction fluid used to practice the extraction methods described herein include, but are not limited to, 40% ethanol, 50% ethanol, 70% ethanol and 95% ethanol. Like water, a benefit of incorporating an ethanolic solvent in extraction method is that an ethanolic solvent is compatible with an ingestible product, and therefore is suitable for incorporation of the extract into a pill, capsule, tablet, or other ingestible form known in the art. In some embodiments, the extraction fluid comprises at least 90%, or at least 91%, or at least 92%, or at least 93%, or at least 94%, or at least 95%, or at least 96%, or at least 97%, or at least 98%, or at least 99% water. In some embodiments, the extraction fluid comprises less than 10%, or less than 9%, or less than 8%, or less than 7%, or less than 6%, or less than 5%, or less than 4%, or less than 3%, or less than 2%, or less than 1% of an organic solvent other than water. Exemplary organic solvents other than water include, but are not limited to, straight and branched chain alkanes, alcohols, ethers, esters, aldehydes, ketones, and hydrocarbons of CI to CIO, e.g., ethanol, methanol, n-butanol, n-propanol and isopropanol. In some embodiments, the extraction fluid comprises 95% ethanol.

[0056] In some embodiments, the injured fresh leaves or sprouts of the plant are contacted with a volume of extraction fluid at an exemplary ratio of 1: 1 (i.e., grams of fresh plant material used to volume of extraction fluid (mL)). In other embodiments, the injured fresh leaves or sprouts of the plant are contacted with a volume of extraction fluid at an exemplary ratio of 1:2, or 1:3, or 1:4, or 1:5, or 1:6, or 1:7, or 1:8, or 1:9 or 1: 10. In some

embodiments, the injured fresh leaves or sprouts of the plant are contacted with a volume of extraction fluid at a ratio of 1:5.

[0057] In some embodiments, the injured seeds (that are either incubated in a solution comprising water after injury, or are injured in a solution comprising water prior to the contacting step) are contacted with a volume of extraction fluid at an exemplary ratio of 1:5 (e.g., grams of injured seed material to volume of extraction fluid (mL). In other

embodiments, the injured seeds are contacted with a volume of extraction fluid at an exemplary ratio of 1:6, or 1:7, or 1:8, or 1:9, or 1: 10, or 1: 11, or 1: 12, or 1: 13, or 1: 14 or 1: 15, or 1: 16, or 1: 17, or 1: 18, or 1: 19, or 1:20. In some embodiments, the injured seeds are contacted with a volume of extraction fluid at a ratio of 1:5.

[0058] The extract produced by the extraction methods described herein comprises a high concentration of moringa isothiocyanates (MICs) compared to extracts produced using a dried moringa leaf powder as the starting material. In some embodiments, the extract comprises at least 0.5% MICs per gram of plant material (e.g., seeds, leaves or sprouts) used in the extraction method. In some embodiments, the extract comprises at least 0.6%, or at least 0.7%, or at least 0.8%, or at least 0.9%, or at least 1%, or at least 1.1%, or at least 1.2%, or at least 1.3%, or at least 1.4%, or at least 1.5%, or at least 1.6%, or at least 1.7%, or at least 1.8%, or at least 1.9%, or at least 2%, or at least 5%, or at least 10% or more MICs per gram of plant material (e.g., seeds, leaves or sprouts) used in the extraction method. In some embodiments, the extract comprises about 1% MICs per gram of plant material (e.g., seeds, leaves or sprouts) used in the extraction method. In some embodiments, the extract comprises about 1.5% MICs per gram of plant material (e.g., seeds, leaves or sprouts) used in the extraction method.

[0059] The MICs present in an extract produced by the methods disclosed herein demonstrate greater stability than other isothiocyanates, such as sulforaphane. For example, in some embodiments, an extract produced by the methods disclosed herein comprise a MIC that demonstrated less than 50% degradation when the extract was stored at 37°C for about 30 days compared to the MIC present in the extract at day 0. In some embodiments, a MIC in the extract degrades less than 50%, less than 45%, less than 40%, less than 35%, less than 30%, less than 25%, less than 20%, less than 19%, less than 18%, less than 17%, less than 16%, less than 15%, less than 14%, less than 13%, less than 12%, less than 11%, less than 10%, less than 9%, less than 8%, less than 7%, less than 6%, less than 5%, less than 4%, less than 3%, less than 2% or less than 1% when the extract is stored at 37°C for about 30 days. In some embodiments, MIC-4 in the extract produced by the methods disclosed herein degrades less than 20% when the extract is stored at 37°C for about 30 days compared to the amount of MIC-4 present in the extract at day 0.

[0060] Extracts obtained from plants of the Moringaceae family contain other beneficial phytochemicals, such as polyphenols, flavonols, carotenoids, and ascorbic acid. Polyphenols found in plants of the Moringaceae family include, but are not limited to, to 5-caffeoylquinic acid (5-CQA), 3-caffeoylquinic acid (3-CQA), quercetin 3-O-rutinoside, quercetin 3-O- glucoside, kaempferol 3-O-rutinoside, quercetin 3-0-(6"-malonylglucoside), kaempferol 3- O-glucoside, quercetin 3-0-(X"-malonylglucoside),isorhamnetin 3-O-glucoside, quercetin 3- 0-(X"-acetylglucoside, quercetin 3-0-(Y"-malonylglucoside), kaempferol 3-0-(6"- malonylglucoside), isorhamnetin 3-0-(6"-malonylglucoside), kaempferol 3-0-(X"- malonylglucoside), kaempferol 3-0-(X"-acetylglucoside), quercetin aglycone, kaempferol aglycone, isorhamnetin aglycone. The most abundant being 5-caffeoylquinic acid (5-CQA) known as chlorogenic acid, quercetin-3-O-rutinoside known as rutin, quercetin 3-O-glucoside and quercetin 3-0-(6"-malonylglucoside). In some embodiments, an extract produced by the methods described herein comprises (in addition to a high concentration of MICs) at least 1% total polyphenol content per gram of plant material used in the extraction method. In some embodiments, an extract produced by the methods described herein comprises at least 1%, or at least 2%, or at least 3%, or at least 4%, or at least 5%, or at least 6%, or at least 7%, or at least 8%, or at least 9%, or at least 10% total polyphenol content per gram of plant material. In some embodiments, an extract produced by the methods described herein comprises a total polyphenol content ranging from 2-5%, or 1-3%, or 2-4%, or 1-5%, or 3-5%, or 3-7% or 4- 8% or 5-10% per gram of plant material.

Use of the plant compositions or extracts

[0061] In some embodiments, a plant composition or extract produced by the methods described herein is incorporated into consumer products. Consumer products are products available for purchase and/or use by individual consumers and include food products (including, but not limited to, enriched food products (see below), dietary supplements (see below) and medical foods (see below)), cosmetic products and other personal care products. [0062] In some embodiments the plant composition or extract produced by the methods described herein is incorporated into a food product to produce an enriched food product. The term "food product" as used herein refers to any substance containing nutrients that can be ingested by an organism to produce energy, promote health and wellness, stimulate growth, and maintain life. In some embodiments, the plant composition or extract produced by the methods described herein is used in the preparation of enriched food products comprising high amounts of MICs. The term "enriched food product" as used herein refers to a food product that has been modified to include the plant composition or extract produced by the methods described herein described herein, which provides a benefit such as a health/wellness-promoting and/or disease-preventing/mitigating/treating property beyond the basic function of supplying nutrients.

[0063] The plant composition or extract produced by the methods described herein can be incorporated into any food product. Exemplary food products include, but are not limited to, baked goods (cakes, cookies, crackers, breads, scones and muffins), dairy-type products (including, but not limited to, cheese, yogurt, custards, rice pudding, mousses, ice cream, frozen yogurt, frozen custard), desserts (including, but not limited to, sherbet, sorbet, water- ices, granitas and frozen fruit purees), spreads/margarines, pasta products and other cereal products, meal replacement products, nutrition bars, trail mix, granola, beverages (including, but not limited to, smoothies, water or dairy beverages, and soy-based beverages), and breakfast-type cereal products such as oatmeal. For beverages, the plant composition or extract (or MICs isolated from the plant composition or extract) may be in solution, suspended, emulsified or present as a solid.

[0064] In one embodiment, the enriched food product is a meal replacement product. The term "meal replacement product" as used herein refers to an enriched food product that is intended to be eaten in place of a normal meal. Nutrition bars and beverages that are intended to constitute a meal replacement are types of meal replacement products. The term also includes products which are eaten as part of a meal replacement weight loss or weight control plan, for example snack products which are not intended to replace a whole meal by themselves, but which may be used with other such products to replace a meal or which are otherwise intended to be used in the plan. These latter products typically have a calorie content in the range of from 50-200 kilocalories per serving.

[0065] In another embodiment, the food product is a dietary supplement. The term

"dietary supplement" as used herein refers to a substance taken by mouth that contains a "dietary ingredient" intended to supplement the diet. The term "dietary ingredient" includes, but is not limited to, the MICs as disclosed herein, as well as vitamins, minerals, herbs or other botanicals, amino acids, and substances such as enzymes, organ tissues, glandulars, and metabolites.

[0066] In yet another embodiment, the food product is a medical food. The term "medical food" as used herein means a food which is formulated to be consumed or administered entirely under the supervision of a physician and which is intended for the specific dietary management of a disease or condition for which distinctive nutritional requirements, based on recognized scientific principles, are established by medical evaluation.

[0067] In some embodiments, the plant composition or extract produced by the methods described herein (or MICs isolated from the plant composition of extract) are useful as cosmeceuticals. The term "cosmeceutical" as used herein means an ingredient for a cosmetic, body care or hair care personal product having a positive effect on the physical condition of the body (e.g., the skin, the nails, or hair).

[0068] Compositions suitable for personal care products generally are formulated as, e.g., shampoos, conditioners, shower gels, liquid hand cleansers, facial cleansers, moisturizers, lotions, skin lotions and creams (such as eye creams and lip creams), facial skin cosmetics (such as blusher and highlighter), eye cosmetics (such as eye shadow, eye brow color, and eye liner), lip cosmetics (such as lip rouge), foundation, concealer, wrinkle- smoothing serums or creams, mascaras, skin facial masks, sunscreens, scalp hair-styling aids, facial hair-styling aids, emulsions, oils, mousses, ointments, milks, pomades, solutions, sprays, aerosols, powders, foams, gels (such as skin gels, eye gels, and lip gels), or other skin or hair products known in the art.

Additional Uses

[0069] The data provided herein demonstrate that serum levels of insulin, leptin, resistin, triglycerides and cholesterol (all of which are associated with metabolic disorders and healthy body weight maintenance) were reduced in animals receiving a moringa extract produced by the methods described herein. Thus, the disclosure also provides a method for maintaining healthy body weight in a mammalian subject in need thereof comprising administering a plant composition or extract prepared according to the methods described herein to the subject in an amount sufficient to maintain a healthy body weight in the subject. The phrase "healthy body weight" as used herein refers to a body weight that is within the normal range on the body mass index (BMI). BMI is a number calculated from a person's weight and height. A BMI of 19-24 is considered normal, while BMIs of 25-29 are defined as overweight. In some embodiments, the disclosure provides a method of promoting or maintaining a normal BMI comprising administering a plant composition or extract prepared according to the methods described herein to the subject in an amount sufficient to maintain or promote a normal BMI in the subject.

[0070] In another aspect, the disclosure provides a method for promoting a healthy metabolism in a mammalian subject in need thereof comprising administering a plant composition or extract prepared according to the methods described herein to the subject in an amount sufficient to promote a healthy metabolism in the subject. In some embodiments, the subject is suffering from a metabolic disorder.

[0071] In another aspect, the disclosure provides a method for treating a mammalian subject suffering from a metabolic disorder comprising administering to the subject in need thereof a plant composition or extract produced by the methods described herein in an amount sufficient to treat the metabolic disorder. In some embodiments, the subject is suffering from type II diabetes. In some embodiments, the subject is suffering from obesity.

[0072] The term "metabolic disorder" is used broadly herein to refer to the conditions, diseases, and disorders associated with insulin and/or glucose dysregulation. Such disorders include those resulting from an alteration in glucose homeostasis resulting, for example, in hyperglycemia. In some embodiments, an alteration in glucose levels is an increase in glucose levels by at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or even 100% relative to such levels in a healthy individual. Metabolic disorders include, but are not limited to, obesity and diabetes (e.g., diabetes type I, diabetes type II, MODY, and gestational diabetes), satiety, endocrine deficiencies of aging, diabetes as a consequence of obesity, hyperglycemia, dyslipidemia, hypertriglyceridemia, syndrome X (metabolic syndrome), insulin resistance, impaired glucose tolerance (IGT), diabetic dyslipidemia, hyperlipidemia, a cardiovascular disease, and hypertension. Metabolic disorders are also described in Kinzig et al., J. Neurosci. 23:6163-6170, 2003, which is hereby incorporated by reference.

[0073] By "treating" is meant ameliorating at least one symptom of a condition or disease in a subject having the condition or disease (e.g., a subject diagnosed with a metabolic disorder), as compared with an equivalent untreated control. Such reduction in the symptom (e.g., a reduction in blood glucose levels or weight) is at least 5%, 10%, 20%, 40%, 50%, 60%, 80%, 90%, 95%, or 100%, as measured by any standard technique.

[0074] In some embodiments, a desired outcome of treatment is the ability to reduce glucose levels in the subject. The phrase "reducing glucose levels" refers to reducing the level of glucose in a blood sample from the subject by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 100% relative to an untreated control. In some

embodiments, glucose levels are reduced to normoglycemic levels, i.e., levels between 150 to 60 mg/dl, between 140 to 70 mg/dl, between 130 to 70 mg/dl, between 125 to 80 mg/dl, or between 120 to 80 mg/dl.

[0075] In some embodiments, a desired outcome of treatment is the ability to maintain a healthy body weight. The phrase "healthy body weight" as used herein refers to a body weight that is within the normal range on the body mass index (BMI). BMI is a number calculated from a person's weight and height. A BMI of 19-24 is considered normal, while BMIs of 25-29 are defined as overweight. In some embodiments, the disclosure provides a method of promoting or maintaining a normal BMI comprising administering a plant composition or extract prepared according to the methods described herein to the subject in an amount sufficient to maintain or promote a normal BMI in the subject.

Formulations and Dose Regimens

[0076] The disclosure contemplates compositions comprising a plant composition or extract produced by the methods described herein (or MICs isolated from such plant compositions and/or extracts) that are, in some embodiments, tabletted, encapsulated or otherwise formulated for oral administration. The compositions may be provided as pharmaceutical compositions, nutraceutical compositions (e.g., a dietary supplement), or as a food or beverage additive, as defined by the U.S. Food and Drug Administration. The dosage form for the above compositions is not particularly restricted. For example, liquid solutions, suspensions, emulsions, tablets, pills, capsules, sustained-release formulations, powders, suppositories, liposomes, microparticles, microcapsules, sterile isotonic aqueous buffer solutions, and the like are all contemplated as suitable dosage forms.

[0077] The compositions typically include one or more suitable diluents, fillers, salts, disintegrants, binders, lubricants, glidants, wetting agents, controlled release matrices, colorings, flavoring, carriers, excipients, buffers, stabilizers, solubilizers, commercial adjuvants, and/or other additives known in the art. [0078] Any pharmaceutically acceptable (i.e., sterile and acceptably non-toxic as known in the art) liquid, semisolid, or solid diluent that serves as a pharmaceutical vehicle, excipient, or medium can be used. Exemplary diluents include, but are not limited to, polyoxyethylene sorbitan monolaurate, magnesium stearate, calcium phosphate, mineral oil, cocoa butter, and oil of theobroma, methyl- and propylhydroxybenzoate, talc, alginates, carbohydrates, especially mannitol, a-lactose, anhydrous lactose, cellulose, sucrose, dextrose, sorbitol, modified dextrans, gum acacia, and starch. Such compositions may influence the physical state, stability, rate of in vivo release, and rate of in vivo clearance of the functional compounds that are compatible with the disclosed methods and extracts comprising relatively stabilized MICs.

[0079] Pharmaceutically acceptable fillers can include, for example, lactose,

microcrystalline cellulose, dicalcium phosphate, tricalcium phosphate, calcium sulfate, dextrose, mannitol, and/or sucrose. Salts, including calcium triphosphate, magnesium carbonate, and sodium chloride, may also be used as fillers in the pharmaceutical

compositions.

[0080] Binders may be used to hold together the composition containing the enriched substance to form a hard tablet. Exemplary binders include materials from organic products such as acacia, tragacanth, starch and gelatin. Other suitable binders include methyl cellulose (MC), ethyl cellulose (EC) and carboxymethyl cellulose (CMC).

[0081] In some embodiments, the composition further comprises a bioavailability enhancer, which acts to increase the absorption of the MICs by the body. Bioavailability enhancers can be natural or synthetic compounds. In one embodiment, the enriched food product comprising the enriched solid further comprises one or more bioavailability enhancers in order to enhance the bioavailability of the bioactive natural product(s).

[0082] Natural bioavailability enhancers include ginger, a caraway extract, a pepper extract and chitosan. The active compounds in ginger include 6-gingerol and/or 6-shogoal. Caraway oil can also be used as a bioavailability enhancer (U.S. Patent Application Publication No. 2003/022838). Piperine is a compound derived from pepper (Piper nigrum or Piper longum) that acts as a bioavailability enhancer (U.S. Pat. No. 5,744,161). Piperine is available commercially under the brand name Bioperine® (Sabinsa Corp., Piscataway, N.J.). In some embodiments, the natural bioavailability enhancers is present in an amount of from about 0.02% to about 0.6% by weight based on the total weight of enriched food product. [0083] Examples of suitable synthetic bioavailability enhancers include, but are not limited to, Gelucire ® ., Labrafil ® and Labrasol ® , Lauroglycol ® , Pleural Oleique ® (Gattefosse Corp., Paramus, N.J.) and Capmul ® (Abitec Corp., Columbus, Ohio).

[0084] The amount and administration regimen of the composition is based on various factors relevant to the purpose of administration, for example human or animal age, sex, body weight, hormone levels, or other nutritional need of the human or animal. In some embodiments, the composition is administered to an animal in an amount from about 0.001 mg/kg body weight to about 10 g/kg body weight. In some embodiments, the composition is administered to an animal in an amount of about 0.005 mg/kg body weight. In some embodiments, the composition is administered to an animal in an amount of about 0.01 mg/kg body weight, or about 0.05 mg/kg body weight, or about 0.1 mg/kg body weight, or about 1 mg/kg body weight, or about 10 mg/kg body weight, or about 100 mg/kg body weight, or about 250 mg/kg body weight, or about 500 mg/kg body weight, or about 1 g/kg per body weight, or about 2.5 g/kg body weight, or about 5 g/kg body weight, or about 7.5 g/kg body weight, or about 10 g/kg body weight.

[0085] A typical regimen may comprise multiple doses of the composition. In one embodiment, the composition is administered once per day and may be administered to an individual at any time. In some embodiments, the composition is administered concurrently, prior to, or at the consumption of a meal. The composition is administered on any periodic schedule suitable for the desired or needed effect, or on an as-needed basis.

[0086] It will be appreciated that the plant composition and extract produced by the methods described herein is useful in the fields of human medicine and veterinary medicine to provide high levels of MICs to a subject in need thereof. Thus, the subject or individual to be treated may be a mammal, such as a human. For veterinary purposes, subjects include, for example, farm animals such as cows, sheep, pigs, horses, and goats; companion animals such as dogs and cats; exotic and/or zoo animals; laboratory animals including mice, rats, rabbits, guinea pigs, and hamsters; and poultry such as chickens, turkeys, ducks, and geese.

EXAMPLES

[0087] The following Examples are provided to describe the invention in greater detail, and are intended to illustrate, not to limit, the appended claims. Example 1 provides the materials and methods for the experiments performed in Example 2. Example 2 describes the optimization of the extraction method. Example 3 demonstrates that the moringa extracts prepared as described in Example 2 have anti-inflammatory activity. Example 4 provides additional parameters for the preparation of moringa plant compositions and extracts.

Example 5 provide an alternative method for the preparation of a moringa extract. Example 6 provides the materials and methods for the experiments performed in Example 7. Example 7 demonstrates that the moringa extracts prepared as described in Example 2 reduced body weight and fat accumulation in mice. Example 8 provides materials and methods for the preparation of moringa plant compositions and extracts from seeds of a M. oleifera plant.

Example 1 - Materials and Methods

[0088] Plant Material and Sample Preparation. Fresh leaves and seeds from M. oleifera (Indian PKM-1 variety) were shipped overnight from Moringa Farms, CA. The leaves were extracted using the methods disclosed herein on the day of arrival to produce a moringa extract. Moringa seeds were cultivated at the Rutgers University greenhouse until the plants flowered. A voucher specimen (CWl) was prepared and deposited at the Chrysler Herbarium of Rutgers University (CHRB).

[0089] Fresh M. oleifera leaves were blended (Vitamix 5200 Blender, Cleveland, OH) thoroughly with room temperature Millipore water in a ratio of 1 g of leaves to 5 mL of water (1:5) for moringa extract preparation used in stability tests, all biological assays and batch reproducibility assessment. Micro preparation of moringa extract for temperature/dilution optimization was performed by grinding fresh leaves in a coffee grinder (Krups, Millville, NJ) and then placing them in water. The leaf extracts (either prepared with the blender or coffee grinder) were placed on a shaker for 30 min at room temperature. In temperature experiments, the extracts were placed in water baths at designated temperatures for 30 min. Following incubation, the extracts were filtered through Miracloth (Calibiochem, Billerica, MA) and centrifuged for 10 min at 3200 g and 4°C. The supernatant, which appeared as a brown clear tea was decanted and lyophilized to produce moringa extract. In some cases, particularly with larger batches, centrifugation was repeated in order to clear all solid materials from the supernatant.

[0090] Compound Extraction and Isolation. MIC-1 and MIC-4 were isolated from fresh moringa leaves using a modified approach to previously published methods (Cheenpracha et al., 2010). Briefly, MICs were initially extracted from ground leaves in methanol (MeOH). The methanolic extract was dried down and partitioned in H 2 0 and hexanes (1: 1 v/v). An equal volume of ethyl acetate (EtOAc) was then added to the H 2 0 fraction. The EtOAc fraction was dried down and resuspended in acetonitrile (CAN): H 2 0 (1: 1), sonicated briefly, and filtered through a 0.45 μηι filter prior to preparative high-performance liquid

chromatography (HPLC).

[0091] Replicate HPLC injections of 100 μΐ. of the EtOAc fraction (200 mg/mL) were eluted with ACN/H 2 0/trifluoroacetic acid (TFA) (50:50:0.05) to yield MIC-1 (retention time (Rt)= 8.2 min) and MIC-4 (Rt = 17.5 min). Reverse-phase HPLC was carried out on a Waters System consisting of a four-channel Waters 616 pump with semi-preparative pump heads operated on a Waters 600 Controller; Waters 490E Programmable Multiwavelength Detector set to monitor at 222 nm; and a Waters 717 Plus Autosampler. A Phenomenex semi- preparative Synergi Hydro column (4μΜ, 250 x 20 mm) was run with a flow rate of 10 mL/min.

[0092] Compound Quantification. The chemical purity of isolated MICs was confirmed by liquid chromatography mass spectrometry (LCMS) and 1H NMR. The UV peak area of LCMS injections of MIC-1 and MIC-4 (> 98% purity) at 3 concentrations (3x) were averaged and used to generate standard curves to quantify MIC content in moringa extract

preparations. One μL· injections (3x) of MIC-1 at 20, 100, and 200 ng/μL dissolved in ACN: H 2 0 (1: 1) generated a standard curve (y = 123x-0.098, R2= 1) and MIC-4 at 10, 50, and 100 ng/μί generated a standard curve (y = 104.32x-0.098, R2> 0.99).

[0093] LCMS analysis was performed using the Dionex ® UltiMate 3000 RSLC ultra-high- pressure liquid chromatography system, consisting of a workstation with Dionex ® ' s

Chromeleon v. 6.8 software package, solvent rack/degasser SRD-3400, pulseless

chromatography pump HPG-3400RS, autosampler WPS-3000RS, column compartment TCC-3000RS, and photodiode array detector DAD-3000RS. After passing the photodiode array detector, the eluent flow was guided to a Varian 1200L (Varian Inc., Palo Alto, CA) triple quadrupole mass detector with electrospray ionization interface, operated in negative ionization mode. The voltage was adjusted to -4.5 kV, heated capillary temperature was 280°C, and sheath gas was compressed air, zero grade, for the negative ionization mode. The mass detector was used in scanning mode from 65 to 1500 atomic mass units. Data from the Varian 1200L mass detector was collected, compiled and analyzed using Varian' s MS Workstation, v. 6.9, SP2. Compounds were separated on a Phenomenex™ C8 reverse phase column, size 150 x 2 mm, particle size 3 μιη, pore size 100 A. The mobile phase consisted of 2 components: Solvent A (0.5% ACS grade acetic acid in double-distilled de-ionized water, pH 3-3.5), and Solvent B (100% Acetonitrile). The mobile-phase flow was 0.20 mL/min, and a gradient mode was used for all analyses. The initial conditions of the gradient were 95% A and 5% B over 30 min the proportions of A and B continuously changed, reaching 5% A and 95% B, which was kept for the next 8 min. During the following 4 min, the ratio was brought to initial conditions. An 8 min equilibration interval was included between subsequent injections. 1H NMR spectra were recorded in methanol-d4 on a 500 Varian VNMRS 500 MHz.

[0094] Optimization and Reproducibility of Extraction. Moringa extract was prepared in ratios of 1:2, 1:5, and 1: 10 (g of fresh leaves: mL water) for optimization of MIC content and percent yield. Triplicate samples of fresh moringa leaves (8 g) were ground in a coffee grinder, diluted accordingly in room temperature water and mixed for 30 min. For temperature experiments, fresh moringa leaves (8 g) were ground in a coffee grinder and added to water (40 mL) at 22, 40, 60, 80, and 100°C as triplicate samples. The mixtures were maintained at these temperatures for 30 min in temperature-controlled water baths.

Following 30 minutes of incubation, moringa extract was prepared as described herein.

Analysis for percent yield (weight of moringa extract as a percent of starting fresh weight of leaves) and MIC content were determined.

[0095] Once the optimum temperature (22°C) and dilution factor (1:5) were established using micro preparations, larger batches of moringa extract were made using a Vitamix blender (200 g: 1000 mL). Triplicate samples of moringa extract prepared in this manner from three separate batches of moringa leaves were compared for reproducibility tests.

[0096] Compound Stability. Triplicate 100 mg samples of optimized moringa extract were placed in a 37°C dark incubator and subjected to LCMS analysis for quantification of MICs at O, 18 and 30 days.

[0097] Characterization of Extract. The optimized moringa extract preparation, prepared with 22°C water at a ratio of fresh leaves to water of 1:5 (w/v) was additionally characterized for total polyphenol content (TP) and oxygen radical absorbance capacity (ORAC). Total polyphenols were quantified by the Folin-Ciocalteu method (Singleton & Rossi, 1965) and samples were read at 726 nm against a gallic acid standard curve. ORAC was determined as μΜ Trolox equivalents (TE) using fluorescein as the fluorescent probe and 2,2'-azobis(2- amidinopropane) dihydrochloride(AAPH) as a peroxyl radical generator in a procedure adapted from previously published methods (Prior et al., 2003). [0098] Cell Culture. All reagents were supplied from Sigma- Aldrich Co. (St. Louis, MO) unless otherwise noted. RAW 264.7 macrophages (ATCC TIB-71) were maintained in Dulbecco's modified Eagle's medium (Caisson, North Logan, UT) supplemented with 100 U/mL penicillin, 100 μg/mL streptomycin, and 10% fetal bovine serum. Cells were incubated at 37°C in a 5% C0 2 humidified atmosphere and subcultured by cell scraping. For experiments, RAW cells were plated at a density of 4 x 10 5 cells/mL in 24-well plates. Cells were incubated overnight (18 h), washed with warm 37°C PBS, and medium was replaced fresh DMEM medium. Cells were pretreated with designated doses of moringa extract, MICs or vehicle (50% EtOH). Lipopolysaccharide (1 μg/mL) was added after 2 h incubation with treatments to elicit inflammatory responses. Cells were treated in duplicate or triplicate. After an additional 6 h incubation period, media were collected and cells were washed with PBS prior to collection in TRIzol ® Reagent (Ambion, Life Technologies). Samples were stored at -80°C prior to processing.

[0099] Gene Expression Analyses. Total RNA was extracted from cells according to manufacturer's specifications. Briefly, 200 of chloroform was added to 600 of TRIzol-harvested samples. Samples were vigorously mixed for 30 s, incubated at room temperature for 5 min, and centrifuged at 12,400 g Eppendorf tube and isopropanol was added to the aqueous phase to obtain a ratio of 7: 10 supernatant to isopropanol. Samples were mixed by inverting, vortexed briefly and incubated for 10 min at -20°C. Samples were centrifuged at 12,400 g for 15 min at 4°C. Next, supernatant was removed and each sample was washed twice with 75% ethanol and centrifuged at 6000 g for 10 min. Samples were allowed to dry and resuspended in diethylpyrocarbonate (DEPC)-treated-water. RNA integrity was evaluated by running aboutl μg of RNA on a 1% agarose gel.

[00100] RNA was then treated with Deoxyribonuclease I (DNAse I) Amplification grade (Invitrogen), following the manufacturer' s guidelines. RNA quality was checked on the NanoDrop 1000 system (NanoDrop Technologies). A ratio of OD 260:280 > 2.0 and OD 260:230 > 1.8 was considered to be good quality RNA. First-strand cDNA synthesis was performed using the ABI High-Capacity cDNA Reverse Transcription kit (Applied

Biosystems, Foster City, CA) with RNAse I inhibitor, according to the manufacturer' s instructions, using 1 μg of RNA. The thermal cycle program was set as follows: 10 min, 25°C; 60 min, 37°C; 60 min, 37°C; 5 s, 85°C; and final hold at 4°C.

[00101] Synthesized cDNAs were diluted 25-fold and 5 of dilution was used for qPCR with 12.5 μΐ ^ of Power SYBR Green PCR master mix (Applied Biosystems), 0.5 μΐ ^ primers (6 μΜ) and BPC grade water (Sigma) to a final reaction volume of 25 μh. Exon-spanning primer sequences were designed on Primer Express® (Life Tech) and are as follows: β-actin forward 5'- AAC CGT GAA AAG ATG ACC CAG AT - 3' (SEQ ID NO: 1), reverse: 5'- CAC AGC CTG GAT GGC TAC GT-3' (SEQ ID NO: 2), IL-Ιβ forward 5'- CAA CCA ACA AGT GAT ATT CTC CAT - 3' (SEQ ID NO: 3), reverse 5'- GAT CCA CAC TCT CCA GCT GCA - 3' (SEQ ID NO: 4), iNOS forward 5'- CCC TCC TGA TCT TGT GTT GGA - 3' (SEQ ID NO: 5), reverse 5'- TCA ACC CGA GCT CCT GGA A-3' (SEQ ID NO: 6), COX-2 forward 5' - TGG TGC CTG GTC TGA TGA TG -3' (SEQ ID NO: 7), reverse 5'- GTG GTA ACC GCT CAG GTG TTG-3' (SEQ ID NO: 8), TNF-a forward 5'- TGG GAG TAG ACA AGG TAC AAC CC - 3' (SEQ ID NO: 9), reverse 5'- CAT CTT CTC AAA ATT CGA GTG AGA A - 3' (SEQ ID NO: 10), IL-6 forward 5' - TCG GAG GCT TAA TTA CAC ATG TTC - 3' (SEQ ID NO: 11), reverse 5' TGC CAT TGC ACA ACT CTT TTC T - 3' (SEQ ID NO: 12). All primers were validated by analyzing amplification efficiencies and melt-curve profiles.

[00102] Quantitative PCR amplifications were performed on an ABI 7300 Real-Time PCR System (Applied Biosystems) with the following thermal cycler profile: 2 minutes, 50°C; 10 minutes, 95°C; 15 seconds, 95°C; 1 minute, 60°C for the dissociation stage; 15 seconds, 95°C; 1 minutes, 60°C; and 15 seconds, 95°C. Inflammatory marker mRNA expressions were validated and samples were analyzed by the comparative AACt method and normalized with respect to the average Ct value of β-actin. Vehicle with LPS served as the calibrator for AACt analysis and was assigned a value of 1.0. Lower values indicate inhibition of gene expression relative to vehicle treated with LPS control. All experimental samples were run in triplicate and each reaction plate included no-template controls.

[00103] TNF-a Secretion Analysis. RAW 264.7 macrophages were cultured and treated with moringa extract or MICs as stated above. After treatments, 1 mL of media was collected and immediately centrifuged at 13,500 g at 4°C for 10 minutes. The supernatant was preserved at -80°C until further processing with the BD OptEIATM Mouse TNF ELISA kit (BD Bioscience, San Jose, CA) following the manufacturer's protocol. All the samples were assayed in duplicate. TNF-a levels were quantified using a reference standard curve provided with the kit. Absorbance was read at 450 nm and corrected at 570 nm.

[00104] Nitric Oxide Production Analysis. RAW 264.7 macrophages were cultured and treated with moringa extract or MICs as stated herein. After treatments, 1 mL of media was collected and assayed in duplicate following the Griess Reagent System provided by Promega (Promega Corporation; Madison, WI). The nitrite standard (0.1 M sodium nitrite) reference curve was built performing a serial dilution (0 to 100 μΜ). Absorbance was read at 540 nm.

[00105] Cell Viability. The effect of the treatments on cell viability was measured using MTT [3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyltetrazolium bromide] (TCI, Portland, OR) (Mosmann, 1983). MTT (5 mg/mL) was dissolved in PBS (Cayman Chemical, Ann Arbor, MI), filtered through a 0.22 μιη membrane and added to treated cells during the last 3-4 hours of treatment. Media were carefully aspirated and cells were dissolved in DMSO. The absorbance was read at 570 nm.

[00106] Statistical Analysis. Data were expressed as mean + SEM. Statistical

comparisons for optimization experiments were made by use of 1-way analysis of variance (ANOVA) followed Tukey's post-hoc test in the moringa extract optimization and stability experiments. Statistical comparisons for anti-inflammatory experiments were made by use of ANOVA followed by a Dunnett's or Wilcoxon test, as indicated, and p < 0.05 were considered significant. *** = p < 0.001, ** = p < 0.01, * = p < 0.05. For statistical analysis, GraphPad Prism version 6.02 for Windows (GraphPad Software, Inc.) was used.

Example 2 - Optimization of Extraction Method

[00107] Experiments were performed to optimize the in situ biotransformation of MGLs into MICs by myrosinase and to maximize the yield of MICs present in fresh leaves. The solvent ratio (weight of fresh leaves to volume of water) and temperature (22 -100°C) were tested to determine the optimal conditions for moringa extract yield and MICs content. The solvent ratio affected both the concentrations of MICs and the percent yield (Fig. 2A & 2B). The 1:2 solvent ratio resulted in a lower average MIC-1 content (0.45% of moringa extract) compared with the 1:5 and 1: 10 dilution, (0.59% and 0.62% of moringa extract, respectively). The amount of MIC-4 was higher in the 1:5 and 1 : 10 dilutions, but not to a statistically significant degree (0.12% of moringa extract compared with 0.20%, 0.19% respectively). Larger dilutions resulted in a proportional percent yield increase of moringa extract: (1:2) 5.47%, (1:5) 6.13%, (1: 10) 7.87%. The 1:5 dilution factor was selected as optimum to maximize the amount of MICs captured in moringa extract, while minimizing the amount of water used for extraction.

[00108] The 1:5 solvent ratio was used in the evaluation of the effect of water temperature on MIC concentration and percent yield. There was a significant difference in the amount of MICs extracted at water temperatures between 22 to 80°C (MIC-1: 0.49-0.72% of moringa extract, MIC-4: 0.19-0.21% of moringa extract) with undetectable amounts at 100°C (Fig. 2C). The LCMS mass chromatogram of MGLs and MICs in a 22°C and 100°C extraction shows the heat sensitivity of myrosinase activity at high temperatures (Fig. 3). At 22°C myrosinase converted significant quantities of MGLs to MICs. At 100°C myrosinase was inactivated and MGLs were not converted to their respective MICs. The thermal stability of moringa myrosinase is similar to broccoli myrosinase, with a reported thermal stability to 50- 60°C (Eylen, Oey, Hendrickx, & Loey, 2008) and complete destruction of the enzyme above 80°C (Gallaher, Gallaher, & Peterson, 2012). Extraction with the 1:5 solvent ratio at room temperature (22°C) was adopted to maintain full enzymatic conversion of MGLs into MICs.

[00109] Once moringa extract preparation had been optimized over temperature and solvent ratio, larger scale production of moringa extract required the use of a blender instead of a coffee grinder. This unintended parameter of scaling up the extraction resulted in a significant increase in MIC content. Under the same conditions, 1:5 solvent ratio at 22°C, the coffee grinder produced a lower amount of MIC-1 and MIC-4, approximately 1.00% total, in moringa extract while the blender increase the concentration of total MICs to 1.66% of moringa extract. This was likely due to finer fractionating of the leaves and the presence of water at the time of blending rather than grinding prior to combining with water in the case of the coffee grinder. Use of the blender, like use of the coffee grinder, did not create the harsh conditions of pulverization and not characteristic of prior art methods. It is expected that fine chopping of plant materials (e.g., use of or cutting or slicing implement moving at blender speeds) at lower temperatures (e.g., 4°C to 60°C or 80°C) will yield significant quantities of MICs from moringa plant materials.

[00110] Preparation of moringa extract using the blender was performed with three separate batches of moringa leaves and subjected to MIC quantification by LCMS to ensure reproducibility of the extraction method. The content of MICs in moringa extract (1.66%) is approximately 3 times higher than the SF content obtained from broccoli sprouts (calculated by a reported 61% conversion rate of glucoraphanin to SF and converted to dry weight factoring 89% moisture content (Force, O'Hare, Wong, & Irving, 2007; Pereira et al., 2002; Song & Thornalley, 2007). Moringa extracts, prepared in large batches, were subsequently evaluated for chemical stability of MICs, total polyphenol content, oxygen radical absorbance capacity and anti-inflammatory activity in vitro.

[00111] Stability of Compounds. The accelerated stability studies of MICs in moringa extract at 37°C for 30 days showed approximately 80% and 20% degradation of MIC-1 and MIC-4, respectively (Fig. 4) compared to day 0. The higher stability of MIC-4 may be due to the monoacetylation at the 4' position of the rhamnose sugar. Greater acetylation is known to increase the stability of glycosylated molecules such as anthocyanins (Giusti & Wrolstad, 2003). However, both MIC-1 and MIC-4 demonstrated superior stability compared to reported values of SF, the main broccoli isothiocyanate, which degraded by 75% after 6 days at 37°C (Franklin et al., 2013). SF is a volatile, viscous liquid, whereas MICs obtained by the methods disclosed herein are solids at room temperature. This is may be due to the higher molecular weight and rhamnose substitution compared with SF.

[00112] Total polyphenol Content. Total polyphenol (TP) content of moringa extract, determined by the Folin-Ciocalteu method (Singleton & Rossi, 1965) was 3.82 mg of gallic acid equivalents per 100 mg of moringa extract (+ 0.22), which is similar to the reported TP content of dried moringa leaves (3.6 to 4.5 % DW) (Sreelatha & Padma, 2009). This indicates that the aqueous moringa extract extraction methods disclosed herein captured the majority of polyphenols present in fresh leaves. Predominant polyphenols identified in moringa include rutin, chlorogenic acid, and quercetin-malonyl-glucoside (Amaglo et al., 2010; Bennett et al., 2003). The molecular weights of these compounds were detected by LCMS analyses of moringa extract, but quantification of specific polyphenols was not performed.

[00113] Oxygen Radical Absorbance Capacity. The ORAC value of moringa extract was 3.6 mmol Trolox equivalents (TE) per gram of moringa extract (+0.69 SEM). This is greater than reported values for spices with high ORAC, such as dried cinnamon powder (2.6 mmol TE per gram) (Wu et al., 2004). Fresh and dried moringa leaves were previously reported to contain high levels of antioxidant compounds, including phenolics, flavonols, carotenoids and ascorbic acid (Siddhuraju & Becker, 2003; Dillard & German, 2000). Antioxidants in various moringa leaf extracts (total polyphenols, total flavonoids) have been shown in vitro to possess free -radical scavenging activity and ferric-reducing power (Vongsak et al., 2012). In vivo moringa extracts have also been shown to increase the antioxidant activity of reduced glutathione, superoxide dismutase, and catalase, while decreasing lipid peroxidation (Moyo, Oyedemi, Masika, & Muchenje, 2012).

[00114] Moringa' s antioxidant capacity has primarily been attributed to the presence of polyphenols and flavonoids, while little attention has been paid to the antioxidant potential of MGLs and MICs present in moringa extracts. Yet, isothiocyanates (ITCs) from crucifers have been shown to possess strong antioxidant activity. SF, the primary ITC in broccoli, is one of the most potent inducers of phase II enzymes (Traka & Mithen, 2009). Reduction of oxidants has been correlated with reduced pathogenesis of inflammation (Geronikaki & Gavalas, 2006).

Example 3 - Moringa extracts demonstrate anti-inflammatory activity

[00115] Moringa extract produced by the methods disclosed herein demonstrated a dose dependent inhibitory effect on iNOS and IL-Ιβ gene expression in RAW 264.7 macrophages in vitro (Fig. 5A). Tested concentrations of moringa extracts ranged from 5 to 100 μg/mL (0.1% to 1.6% MIC content). The molar concentration of MICs in moringa extract at the various doses ranged from approximately 0.28 μΜ to 5.5 μΜ. Almost complete suppression of iNOS and IL-Ιβ gene expression was observed at 100 μg/mL of moringa extract (5.5 μΜ MIC content).

[00116] Purified MIC-1 and MIC-4 tested at 1 and 5 μΜ concentrations also showed significant reduction of mRNA expression of iNOS and IL-Ιβ (Fig. 5B). Additionally, moringa extract at 100 μg/mL (Fig. 5C) and MIC-1 at 5 μΜ (Fig. 5D) were able to decrease IL-6 gene expression significantly. However, no reduction of TNF-a gene expression was seen at any of the concentrations of moringa extract, MIC-1 and MIC-4 tested.

[00117] Nitric oxide (NO) and TNF-a cytokine production were reduced by moringa extract, MIC-1 and MIC-4 (Fig. 6 A & B). Moringa extract at 100 μg/mL, containing 1.15% MIC-1 and 0.51% MIC-4, inhibited TNF-a production by 70% compared to the control. MIC-1 and MIC-4 at 5 μΜ reduced TNF-a production by 20% and 27%, respectively. The enhanced anti-inflammatory activity of moringa extract compared with MIC-1 and MIC-4 alone could be the result of additive or synergistic activities of moringa extract polyphenols or could be the presence of less abundant, but perhaps highly active MIC-2 and MIC-3. These results demonstrate the plausible advantage of delivering MICs in a food-grade product. TNF-a RNA expression was not significantly inhibited by moringa extract, MIC-1, or MIC-4, indicating that moringa phytochemicals may inhibit TNF-a production at the translational level or at the level of TNF-a turnover.

[00118] Moringa extract, MIC-1 and MIC-4 inhibited the production of NO significantly (Fig. 6 A & B). This is consistent with previously reported NO inhibition by MIC-1 and MIC-4 (IC50 of 14.43 and 2.71 μΜ, respectively) (Cheenpracha et al., 2010). Moringa extract, at 100 μg/mL, was able to inhibit NO formation by 90%. MIC-1 and MIC-4 are at least partially responsible for this effect, because they inhibited NO formation at 5 μΜ. Moringa extract also contains MIC-2 and MIC-3, reported to inhibit NO formation at low micromolar concentrations (IC50 of 1.67 μΜ and 2.66 μΜ, respectively (Park et al., 2011)). Moringa extract, MIC-1 and MIC-4 showed no signs of cytotoxicity at the concentrations tested in anti-inflammatory assays, as demonstrated in MTT-based cell viability assays.

Example 4 - Additional parameters for preparation of moringa plant compositions and extracts

[00119] Moringa plant compositions and extracts are prepared in a variety of ways to accommodate situations where outdoor cultivation or manufacturing equipment is limiting. For example, moringa can be grown indoors as sprouts (5-10 days old) and readily used for the preparation of moringa extract. Fresh moringa leaves can be injured without the addition of water, by blending or rolling, to activate myrosinase and convert MGLs to MICs. This injured material can be easily dried, sold as is, or shipped for further extraction/concentration of MICs. Alternatively moringa extract can be made in al : 1 dilution to increase the concentration of MICs, while still allowing for rapid drying (solar/ oven dryers). Provided below in Table 1 is a list of various methods of extraction and filtration performed and percent yield (product dry weight as a percent of the starting fresh weight), % MIC-1 (as determined by LCMS as a percent of product dry weight), % MIC-4 (as determined by LCMS as a percent of product dry weight) and MIC-4:MIC-1 ratio of the resulting plant compositions and extracts.

[00120] Table 1.

a Product dry weight as a percent of starting fresh weight

b Amount of MIC-l/MIC-4 determined by LCMS as a percent of product dry weight

c Leaves were ground in a blender (Vitamix 5200 Blender, Cleveland, OH)

d Leaves were placed on a screen and crushed with a rolling pin

c Filtered using Miraclofh (Calibiochem, Billerica, MA) and hand squeezed

f Dilution faction is stated as the ratio of fresh leaves used (g): amount of water used (mL)

E Centrifuged for 10 min at 3200 g and 4°C

h Mixture was placed in a Juicer (Jack LaLanne's Ultimate Power Juicer, Fairfield, NJ)

[00121] A 1:5 dilution ratio provided a significantly higher concentration of MICs compared to the 1: 1 dilution ratio. The best preparation method for the 1:5 ratio was determined to be blending, followed by juicing. Centrifugation in all cases led to lower levels of MIC-4, the more stable MIC and is thus not recommended. Drying the 1:5 ratio preparations can be achieved by rotoevaporation, followed by freeze-drying or spray drying. The 1:5 ratio preparations were subjected to rotoevaporation for 1 hour at 50°C which removed approximately 75% of the water. This increased the solid concentration to levels required for spray drying. No significant loss of either MIC-1 or MIC-4 was observed under rotoevaporation conditions when compared to samples that were freeze-dried. Further concentration of MICs and removal of sugars from the extract can also be performed with solid phase-extraction (SPE).

Example 5 - Additional extraction method

[00122] Fresh moringa leaves were injured (e.g., crushed with mortar and pestle) to bioconvert MGLs to MICs and then dried at 37°C for 18 hours. The resulting injured and dried plant material can be used in this condition as a product with enhanced MIC content or stored/shipped and processed at a later date by extraction for further concentration of MICs. Extraction of these crushed-dried leaves in a 1:5 ratio (g of fresh weight equivalents: mL of water) resulted in a MIC-1 content of 1.01% and MIC-4 content of 0.57%. This

demonstrated bioconversion of MGLs to MICs in the crushing step, stability of MICs in the drying, storage and potential shipping step, and concentration of MICs in the extraction step. This procedure allows for spatial and temporal separation between a supply of fresh moringa leaves and extraction concentration.

Example 6 - Materials and Methods

[00123] Materials: Moringa extract was produced by the methods disclosed herein. Food formulation for the experiments provided in Example 7 was standardized to deliver 800 mg of MICs/kg of food. In the long-term study, a very high-fat diet (VHFD) (60% kcal from fat) contained 5% moringa extract (1.66% MIC by DW). The diet was formulated by Research Diets (New Brunswick, NJ) to be isocaloric for fat, protein and carbohydrate content (Suppl. Table 1).

[00124] Animals: Twenty-four male C57BL/6J mice at 5 weeks of age were obtained from Jackson Laboratories (Bar Harbor, ME). Mice were acclimated for 9 days and housed 4 animals per cage under a 12-hour light/dark cycle, with ad libitum access to water and a VHFD or VHFD + 5% moringa extract for twelve weeks. Body weight and food intake was recorded weekly. Food intake was estimated as follows: [total food consumed per

cage]/[mice per cage]x[day of food consumption]. Body composition was determined at 4, 8, and 12 weeks by magnetic resonance imaging using an EchoMRI-100 instrument (Echo Medical Systems, Houston, TX). At the end of the study, mice were euthanized with carbon dioxide. Blood and tissues (liver, inguinal fat, gastrocnemius muscle and ileum) were collected immediately and preserved at -80 °C. [00125] Oral glucose tolerance test (OGTT): Mice in the three-month study were first fasted overnight before fasting glycemic levels were recorded using a glucometer

(AlphaTRAK® 32004-02, Abbott Animal Health, Abbott Park, IL) and gavaged with 2 g/kg of glucose at weeks 4, 8 and 12 weeks of treatment. An additional six mice on the VHFD at the same age were gavaged with 300 mg/kg of metformin (positive control) 3 hours prior to glucose gavage. Glycemic levels were measured at intervals up to 120 minutes.

[00126] Acute OGTT: Fifteen male C57BL/6J mice were purchased, acclimated and housed as described in the 3-month study. Mice were fed ad libitum a VHFD for 12 weeks. The OGTT was performed as described above except for gavage treatments of 2 g/kg of moringa extract(n = 6), water (vehicle; n = 6), or 300 mg/kg of metformin (n = 3).

[00127] Blood chemistry analysis: Animals were fasted overnight and trunk blood was collected immediately after euthanization. Samples were collected in tubes with EDTA and plasma was aliquotted into cryovials and stored at -80°C for analysis. Insulin, leptin, resistin, interleukin- 1 beta (IL-Ιβ) and tumor necrosis factor alpha (TNFa) were measured using a multiplex assay (Millipore, Temecula, CA) measured on a Luminex 200 (Luminex, Austin, TX). Total cholesterol and triglycerides (TG) were assayed on a DxC 600 Pro (Beckman Coulter, Inc., Indianapolis IN).

[00128] Liver histology, total lipid extraction, and TG levels: Randomly selected liver sections were fixed in 10% neutral-buffered formalin for 48 hours, then processed and embedded in Paraplast. Six-micrometer sections were cut and stained in hematoxylin and eosin. A diagnosis of fatty liver was made based on the presence of macro or microvesicular fat > 5% of the hepatocytes in a given slide. Total lipid content of liver and feces was determined by Folch's method (19). Briefly, liver (about 300 mg) and feces (about 200 mg) were extracted 20: 1 (v/w) with CHC 12 /CH 3 OH (2: 1), followed by solvent evaporation and DW calculation.

[00129] Gene expression analysis by quantitative RT-PCR:

[00130] Liver and ileum. Total RNA was isolated from liver and ileum for TNFa, IL-Ιβ, interleukin-6 (IL-6) expression; and additionally for glucose-6 phosphatase (G6P), PEPCK and glucokinase (GcK) expression from liver tissue using the PureLink® RNA mini kit plus on-column DNase treatment (Applied Biosystems, Foster City, CA). Tissue (100 mg) was homogenized with TRIzol® using zirconium beads in a Bead Bug homogenizer (Benchmark Scientific, Inc. Edison, NJ). First-strand cDNA was synthetized from 2 μg total RNA using the high capacity cDNA reverse transcription kit plus RNase inhibitor (Applied Biosystems) with oligo-d(T)s as primers. PCR analyses were performed on a 7300 Real-Time PCR system using the TaqMan Assays (Applied Biosystems). Hydroxymethylbilane synthase (Hmbs) was used to normalize target gene expression and effect of treatment on gene expression levels was evaluated by the AACt method (20).

[00131] In vitro gluconeogenesis studies. H4IIE rat hepatoma cells (CRL-1548, American Type Culture Collection, Manassas, VA) were assayed for glucose production as previously described (21). Cell viability was measured by the 3-(4,5-Dimethyl-2-thiazolyl)-2,5- diphenyltetrazolium bromide (MTT; TCI, Portland, OR) assay (22). RNA extraction, cDNA synthesis and qPCR for gene expression of PEPCK and G6P were performed as described above.

Example 7 - Moringa extract reduced body weight and fat accumulation in mice.

[00132] This Example determined the effect of a moringa extract produced by the methods disclosed herein on body weight, body composition, OGTT, liver composition and lipid content of mice fed either a very high-fat diet (VHFD) + moringa extract or a VHFR without moringa extract (control).

[00133] The VHFD + 5% moringa extract— fed mice gained significantly less weight over the 3-month study compared to the VHFD-control mice (P<0.001 from 4-12 weeks) with a final average weight of 38.4 ± 1.0 g vs. 46.9 + 1.0 g (mean + SEM), respectively (Figure 6A). All animals involved in the study looked healthy at the end of the study with no adverse effects noticed. Weekly food consumption remained stable throughout the 12-week study, averaging 2.22 + 0.02 g /day for the VHFD + 5% moringa extract group versus 2.42 + 0.05 g/d for control mice. The 5% moringa extract diet contained 800 mg of MICs/kg. Therefore, the mice were consuming approximately 66 mg of MICs per day. Accumulated food intake only became significantly less in the VHFD + 5% moringa extract-fed group at the 12th week (P < 0.05). The ratio of accumulated food intake to body weight, however, was significantly higher in the VHFD + 5% moringa extract-fed mice compared to the VHFD group

throughout the entire study (Figure 6B). Body composition at 4, 8 and 12 weeks showed lower fat accumulation (Figure 6C) and greater free fat (lean mass) as a percentage of body weight in the VHFD + 5% moringa extract-fed mice compared to the VHFD-fed mice (Figure 6D). [00134] OGTT performed at 4, 8 and 12 weeks demonstrated lower blood-glucose levels and faster return to fasting levels in VHFD + 5% moringa extract -fed mice compared to VHFD-fed mice (Figure 7). Compared to fatty livers of VHFD-fed mice, livers from the VHFD + 5% moringa extract-fed animals did not show the appearance of fatty-liver disease (Figures 8A and 8B) as also evident from the histological comparison (Figures 8D and 8E). The livers of VHFD + 5% moringa extract-fed mice weighed less (Fig. 8C) and contained lower levels of lipids in relation to the VHFD-fed mice (Figure 8F). There was no significant difference in the lipid content as a percent of dry fecal weight from the two experimental groups (VHFD, 0.47 + 0.14%; VHFD + 5% moringa extract, 0.46 + 0.04%).

[00135] Effect ofmoriga extract on blood composition. VHFD + 5% moringa extract- fed mice had lower blood plasma levels of glucose regulators (insulin, leptin, resistin) (Figure 9A), inflammatory cytokines (IL-Ιβ and TNFa) (Figure 9B), cholesterol and triglycerides (Figure 9C) compared to the VHFD group. Reduced gene expression of pro-inflammatory markers, TNFa, IL-6, and IL-Ιβ, were observed in the liver (Figure 10A) and ileum (Figure 10B) tissue from the VHFD + 5% moringa extract-fed mice compared to the VHFD group.

[00136] Effect of moringa extract and MICs on glucose metabolism and OGTT. Moringa extract (produced by the methods disclosed herein) and MICs significantly reduced glucose production by approximately 60% in HII4E liver cells at 10 μg/mL and 1 μΜ, respectively (P < 0.001). MIC-1 and MIC-4 demonstrated superior activity to SF at the same concentrations (Figure 11A). To further explore the activity of MICs in comparison to the prescription drug metformin, MIC-4 and metformin were tested over a range of 5 concentrations, showing IC 50 of glucose production at 7 μΜ for MIC-4 versus 800 μΜ for metformin (Figure 1 IB).

Moringa extract and MICs also significantly decreased expression of G6P and PEPCK in HII4E liver cells relative to the vehicle (Figure 11C). G6P expression was significantly lower in the hepatic tissue of VHFD + 5% moringa ex tract- fed mice compared to the controls (Figure 1 ID). Glucose lowering effects of moringa extract were further tested in vivo by the acute OGTT, to eliminate the weight difference variable in the long-term feeding study. The acute OGTT resulted in significantly lower blood glucose levels at 15 and 30 minutes in the moringa extract-gavaged mice (2 g/kg) compared to the vehicle (Figure HE).

[00137] This study provides justification and mechanistic evidence for the uses of moringa extract prepared as disclosed herein as a dietary agent in preventing type 2 diabetes by demonstrating that MIC-enriched moringa extract caused significant reduction in weight gain, hepatic adiposity, gluconeogenesis, insulin, cholesterol, and inflammatory markers. This study also establishes the role of MICs as primary anti-diabetic actives in moringa extract. The most notable result of the long-term feeding study was the significant reduction in weight gain observed in the moringa extract-fed mice. Healthy C57BL/6J mice fed a low fat diet (10% kcal from fat) typically gain 25-32% less weight than mice on a VHFD (25, 26). In this experiment, the moringa extract-fed mice gained 18% less weight than the VHFD-fed mice, demonstrating almost complete abolition of excess weight gain caused by the VHFD, without any other observable side effects. Slight differences in accumulated food intake or food aversion cannot explain the reduced weight gain in moringa extract-fed mice, because the ratio of accumulated food intake to body weight was actually higher in the VHFD + 5% moringa extract-fed mice compared to the VHFD group. Previous in vitro work

demonstrated MICs and MC possess anti-inflammatory activity manifested as decreased IL- 1β and TNFa expression and nitric oxide (NO) production (2); effects that were also observed in this in vivo study. TNFa over-expression was previously identified as a contributing factor to obesity-induced type 2 diabetes (27), particularly by studies showing TNFa knockout mice had increased insulin sensitivity (28-30). However, only slight decreases in body weight gain were noted in these studies, indicating that the antiinflammatory effects of MICs alone are not likely responsible for anti-obesity effects observed by moringa extract treatment. MICs are very effective, however, in blocking glucose production in HII4E hepatocytes, showing activity at nanomolar concentrations (Figure 12A-B) and being close to two orders of magnitude more active than metformin (Figure 12B). Because MICs were able to decrease PEPCK and G6P gene expression at similarly low concentrations, it is tempting to speculate that MICs act via blocking these rate- limiting steps in liver gluconeogenesis. Decreased G6P and PEPCK gene expression was also observed in liver tissue from the moringa extract feeding study, further supporting this mode of action (Figure 12D). In the long term, reduced gluconeogenesis may contribute to improved insulin sensitivity, as metformin's inhibition of gluconeogenesis (31) has been a successful target for treating type 2 diabetes (32), although other studies suggest that metformin may have other modes of action (33-35). Additional symptoms of type 2 diabetes include impaired insulin sensitivity and increased serum levels of insulin, leptin, resistin, TG, and cholesterol (36-39); all of which were reduced by moringa extract treatment.

[00138] Collectively, the results of in vitro and in vivo experiments establish that MICs are the primary biologically active anti-obesity and anti-diabetes constituents of moringa extract, and the primary mechanism of action of the extract is the inhibition of liver gluconeogenesis, which directly or indirectly results in systemically increased insulin sensitivity. These effects are expected, in turn, to reduce lipid accumulation in the liver and body. These conclusions, combined with previous data on MICs anti-inflammatory effects (2), indicate that moringa extract and MICs have beneficial effects for the prevention and treatment of metabolic disorders such as obesity and diabetes.

Documents referenced in Example 7.

1. Mbikay M. Therapeutic potential of Moringa oleifera leaves in chronic hyperglycemia and dyslipidemia: a review. Front Pharmacol. 2012;3: 1-12.

2. Waterman C, Cheng DM, Rojas-Silva P, Poulev A, Dreifus J, Lila MA, et al. Stable, water extractable isothiocyanates from Moringa oleifera leaves attenuate

inflammation in vitro. Phytochem. 2014;103: 114-22.

3. Cheenpracha S, Park E-J, Yoshida WY, Barit C, Wall M, Pezzuto JM, et al. Potential anti-inflammatory phenolic glycosides from the medicinal plant Moringa oleifera fruits. Bioorgan Med Chem. 2010;18(17):6598-602.

4. Bae JY, Lim SS, Kim SJ, Choi JS, Park J, Ju SM, et al. Bog blueberry anthocyanins alleviate photoaging in ultraviolet-B irradiation-induced human dermal fibroblasts. Mol Nutr Food Res. 2009;53(6):726-38.

5. Brunelli D, Tavecchio M, Falcioni C, Frapolli R, Erba E, Iori R, et al. The isothiocyanate produced from glucomoringin inhibits NF-kB and reduces myeloma growth in nude mice in vivo. Biochem Pharmacol. 2010;79(8): 1141-8.

6. Faizi S, Siddiqui BS, Saleem R, Siddiqui S, Aftab K, Gilani AH. Isolation and structure elucidation of new nitrile and mustard oil glycosides from Moringa oleifera and their effect on blood pressure. J Nat Prod. 1994;57(9): 1256-61.

7. Shapiro TA, Fahey JW, Wade KL, Stephenson KK, Talalay P.

Chemoprotective glucosinolates and isothiocyanates of broccoli sprouts metabolism and excretion in humans. Cancer Epidem Biomar. 2001;10(5):501-8.

8. Higdon JV, Delage B, Williams DE, Dashwood RH. Cruciferous vegetables and human cancer risk: epidemiologic evidence and mechanistic basis. Pharmacol Res.

2007;55(3):224-36. 9. Verhoeven DT, Goldbohm RA, van Poppel G, Verhagen H, van den Brandt PA. Epidemiological studies on brassica vegetables and cancer risk. Cancer Epidem Biomar. 1996;5(9):733-48.

10. Traka M, Mithen R. Glucosinolates, isothiocyanates and human health.

Phytochem Rev. 2009;8(l):269-82.

11. Mirmiran P, Bahadoran Z, Hosseinpanah F, Keyzad A, Azizi F. Effects of broccoli sprout with high sulforaphane concentration on inflammatory markers in type 2 diabetic patients: A randomized double-blind placebo-controlled clinical trial. J Funct Foods. 2012;4(4):837-41.

12. Bahadoran Z, Tohidi M, Nazeri P, Mehran M, Azizi F, Mirmiran P. Effect of broccoli sprouts on insulin resistance in type 2 diabetic patients: a randomized double-blind clinical trial. Int J Food Sci Nutr. 2012;63(7):767-71.

13. Bahadoran Z, Mirmiran P, Azizi F. Potential Efficacy of Broccoli Sprouts as a Unique Supplement for Management of Type 2 Diabetes and Its Complications. J Med Food. 2013.

14. Wu H, Liang H, Yuan Q, Wang T, Yan X. Preparation and stability investigation of the inclusion complex of sulforaphane with hydroxypropyl-P-cyclodextrin. Carbohyd Polym. 2010;82(3):613-7.

15. Park E-J, Cheenpracha S, Chang LC, Kondratyuk TP, Pezzuto JM. Inhibition of lipopolysaccharide-induced cyclooxygenase-2 and inducible nitric oxide synthase expression by 4-[(2'-0-acetyl-a-L-rhamnosyloxy)benzyl]isothiocyanate from Moringa oleifera. Nutr Cancer. 2011;63(6):971-82.

16. Shetty P. Public health: India's diabetes time bomb. Nature.

2012;485(7398):S 14-S6.

17. Mbanya JCN, Motala AA, Sobngwi E, Assah FK, Enoru ST. Diabetes in sub- Saharan Africa. Lancet. 2010;375(9733):2254-66.

18. Ng M, Fleming T, Robinson M, Thomson B, Graetz N, Margono C, et al. Global, regional, and national prevalence of overweight and obesity in children and adults during 1980-2013: a systematic analysis for the Global Burden of Disease Study 2013.

Lancet. 2014.doi: 10.1016/S0140-6736(14)60460-8 19. Folch J, Lees M, Sloane-Stanley G. A simple method for the isolation and purification of total lipids from animal tissues. J Biol Chem. 1957;226(l):497-509.

20. Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative CT method. Nature Protocols. 2008;3(6): 1101-8.

21. Cheng DM, Kuhn P, Poulev A, Rojo LE, Lila MA, Raskin I. In vivo and in vitro antidiabetic effects of aqueous cinnamon extract and cinnamon polyphenol-enhanced food matrix. Food Chem. 2012;135(4):2994-3002.

22. Mosmann T. Rapid colorimetric assay for cellular growth and survival:

application to proliferation and cytotoxicity assays. J Immunol Methods. 1983;65(l):55-63.

23. Jaiswal D, Kumar Rai P, Kumar A, Mehta S, Watal G. Effect of Moringa oleifera Lam. leaves aqueous extract therapy on hyperglycemic rats. J Ethnopharmacol. 2009;123(3):392-6.

24. Ndong Moussa UM, Katsumata Shin-ichi , Suzuki Kazuharu Effects of Oral Administration of Moringa oleifera Lam on Glucose Tolerance in Goto-Kakizaki and Wistar Rats. J Clin Biochem Nutr. 2007;40(3):229-33.

25. Miller RS, Becker KG, Prabhu V, Cooke DW. Adipocyte gene expression is altered in formerly obese mice and as a function of diet composition. J Nutr.

2008;138(6): 1033-8.

26. Korda M, Kubant R, Patton S, Malinski T. Leptin-induced endothelial dysfunction in obesity. Am J Physiol-Heart C. 2008;295(4):H1514-H21.

27. Moller DE. Potential role of TNF-alpha in the pathogenesis of insulin resistance and type 2 diabetes. Trends Endocrin Met. 2000;l l(6):212-7.

28. Uysal KT, Wiesbrock, S.M., Marino, M.W., Hotamisligil, G.S. Protection from obesity-induced insulin resistance in mice lacking TNFalpha function. Nature.

1997;389:610-4.

29. Schreyer SA, Chua Jr SC, LeBoeuf RC. Obesity and diabetes in TNF-alpha receptor-deficient mice. J Clinl Invest. 1998;102(2):402.

30. Hotamisligil GS, Murray DL, Choy LN, Spiegelman BM. Tumor necrosis factor alpha inhibits signaling from the insulin receptor. PNAS. 1994;91(l l):4854-8. 31. Hundal RS, Krssak M, Dufour S, Laurent D, Lebon V, Chandramouli V, et al. Mechanism by which metformin reduces glucose production in type 2 diabetes. Diabetes. 2000;49(12):2063-9.

32. Knowler WC, Barrett-Connor E, Fowler SE, Hamman RF, Lachin JM, Walker EA, et al. Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin. New Engl J Med. 2002;346(6):393-403.

33. Rena G, Pearson ER, Sakamoto K. Molecular mechanism of action of metformin: old or new insights? Diabetologia. 2013;56(9): 1898-906.

34. Geerling JJ, Boon MR, van der Zon GC, van den Berg SA, van den Hoek AM, Lombes M, et al. Metformin lowers plasma triglycerides by promoting VLDL-triglyceride clearance by brown adipose tissue in mice. Diabetes. 2013;63(3):880-91.

35. Madiraju AK, Erion DM, Rahimi Y, Zhang X-M, Braddock DT, Albright RA, et al. Metformin suppresses gluconeogenesis by inhibiting mitochondrial glycerophosphate dehydrogenase. Nature. 2014; 510:542-6.

36. Widjaja A, Stratton IM, Horn R, Holman RR, Turner R, Brabant G. UKPDS 20: plasma leptin, obesity, and plasma insulin in type 2 diabetic subjects. J Clin Endocrinol Metab. 1997;82(2):654-7.

37. Steppan CM, Bailey ST, Bhat S, Brown EJ, Banerjee RR, Wright CM, et al. The hormone resistin links obesity to diabetes. Nature. 2001 ;409(6818):307-12.

38. Srinivasan K, Viswanad B, Asrat L, Kaul C, Ramarao P. Combination of high- fat diet-fed and low-dose streptozotocin-treated rat: a model for type 2 diabetes and pharmacological screening. Pharmacol Res. 2005;52(4):313-20.

39. El Messaoudi S, Rongen GA, de Boer RA, Riksen NP. The cardioprotective effects of metformin. Current Opin Lipidol. 2011 ;22(6):445-53.

Example 8 - Preparation of extracts from seeds of a M. oleifera plant

[00139] Moringa seeds were ground to a fine powder and subjected to the extraction protocols listed below in Table 2. Insoluble seed material was separated from the liquid extract by filtration or centrifugation and dried under vacuum to produce final extract. The amounts of recovered moringa isothiocyanates (MICs) were determined using HPLC and MIC-1 as a standard. Table 2.

* Actual concentration are lower than calculated concentrations because of MICs loss and/or degradation during drying.

[00140] The data provided in Table 2 demonstrates that incubating injured (e.g., ground) seeds in a solution comprising water, or injuring seeds in a solution comprising water produces optimal conditions for activating myrosinase and forming MICs. Adding solvent (e.g., ethanol) to the injured seeds before or with a solution comprising water progressively inhibited formation of MICs. No detectable MICs were formed when 95% ethanol was directly added to the injured seeds or seeds that were injured in the presence of 95% ethanol.

[00141] As shown in Table 2, 95% ethanol was most effective in extracting MICs from seeds that were injured in a solution comprising water (or injured seeds that were incubated in a solution comprising water for a period of time before addition of the ethanol), primarily because it allowed most rapid drying of the extract. Differences between calculated and actual concentration of MICs in the extract can be explained by losses that occurred during the drying process. Long drying time of the samples with high water content resulted in significant losses on MIC from the final extract. Much faster drying times for samples with high ethanol concentration resulted in lower loss of MICs.

[00142] In summary, initial incubation of the injured seeds in a solution comprising water prior to extraction with ethanol was sufficient to activate myrosinase and convert most moringa glucosinolates into MICs, as confirmed by the HPLC analysis. MICs formed in this initial process can be effectively extracted in ethanol or other solvents. However, once MICs are formed as a result of adding a solution comprising water to the injured seeds, ethanol can be added to increase the extraction volume and to facilitate extraction efficiency. It is important to note that MIC-1 is the major isothiocyanates formed in moringa seeds and that other isothiocyanates are formed only in trace amounts. The data provided herein

demonstrates that injuring seeds in a solution comprising water (e.g., 1 : 1 to 1 :3 seed to water ratio w/w) for 30-60 minutes followed by extraction with 95% ethanol or other solvent appropriate for solubilizing MICs is an effective way of producing moringa seed extracts enriched in MICs.

[00143] Numerous modifications and variations in the practice of the invention are expected to occur to those of skill in the art upon consideration of the presently preferred embodiments thereof. Consequently, the only limitations which should be placed upon the scope of the invention are those which appear in the appended claims.

[00144] All of the U.S. patents, U.S. patent application publications, U.S. patent applications, foreign patents, foreign patent applications and non-patent publications referred to in this specification and/or listed in the Application Data Sheet, are incorporated herein by reference, in their entireties or in relevant part, as would be apparent from the context of their citation.

[00145] From the foregoing it will be appreciated that, although specific embodiments of the invention have been described herein for purposes of illustration, various modifications may be made without deviating from the spirit and scope of the invention.

References:

Alberti et al., (2009). Circulation, 120(16), 1640-1645. Amaglo et al., (2010). Food Chemistry, 122(4), 1047-1054.

Bao et al., (2011). Biochimica et Biophysica Acta - Reviews on Cancer, 1815(2), 135-

146.

Bennett et al., (2003), Journal of Agricultural and Food Chemistry, 51(12), 3546-

3553.

Bhargava, P., & Lee, C. (2012), Biochemical Journal, 442, 253-262.

Brunelli et al., 2010), Biochemical Pharmacology, 79(8),1141-1148.

Cheenpracha et al., (2010), Bioorganic & Medicinal Chemistry, 18(17), 6598-6602.

Dillard, C. J., & German, J. B. (2000), Journal of the Science of Food and

Agriculture, 80(12), 1744-1756.

Eylen et al., (2008), Journal of Food Engineering, 89(2), 178-186.

Fahey, J. W. (2005), Trees for Life Journal 1, 5.

Ferrante, A. W. (2007), Journal of Internal Medicine, 262(4), 408-414.

Force et al., (2007), Postharvest Biology and Technology, 44(2), 175-178.

Franklin et al., (2013), Drug Development and Industrial Pharmacy, 0, 1-9.

Gallaher et al., (2012), Journal of Agricultural and Food Chemistry, 60(6), 1358-

1362.

Geronikaki, A. A., & Gavalas, A. M. (2006), Combinatorial Chemistry & High Throughput Screening, 9(6), 425-442. Giusti, M. M., & Wrolstad, R. E. (2003), Biochemical Engineering Journal, 14(3), 217-225.

Hobbs et al., (1999), Annual Review of Pharmacology and Toxicology, 39(1), 191-

220.

Hotamisligil et al., (1994), Proceedings of the National Academy of Sciences, 91(11), 4854-4858.

Mariappan et al., (2010), Cardiovascular Research, 85(3), 473-483. Mbikay, M. (2012), Frontiers in Pharmacology, 3, 1-12. Mirza et al., (2012), Cytokine, 57(1), 136-142.

Mocellin, S., & Nitti, D. (2008), Frontiers in Bioscience, 13, 2774-2783. Mosmann, T. (1983), Journal of Immunological Methods, 65(1), 55-63. Moyo et al., (2012), Meat Science, 91(4), 441-447. Pandey et al., (2012), Medicinal & Aromatic Plants: Open Access, 1, 1-8. Park et al., (2011), Nutrition and Cancer, 63(6), 971-982.

Pereira et al., (2002), Journal of Agricultural and Food Chemistry, 50(21), 6239-6244.

Prior et al., (2003), Journal of Agricultural and Food Chemistry, 51(11), 3273-3279.

Siddhuraju, P., & Becker, K. (2003), Journal of Agricultural and Food Chemistry, 51(8), 2144-2155.

Singleton, V., & Rossi, J. A. (1965), American Journal of Enology and Viticulture, 16(3), 144-158.

Song, L., & Thornalley, P. J. (2007), Food and Chemical Toxicology, 45(2), 216-224. Sreelatha, S., & Padma, P. R. (2009), Plant Foods for Human Nutrition, 64(4), 303-

311.

Srivastava, S. K., & Singh, S. V. (2004), Carcinogenesis, 25(9), 1701-1709.

Traka, M., & Mithen, R. (2009). Glucosinolates, isothiocyanates and human health. Phytochemistry Reviews, 8(1), 269-282.

Vongsak et al., (2012), Industrial Crops and Products, 44, 566-571. Wadsworth, T. L., & Koop, D. R. (1999), Biochemical Pharmacology, 57(8), 941-

949.

Wang, Y., & Beydoun, M. A. (2007), Epidemiologic Reviews, 29(1), 6-28.

Wu et al., (2004). Lipophilic and hydrophilic antioxidant capacities of common foods in the United States. Journal of Agricultural and Food Chemistry, 52(12), 4026-4037.

Xu Cet al., (2005), Oncogene 24, 4486-4495.

Xu et al., (2003), The Journal of Clinical Investigation, 112(12), 1821-1830.