Oranges As Medicine: Healing Miracle or Quackery?

Oranges as Medicine

This article on oranges is a chapter from my book Cancer Cured.  If you find it helpful, please support my work and buy my book on Amazon.

Sweet, powerfully refreshing and bursting with flavor, it’s no surprise that oranges are one of the most popular fruits in the world.  Most people are aware that oranges are a potent source of vitamin C, but there are a number of other nutrients within an orange that boost its medicinal value considerably.

In this chapter, we will investigate the therapeutic potential of citrus flavonoids, modified citrus pectin, vitamin C and orange juice on cancer and overall health.

Citrus Flavonoids

Flavonoids are naturally occurring substances found in fruits, vegetables, teas and wines that are responsible for the diversity of colors produced by plants.1  Epidemiological studies have shown that dietary consumption of flavonoids can reduce the risk of cardiovascular disease,3 asthma, type 2 diabetes, heart disease, prostate cancer and lung cancer.4

More than 60 flavonoids have been identified in orange fruit, making them one of the most concentrated and readily available sources of flavonoids.366  A 2003 study from the UK found that a Sicilian variety of orange called Tarocco had higher concentrations of citrus flavonoids than 6 other orange varieties tested.Interestingly, flavonoids are found more abundantly in the peel than in the pulp of citrus fruit.2

Some examples of citrus flavonoids include naringin, naringenin, diosmin, hesperetin, hesperidin, quercetin, tangeretin, nobiletin.  In this section we will investigate three of them.

Citrus Flavonoids Vs. Cancer

One of the methods used by scientists to test the therapeutic value of nutrients or drugs against cancer is to add them to a test tube containing cancer cells and observe the effects.  Testing cancer cells outside of living organisms in this way is called in vitro.  Sometimes effects observed in vitro are not the same as inside living organisms, or in vivo, but many times they are, as you will see.

Naringin

In vitro studies have confirmed naringin can inhibit the growth of stomach,9 cervical,12 breast,14 and colon cancer cells,11 the spread of brain7,8 and bone cancer cells,10 and trigger cancer cell death (apoptosis) in colon,11,17 pancreatic,17 stomach,17 cervical,12,13,17 liver17 and breast cancer cells.14,17

Another popular method used by scientists to determine the value of nutrients or drugs against cancer is inducing cancer in animals – usually by injecting carcinogenic chemicals into them – and then administering the treatment and observing the outcome.

In vivo studies have confirmed naringin can inhibit the growth of colon,18 oral,19 lung20 and connective tissue tumors,16,17 the spread of skin tumors20and trigger apoptosis in colon tumors.18

A Closer Look…

  • In 2012, scientists from Sao Paulo, Brazil, investigated the effects of naringin on rats bearing connective tissue cancer. Results showed that a 25mg/kg dose of naringin administered daily for 50 days inhibited tumor growth by 75%.  Naringin also increased survival and notably, “two rats presented complete tumor regression.”16

One of the most exciting experiments on naringin was conducted in 2016 by Chinese researchers, who added naringin to skin cancer cells to determine its impact on cellular energy metabolism.  One of the key changes in the metabolism of cancer cells is increased production of lactic acid, and not only did naringin inhibit lactic acid production in the skin cancer cells, but it also completely reversed them back into normal cells.  “In summary, we demonstrated that naringin inhibits the malignant phenotype of A375 cells.” they concluded.15

Perhaps best of all, the beneficial effects of naringin can be obtained “without adverse side effects.” 59

Normally, once a ‘drug’ has proven itself in vitro and in vivo, it would move on to human testing in clinical trials, but since the average cost of phase 1 clinical trial in the United States ranges from $1.4 million to $6.6 million dollars;198 and since natural substances found in foods can’t be patented or sold by drug companies, this type of research doesn’t often receive funding and is thus rarely performed.

Many times, the only reason in vitro and in vivo studies on food nutrients are funded is so drug companies can find medicines that work and then attempt to replicate similar chemicals to patent and sell.

Naringenin

A 2015 study identified naringenin as one of 10 therapeutic agents “that may warrant further investigation to target the tumor microenvironment” for preventing and treating cancer.”64

In vitro experiments have established naringenin can inhibit the growth of breast,65,73 stomach,71 colon,66,73 skin,68,76 leukemia75 and liver cancer cells,67,77 the spread of breast,65 liver,67 skin,68 bladder,69 pancreatic,70 and stomach cancer cells,71 and trigger apoptosis in liver,67,77 stomach,71 colon,72,73 breast,73,74 leukemia75 and skin cancer cells.76

Animal experiments have verified naringenin can inhibit the growth of oral,19 stomach,78 lung81 and brain tumors,80 prevent the spread of breast tumors79 and trigger apoptosis in brain tumors.82

Synergistic enhancement of the anti-cancer effects of naringenin can be obtained by combining it with either curcumin83 or vitamin E,84 and one study reports that nano-encapsulated naringenin exhibits “significantly higher” antioxidant and anticancer properties than naringenin in free form.86

Naringenin has a “promising safety profile”133 and remarkably, it maintains its cancer-killing effects even in the presence of the environmental toxin bisphenol A.85

Hesperidin

In vitro, hesperidin can inhibit the growth of breast,134,135 immune,138 leukemia,140,141 and lung cancer cells,143  the spread of skin cancer cells,136 and trigger apoptosis in breast,135,144 immune,138 colon,139 leukemia,140,141 liver137,142 and lung cancer cells.143

Animal studies have confirmed hesperidin can inhibit the growth of colon,145 lung,146 bladder,147 oral,148,149 throat150 and stomach tumors,151 and trigger apoptosis in stomach151 and colon tumors.152

One study compared the medicinal potencies of a number of citrus flavonoids and found that hesperidin exerted a more powerful anti-cancer effect than neohesperidin, naringin and naringenin.137  With that in mind, Tunisian researchers reported in 2016 that concentrations of hesperidin were greater in organically-grown oranges than in oranges grown conventionally.6

A safety study from 1990 fed rats dietary concentrations of 0%, 1.25% or 5% methyl hesperidin for two years and concluded that the substance “lacked any carcinogenicity” in rats.193

Additional Health Effects

Antibacterial:

  • Naringin exerts a “robust antibacterial effect”21
  • Hesperidin inhibits growth of bifidobacteria153

Antioxidant:

  • Naringin neutralizes the toxic effects of herbicide paraquat22
  • Naringin prevents kidney and liver damage from acetaminophen23 and sodium arsenite24
  • Naringin prevents chemotherapy-induced kidney25 and lung damage26
  • Naringin reverses side effects of HIV medication29,30
  • Naringenin prevents damage from lead108 and endotoxin87
  • Hesperidin prevents chemically-induced kidney damage155-158
  • Hesperidin prevents chemotherapy-induced liver damage161

Antiviral:

  • Naringin28 and naringenin93 inhibit infection by sindbis virus28
  • Naringenin reduces hepatitis C virus secretions from infected cells by 80%92
  • Hesperidin prevents replication of influenza A virus162
  • Hesperidin inhibits infection by canine distemper virus163 and rotavirus164
  • Hesperidin inhibits combined viral-bacterial infections165

Arthritis:

  • Naringin prevents inflammation associated with arthritis31-34
  • Naringenin prevents inflammatory pain in mice94,95
  • Hesperidin prevents chemically-induced arthritis154,166,167

Asthma:

  • Naringin significantly reduces coughing (associated with a type of asthma that causes chronic coughing)63

Bone Health:

  • Naringin prevents the destruction of cartilage38
  • Naringin,35,36 naringenin96-99 and hesperidin168-170 prevent bone loss and accelerate bone formation

Brain Health:

  • Naringin reverses chemically-induced memory deficits39-41,47
  • Naringin improves brain function in mice with Alzheimer’s disease42,43
  • Naringin prevents brain degeneration in rats with Parkinson’s disease44-46
  • Naringin prevents chemically-induced seizures48
  • Naringenin reduces anxiety caused by lead poisoning89
  • Naringenin prevents brain damage caused by neurotoxins90 and iron102
  • Naringenin prevents Alzheimer’s disease100
  • Naringenin improves learning and memory in rats with Alzheimer’s disease101
  • Naringenin prevents cognitive decline in rats with Parkinson’s disease103
  • Hesperidin prevents brain damage from pesticides,171 heavy metals,172 and other poisons174
  • Hesperidin prevents cognitive impairment in mice with Alzheimer’s disease173
  • Hesperidin prevents brain damage caused by ionizing radiation188

Cooking:

  • Naringenin prevents toxic acrylamides from forming in food during high-heat cooking132

Dental Health:

  • Naringin remineralizes root caries (cavities) in teeth37

Depression:

  • Naringenin produces antidepressant-like behavior in rats106,107
  • Naringenin105 and hesperidin175-177 exert potent antidepressant effects

Detoxification:

  • Naringenin chelates lead from the body108

Diabetes:

  • Naringin prevents scarring of the heart caused by diabetes49
  • Naringenin prevents kidney damage caused by diabetes52,109
  • Naringenin improves glucose metabolism110,111
  • Naringin improves insulin sensitivity50,51
  • Hesperidin reduces diabetes and its complications178

Digestive Health:

  • Naringenin an effective treatment for inflammatory bowel disease120,121
  • Naringenin prevents defects of the intestinal barrier122

Exercise:

  • Naringin in combination with treadmill exercise is more effective at increasing bone strength and density than either therapy alone53
  • Hesperidin synergistically enhances the health benefits of exercise179

Eye Health:

  • Naringenin eye drops prevent chemically-induced retinal damage91
  • Hesperidin prevents eye damage caused by chemotherapy159

Food Production:

  • Naringin and Hesperidin fed to chickens elevates antioxidant levels in chicken meat; are “important additives for both the consumer and the industry.”27

Headaches:

  • Drynaria quercifolia (a plant containing naringin) alleviates painful inflammatory conditions, like headache54
  • Hesperidin may be useful for treating migraines180

Healing:

  • Naringin55 and hesperidin191 accelerate wound healing

Heart Health:

  • Naringin56 and naringenin113 prevent arterial plaque formation
  • Naringinen reduces arterial stiffness112
  • Naringenin prevents thickening of the heart muscle114

Immune System:

  • Naringenin boosts the immune system115,116
  • Naringenin significantly enhances anti-cancer immunity117-119
  • Hesperidin enhances immune systems of mice181 and irradiated mice189
  • Hesperidin enhances immune systems of broiler chickens182

Obesity:

  • Naringin significantly decreases fat mass57,59
  • Naringin prevents formation of new fat tissue58
  • Naringenin reduces body fat and suppresses weight gain123-125
  • Hesperidin improves lipid metabolism in humans183,184

Radiation protective:

  • Naringin protects skin from ultraviolet B radiation61
  • Naringin60 and naringenin126 prevent genetic damage caused by ionizing radiation
  • Naringenin prevents skin aging and wrinkle formation caused by ultraviolet B radiation127-129
  • Naringenin added to conventional sunscreen reduces its toxicity130
  • Hesperidin reduces damage caused by whole-body gamma ray irradiation185
  • Hesperidin prevents ultraviolet B radiation damage186,187

Sexual Health:

  • Naringenin aids the process of conception104
  • Naringenin prevents testicle damage caused by insecticides88
  • Hesperidin prevents testicle and sperm damage caused by chemotherapy160

Skin Health:

  • Naringin62 and hesperidin192 promote the production of skin-protective melanin
  • Naringenin “should be introduced into cosmetic products as natural tanning agents.”131

Other:

  • A mixture of citrus flavonoids Hesperidin, Troxerutin, and Diosmin applied to hemmorhoids reduces pain, bleeding and swelling in humans190

Modified Citrus Pectin

Pectin is a complex of sugar molecules (polysaccharide) heavily concentrated in the pulp and peel of citrus fruit.  Because of its gelling properties, pectin is a traditional ingredient used for making marmalades and jams.

Pectin’s long-branched chains of polysaccharides make it virtually indigestible by humans, but researchers have discovered ways to modify pectin so it can be easily absorbed into the blood stream.  Once in the bloodstream, modified citrus pectin (MCP) has proven useful for treating a number of conditions, including cancer.317  Although heat treatment and pH modifications are commonly used to create modified citrus pectin,315 high-intensity ultrasound is also effective and is said to be more ‘environmentally friendly.’316

“The more we learn about MCP, the more impressive it becomes,” said Dr. Isaac Eliaz. “With its ability to control aggressive cancers, reduce inflammation, enhance immunity, chelate heavy metals and work synergistically with a variety of chemotherapeutic agents, it has earned an important role within anti-cancer and chronic disease protocols.”367

Modified Citrus Pectin vs. Cancer

Scientists have observed modified citrus pectin prevent the growth of prostate cancer cells320,322 and trigger apoptosis in lung,321 liver,321 prostate322 and eight other types of cancer cells.323

Co-administration of modified citrus pectin with two herbal products have revealed synergistic inhibitory effects on the spread of breast and prostate cancer in vitro.324 Modified citrus pectin can also eliminate chemotherapy resistance325 and increase the apoptosis-inducing effects of chemotherapy in cell cultures.326

Modified citrus pectin can prevent the growth of skin327 and sarcoma tumors in mice; including a 51% reduction in tumor size and increased survival compared to control mice.323  Another study reported a 70% reduction in colon tumor size in mice after 20 days of MCP treatment.332  Also in animals, the spread of breast,328 prostate,329 skin330 and colon tumors can be inhibited by as much as 90% using modified citrus pectin.331

Scientists from the Harry S. Truman Memorial Veteran’s Hospital in Missouri discovered that MCP prevents cancer metastasis by inhibiting circulating tumor cells from adhering and establishing themselves onto distal body tissues.318  Another mechanism behind MCP’s therapeutic effects is the inhibition of a substance called Galectin-3, which is involved in inflammation, fibrosis, heart disease, stroke and cancer.319

Studies report modified citrus pectin has no adverse side effects,339 including one study in which 15 grams MCP was administered daily to humans for 12 consecutive months.340

Additional Health Effects

Antioxidant:

  • MCP prevents liver fibrosis334
  • MCP prevents kidney injury335
  • MCP prevents damage caused by endotoxin336

Arthritis:

  • MCP is “a therapeutic approach for the treatment of inflammatory arthritis” 337

Detoxification:

  • MCP dramatically increases urinary excretion of arsenic, cadmium and lead in humans338
  • MCP safely and dramatically detoxifies lead from children339
  • MCP reduces toxic heavy metal burden in humans by an average of 74%340

Heart Health:

  • MCP “may represent a new promising therapeutic option in heart failure”341
  • MCP decreases cardiovascular fibrosis and inflammation342,343,344
  • Galectin-3 inhibition “causes decreased atherosclerosis”345

Immune System:

  • MCP enhances anti-cancer immunity346

Inflammation:

  • MCP reduces inflammation and pain after spinal nerve injury333

Obesity:

  • MCP prevents production of new fat tissue347

Vitamin C

Although vitamin C (aka ascorbic acid or ascorbate) wasn’t officially discovered until 1928 by Hungarian biochemist Albert Szent-Gyorgyi,194 the manifestations of its deficiency, known as scurvy,195 were first documented by the physician Hippocrates in ancient Greece (460BC-370BC).368  In 1945, scientists from the University of Wisconsin found that when they deprived monkeys of vitamin C for just a few weeks, various dental issues including bleeding gums, loosening of the teeth and the formation of heavy tartar deposits were induced.196

The effects of supplemental vitamin C are highly-dependent on the dose administered.  In low doses, vitamin C behaves as an anti-oxidant, helping the body neutralize toxins and eliminate waste products.  And in high doses, vitamin C acts as a pro-oxidant that can selectively target unhealthy and even cancerous cells.197 High-doses of intravenous vitamin C have been used to treat cancer since the 1970s.200

Vitamin C vs. Cancer

One thing cancer patients all have in common is significantly depleted levels of vitamin C.222  Remarkably, some researchers have said that vitamin C might be the most important nutritional factor needed by the body to resist cancer; “There is increasing recognition that resistance to cancer depends, to a certain extent, upon the availability of certain nutritional factors, of which ascorbic acid appears to be the most important,” wrote scientist Ewan Cameron in 1982.201

An epidemiological study of people in Northern Italy reported that vitamin C intake has “possible protective activity” against skin cancer202 and greater consumption of antioxidants was associated with less aggressive prostate cancer in the United States.203  A 2014 systematic review by Chinese researchers concluded that low doses of vitamins, specifically vitamins A, C and E, can significantly reduce the risk of stomach cancer.204

In vitro studies have confirmed vitamin C can trigger apoptosis in colon,206,207,214 breast,207 skin,208 blood,209 bone marrow,209 Ehrlich acites carcinoma,205 melanoma220 and four types of malignant mesothelioma cancer cells.213

Co-administration of vitamin C and vitamin B2 can synergistically enhance apoptosis in multiple types of cancer cells.210  Vitamin C loaded into tiny bubbles of fat (lipid nanoparticles) was shown to enter into cells more efficiently than free vitamin C219 and vitamin C affixed to nano-sized polymer carriers has been shown to trigger apoptosis in brain cancer cells.218

In animals, vitamin C can prevent the growth of lung,211 skin,211 ovarian,212 pancreatic,212 brain,212 malignant mesothelioma,213 colon214 and sarcoma tumors,215 and can trigger apoptosis in liver tumors.216  A nutrient mixture containing lysine, vitamin C, proline, green tea extract and other micronutrients fed to tumor-bearing mice “demonstrated a potent inhibition of [cervical] tumor growth.”217

A Closer Look…

  • An American group of scientists from Kansas administered 500mg/kg/day sodium ascorbate to liver tumor-bearing guinea pigs in 2006. Results showed that “Subcutaneous injections of ascorbate (500 mg/kg/day) inhibited tumor growth by as much as sixty-five percent, with oral supplementation reducing it by roughly fifty percent.” 216

In 1994, researchers from the Oregon Institute of Science and Medicine induced tumors in mice and treated them with high-doses of vitamin C along with variations in diet.  Results showed that survival could be increased by up to 20-times simply by adjusting the animal’s nutritional intake.221

Two-time Nobel Prize winner Linus Pauling and surgeon Ewan Cameron administered 10 grams/day of vitamin C to terminal cancer patients in 1976 and found that survival was increased “more than 4.2 times” compared to patients who weren’t given vitamin C.222  Other studies have confirmed vitamin C can increase survival and significantly improve the quality of life of terminal cancer patients.223-225

How does vitamin C exert its beneficial effects?  When blood levels of vitamin C are maintained at a consistently high level, it is absorbed into cancerous tissue where it produces hydrogen peroxide that kills cancer cells.199

Safety

Thanks to the work of Dr. Frederick R. Klenner, it has been known for over 70 years that vitamin C doses as high as 300,000mg (300 grams) per day in humans are safe and modern research has confirmed this finding.205,209,212,224,225,235,260

A Korean study from 2007 acknowledged that vitamin C “is considered a safe and effective therapy”225 and a 2008 study from the National Institutes of Health found that high-doses of vitamin C displayed “cytotoxicity toward a variety of cancer cells in vitro without adversely affecting normal cells.”212

One thing to be aware of is that vitamin C can increase the absorption of iron,307-309 which in small amounts is essential, but like all heavy metals, becomes toxic in excess311 and can diminish the effectiveness of vitamin C treatment.310  For these reasons, oral supplementation of vitamin C is probably best without food and to lower existing iron levels in the body, iron-chelating substances such as tetracycline, doxycycline, minocycline312 or curcumin369 can be used.

Oral vs. Intravenous

There is some controversy surrounding the efficacy of various vitamin C administration methods.  While Dr. Mark Levine of the NIH claims that “…only injected ascorbate might deliver the concentrations needed to see an anti-tumor effect,”313 Dr. Steve Hickey has said, “it is not clear that intravenous vitamin C necessarily provides an advantage over oral supplements in the treatment of cancer.”314

When the body is given a high enough dose of vitamin C intravenously, much of it goes unused and is excreted in the urine, according to Dr. Hickey.  Furthermore, he makes the case that high-doses of orally administered vitamin C might even be more effective.314

While Dr. Levine claims that maximum blood levels of vitamin C are 200μM/L, Dr. Hickey maintains he has been able to generate blood levels of around 250μM/L with a single 5 gram oral dose of vitamin C and blood levels above 400μM/L with a single oral dose of liposomal vitamin C.314

Dose

For many people, a single oral dose of 2 grams of vitamin C will cause a laxative effect and anything more will be eliminated from the body.  Interestingly, bowel tolerance is said to increase dramatically when a person is ill.314  In other words, a person who would normally be able to tolerate only 2 grams might be able to tolerate 100-times that amount when sick.

Maximum blood levels of orally-ingested vitamin C can be achieved by consuming about 3 grams every four hours.  Since vitamin C is only active in the body for a few hours, frequent doses are critical to maintain consistently high blood levels.

Success Stories

Dr. Victor Marcial, radiation oncologist:

“We studied patients with advanced cancer (stage 4). 40 patients received 40,000-75,000 mg intravenously several times a week. These are patients that have not responded to other treatments. The initial tumor response rate was achieved in 75% of patients, defined as a 50% reduction or more in tumor size… Once you start using IV vitamin C, the effect is so dramatic that it is difficult to go back to not using it.”

Linus Pauling, 2x Nobel Prize Laureate:

“I became interested in vitamin C and cancer in 1971 and began working with Ewan Cameron, M.B., Ch.B., chief surgeon at Vale of Leven Hospital in Scotland.  Cameron gave 10 grams of vitamin C a day to patients with untreatable, terminal cancer.  These patients were then compared by Cameron and me to patients with the same kind of cancer at the same terminal stage who were being treated in the same hospital but by other doctors-doctors who didn’t give vitamin C, but instead just gave conventional treatments.  Cameron’s terminal cancer patients lived far longer compared to the ones who didn’t get 10 grams a day of vitamin C. The other patients lived an average of six months after they were pronounced terminal, while Cameron’s patients lived an average of about six years.”

Dr. Irwin Stone, American biochemist, chemical engineer:

“In one case where complete remission was achieved in myelogenous leukemia… the patient took 24-42 gms vitamin c per day… it is inconceivable that no-one appears to have followed this up… without the scurvy, leukemia may be a relatively benign, non-fatal condition.  I wrote a paper… in an attempt to have the therapy clinically tested… I sent it to 3 cancer journals and 3 blood journals… it was refused by all… Two without even reading it.”

Additional Health Effects

Antioxidant:

  • Vitamin C an antidote for snake venom228
  • Vitamin C a cure for carbon monoxide poisoning233
  • Vitamin C a cure for mushroom poisoning229
  • Vitamin C prevents damage caused by agricultural fungicides230 and insecticides231
  • Vitamin C prevents liver damage caused by dexmedetomidine236
  • Vitamin C prevents damage caused by monosodium glutamate237
  • Vitamin C prevents damage caused by methylmercury,238 formaldehyde239 and endotoxin241,242,245,246,248
  • Vitamin C prevents chemically-induced ulcer formation in rats240
  • Vitamin C prevents septic organ injury in mice243
  • Vitamin C prevents alcohol-induced liver fibrosis in mice244
  • Vitamin C prevents the formation of nitric oxide247
  • Vitamin C prevents kidney249 and liver damage250,251 caused by chemotherapy

Antiviral:

  • Vitamin C “kills influenza virus” 252
  • Vitamin C shortens duration of the common cold, pneumonia, malaria and diarrhea infections253
  • Vitamin C suppresses HIV replication by infected cells254
  • Vitamin C successfully treats hepatitis, mononucleosis and pneumonia255
  • Vitamin C successfully treats polio, diphtheria, herpes zoster, herpes simplex, chicken pox, influenza, measles, mumps and viral pneumonia256
  • Vitamin C successfully treats Epstein-Barr virus257
  • Vitamin C resolves all symptoms of Chikungunya fever in two days260

Arthritis:

  • Elevated free radicals and oxidative stress found in patients with Rheumatoid Arthritis227

Bone Health:

  • Vitamin C prevents bone loss261
  • Vitamin C improves bone mineral density in postmenopausal women262-264
  • Vitamin C is “a skeletal anabolic agent” 265
  • Vitamin A, C and E decrease risk of hip fracture266

Brain Health:

  • Vitamin C prevents brain damage caused by methamphetamine,267 insecticides268,269 and glutamate270
  • Vitamin C deficiency increases risk of seizures271
  • Vitamin C (and zinc) deficiencies impair the physical and mental growth of children287
  • Vitamin C levels in patients with severe Parkinson’s disease were “significantly lower” 272

Depression:

  • Vitamin C reduces anxiety levels in highschool students after 14 days of supplementing 500mg276

Detoxification:

  • Vitamin C chelates lead from the bloodstream277
  • Vitamin C reduces blood levels of chemical pollutants232

Diabetes:

  • Vitamin C reduces fasting blood sugar in diabetics278,280
  • Vitamin C significantly lowers needed insulin dose for blood sugar control279

Exercise:

  • Vitamin C and low-intensity exercise prevent seizures281
  • Vitamin C has anti-seizure effects in rats performing endurance swimming282
  • Vitamin C improves blood flow and oxygen use in muscles283

Food Production:

  • Vitamin C improves growth performance and enhances stress resistance in fish303

Headaches:

  • Vitamin C and Pinus Radiata bark extract ingested for 12 months reduces headache severity and frequency by 50%284

Healing:

  • Vitamin C accelerates healing235,301
  • Vitamin C enhances cell survival and DNA repair in human fibroblasts exposed to x-rays299

Heart Health:

  • Vitamin C decreases length of hospital stay in patients following cardiac surgery304

Immune System:

  • Vitamin C enhances anti-cancer immunity285,289-292
  • Vitamin C significantly enhances immunity286-288

Inflammation:

  • Vitamin C reduces inflammation226

Lifespan:

  • Antioxidant-rich diet extends survival of mice exposed to endotoxin234
  • Vitamin C extends lifespan of tetanus patients258,259

Obesity:

  • Vitamin C intake reduces obesity in women293

Radiation protective:

  • Vitamin C prevents damage caused by ionizing radiation294,297,298
  • Vitamin C and vitamin E synergistically prevent damage caused by ionizing radiation295
  • Vitamin C and melatonin synergistically prevent damage caused by ionizing radiation296

Sexual Health:

  • Vitamin C “significantly improves sperm concentration and mobility” 274
  • Vitamin C prevents infertility in rats subjected to forced swimming stress275
  • Vitamin C promotes a healthy pregnancy273

Sleep:

  • Vitamin C prevents spatial memory impairment in rats following sleep deprivation300

Other:

  • Vitamin C resolves symptoms of burning mouth syndrome in humans302
  • Vitamin C diminishes microparticle elevations caused by SCUBA diving305
  • Vitamin C prevents complex regional pain syndrome in humans306

Orange Juice

If an orange contains all the medicines we’ve investigated above, then one would expect orange juice to also have substantial therapeutic value.

Orange Juice vs Cancer

Although studies are limited, scientists have investigated the therapeutic effects of orange juice on cancer cell cultures and in animals.  In vitro studies have confirmed orange juice can trigger apoptosis in two types of blood cancer cells.348

In vivo, researchers have experimentally induced tumors in rats and then replaced their water with orange juice to determine its effects.  Results show that orange juice can inhibit the growth of breast,349 colon,350-352 tongue352 and lung tumors,352  and can trigger apoptosis in colon tumors.350

A Closer Look…

  • In 2015, Brazilian researchers incubated two types of blood cancer cells with orange juice – one with the juice of a red-fleshed sweet orange and the other with juice from a blond orange – for 24-hours and observed the results. At the end of the study, both varieties of orange juice were found to induce apoptosis in the blood cancer cells.348
  • Mandarin orange juice was tested on rats induced with three types of cancers in a 2012 Japanese study from the Journal of Biomedicine & Biotechnology. Citrus juices from the satsuma mandarin orange were found to inhibit the formation of chemically-induced colon, tongue and lung tumors in rats.352
  • Canadian scientists from Western University in London, Ontario, chemically-induced breast tumors in rats and fed them orange juice to determine if it could prevent cancer formation. Published in the journal Nutrition and Cancer in 1996, results showed that rats given orange juice “had a smaller tumor burden than controls.” 349

Additional Health Effects

Antibacterial:

  • Tangerine juice concentrate is effective for “controlling unwanted microbial growth” 353

Antioxidant:

  • Orange juice consumption results in a “marked antioxidant effect”355,356

Bone Health:

  • Orange juice increases bone strength in rats358
  • Orange juice increases bone mineral density in children and adults357

Exercise:

  • Orange juice prevents exercise-induced hypoxia359
  • Orange juice improves physical performance in overweight women360

Heart Health:

  • Fermented orange juice reduces cardiovascular risk factors361

Immune System:

  • Orange juice enhances the immune system362

Inflammation:

  • Orange juice reduces inflammation354

Obesity:

  • Orange juice associated with healthier body composition in adults363
  • Orange juice decreases risk of obesity364
  • Orange juice prevents fatty liver disease365

References

  1. Patel K, Singh GK, Patel DK. A review on pharmacological and analytical aspects of naringenin. Chin J Integr Med. 2014.
    https://link.springer.com/article/10.1007/s11655-014-1960-x
  2. Nakajima VM, Madeira JV, Macedo GA, Macedo JA. Biotransformation effects on anti lipogenic activity of citrus extracts. Food Chem. 2016;197 Pt B:1046-53.
    https://www.sciencedirect.com/science/article/pii/S0308814615302545
  3. Chanet A, Milenkovic D, Manach C, Mazur A, Morand C. Citrus flavanones: what is their role in cardiovascular protection?. J Agric Food Chem. 2012;60(36):8809-22.
    http://pubs.acs.org/doi/abs/10.1021/jf300669s
  4. Knekt P, Kumpulainen J, Järvinen R, et al. Flavonoid intake and risk of chronic diseases. Am J Clin Nutr. 2002;76(3):560-8.
    http://ajcn.nutrition.org/content/76/3/560.long
  5. Proteggente AR, Saija A, De pasquale A, Rice-evans CA. The compositional characterisation and antioxidant activity of fresh juices from sicilian sweet orange (Citrus sinensis L. Osbeck) varieties. Free Radic Res. 2003;37(6):681-7.
    https://www.ncbi.nlm.nih.gov/pubmed/12868495
  6. Letaief H, Zemni H, Mliki A, Chebil S. Composition of Citrus sinensis (L.) Osbeck cv «Maltaise demi-sanguine» juice. A comparison between organic and conventional farming. Food Chem. 2016;194:290-5.
    https://www.researchgate.net/publication/281512187_Composition_of_Citrus_sinensis_L_Osbeck_cv_Maltaise_demi-sanguine_juice_A_comparison_between_organic_and_conventional_farming
  7. Aroui S, Aouey B, Chtourou Y, Meunier AC, Fetoui H, Kenani A. Naringin suppresses cell metastasis and the expression of matrix metalloproteinases (MMP-2 and MMP-9) via the inhibition of ERK-P38-JNK signaling pathway in human glioblastoma. Chem Biol Interact. 2016;244:195-203.
    https://www.sciencedirect.com/science/article/pii/S0009279715301460
  8. Aroui S, Najlaoui F, Chtourou Y, et al. Naringin inhibits the invasion and migration of human glioblastoma cell via downregulation of MMP-2 and MMP-9 expression and inactivation of p38 signaling pathway. Tumour Biol. 2016;37(3):3831-9.
    https://hal-riip.archives-ouvertes.fr/pasteur-01374978
  9. Raha S, Yumnam S, Hong GE, et al. Naringin induces autophagy-mediated growth inhibition by downregulating the PI3K/Akt/mTOR cascade via activation of MAPK pathways in AGS cancer cells. Int J Oncol. 2015;47(3):1061-9.
    https://www.ncbi.nlm.nih.gov/pubmed/26201693
  10. Tan TW, Chou YE, Yang WH, Hsu CJ, Fong YC, Tang CH. Naringin suppress chondrosarcoma migration through inhibition vascular adhesion molecule-1 expression by modulating miR-126. Int Immunopharmacol. 2014;22(1):107-14.
    https://www.sciencedirect.com/science/article/pii/S1567576914002434
  11. Vadde R, Radhakrishnan S, Reddivari L, Vanamala JK. Triphala Extract Suppresses Proliferation and Induces Apoptosis in Human Colon Cancer Stem Cells via Suppressing c-Myc/Cyclin D1 and Elevation of Bax/Bcl-2 Ratio. Biomed Res Int. 2015;2015:649263.
    https://www.hindawi.com/journals/bmri/2015/649263/
  12. Zeng L, Zhen Y, Chen Y, et al. Naringin inhibits growth and induces apoptosis by a mechanism dependent on reduced activation of NF‑κB/COX‑2‑caspase-1 pathway in HeLa cervical cancer cells. Int J Oncol. 2014;45(5):1929-36.
    https://www.medscape.com/medline/abstract/25174821
  13. Ramesh E, Alshatwi AA. Naringin induces death receptor and mitochondria-mediated apoptosis in human cervical cancer (SiHa) cells. Food Chem Toxicol. 2013;51:97-105.
    https://www.sciencedirect.com/science/article/pii/S027869151200511X
  14. Li H, Yang B, Huang J, et al. Naringin inhibits growth potential of human triple-negative breast cancer cells by targeting β-catenin signaling pathway. Toxicol Lett. 2013;220(3):219-28.
    https://www.sciencedirect.com/science/article/pii/S0378427413002063
  15. Guo B, Zhang Y, Hui Q, Wang H, Tao K. Naringin suppresses the metabolism of A375 cells by inhibiting the phosphorylation of c-Src. Tumour Biol. 2016;37(3):3841-50.
    https://link.springer.com/article/10.1007/s13277-015-4235-z
  16. Camargo CA, Gomes-marcondes MC, Wutzki NC, Aoyama H. Naringin inhibits tumor growth and reduces interleukin-6 and tumor necrosis factor α levels in rats with Walker 256 carcinosarcoma. Anticancer Res. 2012;32(1):129-33.
    http://ar.iiarjournals.org/content/32/1/129.figures-only
  17. Kanno S, Tomizawa A, Hiura T, et al. Inhibitory effects of naringenin on tumor growth in human cancer cell lines and sarcoma S-180-implanted mice. Biol Pharm Bull. 2005;28(3):527-30.
    https://www.jstage.jst.go.jp/article/bpb/28/3/28_3_527/_article
  18. Zhang YS, Li Y, Wang Y, et al. Naringin, a natural dietary compound, prevents intestinal tumorigenesis in Apc (Min/+) mouse model. J Cancer Res Clin Oncol. 2016;142(5):913-25.
    https://rd.springer.com/article/10.1007/s00432-015-2097-9
  19. Miller EG, Peacock JJ, Bourland TC, et al. Inhibition of oral carcinogenesis by citrus flavonoids. Nutr Cancer. 2008;60(1):69-74.
    https://www.ncbi.nlm.nih.gov/pubmed/18444138
  20. Menon LG, Kuttan R, Kuttan G. Inhibition of lung metastasis in mice induced by B16F10 melanoma cells by polyphenolic compounds. Cancer Lett. 1995;95(1-2):221-5.
    https://www.sciencedirect.com/science/article/pii/0304383595038873
  21. Ozçelik B, Kartal M, Orhan I. Cytotoxicity, antiviral and antimicrobial activities of alkaloids, flavonoids, and phenolic acids. Pharm Biol. 2011;49(4):396-402.
    http://www.tandfonline.com/doi/full/10.3109/13880209.2010.519390
  22. Blanco-ayala T, Andérica-romero AC, Pedraza-chaverri J. New insights into antioxidant strategies against paraquat toxicity. Free Radic Res. 2014;48(6):623-40.
    http://www.tandfonline.com/doi/abs/10.3109/10715762.2014.899694?journalCode=ifra20
  23. Adil M, Kandhare AD, Ghosh P, Venkata S, Raygude KS, Bodhankar SL. Ameliorative effect of naringin in acetaminophen-induced hepatic and renal toxicity in laboratory rats: role of FXR and KIM-1. Ren Fail. 2016;:1-14.
    http://www.tandfonline.com/doi/full/10.3109/0886022X.2016.1163998
  24. Adil M, Kandhare AD, Visnagri A, Bodhankar SL. Naringin ameliorates sodium arsenite-induced renal and hepatic toxicity in rats: decisive role of KIM-1, Caspase-3, TGF-β, and TNF-α. Ren Fail. 2015;37(8):1396-407.
    http://www.tandfonline.com/doi/figure/10.3109/0886022X.2015.1074462
  25. Chtourou Y, Aouey B, Aroui S, Kebieche M, Fetoui H. Anti-apoptotic and anti-inflammatory effects of naringin on cisplatin-induced renal injury in the rat. Chem Biol Interact. 2016;243:1-9.
    https://www.sciencedirect.com/science/article/pii/S0009279715301216
  26. Turgut NH, Kara H, Elagoz S, Deveci K, Gungor H, Arslanbas E. The Protective Effect of Naringin against Bleomycin-Induced Pulmonary Fibrosis in Wistar Rats. Pulm Med. 2016;2016:7601393.
    http://europepmc.org/abstract/MED/26977316
  27. Goliomytis M, Kartsonas N, Charismiadou MA, Symeon GK, Simitzis PE, Deligeorgis SG. The Influence of Naringin or Hesperidin Dietary Supplementation on Broiler Meat Quality and Oxidative Stability. PLoS ONE. 2015;10(10):e0141652.
    http://journals.plos.org/plosone/article?id=10.1371/journal.pone.0141652
  28. Paredes A, Alzuru M, Mendez J, Rodríguez-ortega M. Anti-Sindbis activity of flavanones hesperetin and naringenin. Biol Pharm Bull. 2003;26(1):108-9.
    https://www.ncbi.nlm.nih.gov/pubmed/12520185
  29. Adebiyi OO, Adebiyi OA, Owira PM. Naringin Reverses Hepatocyte Apoptosis and Oxidative Stress Associated with HIV-1 Nucleotide Reverse Transcriptase Inhibitors-Induced Metabolic Complications. Nutrients. 2015;7(12):10352-68.
    http://www.mdpi.com/2072-6643/7/12/5540
  30. Adebiyi OO, Adebiyi OA, Owira P. Naringin improves zidovudine- and stavudine-induced skeletal muscle complications in rats. Hum Exp Toxicol. 2016.
    http://journals.sagepub.com/doi/abs/10.1177/0960327116638726
  31. Yin FM, Xiao LB, Zhang Y. [Research progress on Drynaria fortunei naringin on inflammation and bone activity]. Zhongguo Gu Shang. 2015;28(2):182-6.
    https://www.ncbi.nlm.nih.gov/pubmed/25924506
  32. Kawaguchi K, Maruyama H, Hasunuma R, Kumazawa Y. Suppression of inflammatory responses after onset of collagen-induced arthritis in mice by oral administration of the Citrus flavanone naringin. Immunopharmacol Immunotoxicol. 2011;33(4):723-9.
    http://www.tandfonline.com/doi/abs/10.3109/08923973.2011.564186?journalCode=iipi20
  33. Ahmad SF, Zoheir KM, Abdel-hamied HE, et al. Amelioration of autoimmune arthritis by naringin through modulation of T regulatory cells and Th1/Th2 cytokines. Cell Immunol. 2014;287(2):112-20.
    https://www.sciencedirect.com/science/article/pii/S0008874914000033
  34. Lee JH, Kim GH. Evaluation of antioxidant and inhibitory activities for different subclasses flavonoids on enzymes for rheumatoid arthritis. J Food Sci. 2010;75(7):H212-7.
    http://onlinelibrary.wiley.com/doi/10.1111/j.1750-3841.2010.01755.x/abstract
  35. Yin FM, Xiao LB, Zhang Y. [Research progress on Drynaria fortunei naringin on inflammation and bone activity]. Zhongguo Gu Shang. 2015;28(2):182-6.
    https://www.ncbi.nlm.nih.gov/pubmed/25924506
  36. Xu T, Wang L, Tao Y, Ji Y, Deng F, Wu XH. The Function of Naringin in Inducing Secretion of Osteoprotegerin and Inhibiting Formation of Osteoclasts. Evid Based Complement Alternat Med. 2016;2016:8981650.
    https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4738947/
  37. Epasinghe DJ, Yiu C, Burrow MF. Effect of flavonoids on remineralization of artificial root caries. Aust Dent J. 2015.
    http://onlinelibrary.wiley.com/doi/10.1111/adj.12367/full
  38. Zhao Y, Li Z, Wang W, et al. Naringin Protects Against Cartilage Destruction in Osteoarthritis Through Repression of NF-κB Signaling Pathway. 2016;39(1):385-92.
    http://europepmc.org/abstract/MED/26438631
  39. Chtourou Y, Gargouri B, Kebieche M, Fetoui H. Naringin Abrogates Cisplatin-Induced Cognitive Deficits and Cholinergic Dysfunction Through the Down-Regulation of AChE Expression and iNOS Signaling Pathways in Hippocampus of Aged Rats. J Mol Neurosci. 2015;56(2):349-62.
    https://link.springer.com/article/10.1007/s12031-015-0547-0
  40. Chtourou Y, Aouey B, Kebieche M, Fetoui H. Protective role of naringin against cisplatin induced oxidative stress, inflammatory response and apoptosis in rat striatum via suppressing ROS-mediated NF-κB and P53 signaling pathways. Chem Biol Interact. 2015;239:76-86.
    https://www.sciencedirect.com/science/article/pii/S0009279715300089
  41. Ramalingayya GV, Nampoothiri M, Nayak PG, et al. Naringin and Rutin Alleviates Episodic Memory Deficits in Two Differentially Challenged Object Recognition Tasks. Pharmacogn Mag. 2016;12(Suppl 1):S63-70.
    https://www.researchgate.net/publication/294576255_Naringin_and_Rutin_Alleviates_Episodic_Memory_Deficits_in_Two_Differentially_Challenged_Object_Recognition_Tasks
  42. Wang DM, Yang YJ, Zhang L, Zhang X, Guan FF, Zhang LF. Naringin Enhances CaMKII Activity and Improves Long-Term Memory in a Mouse Model of Alzheimer’s Disease. Int J Mol Sci. 2013;14(3):5576-86.
    http://europepmc.org/articles/PMC3634479
  43. Wang D, Gao K, Li X, et al. Long-term naringin consumption reverses a glucose uptake defect and improves cognitive deficits in a mouse model of Alzheimer’s disease. Pharmacol Biochem Behav. 2012;102(1):13-20.
    https://www.sciencedirect.com/science/article/pii/S0091305712000846
  44. Kim HD, Jeong KH, Jung UJ, Kim SR. Naringin treatment induces neuroprotective effects in a mouse model of Parkinson’s disease in vivo, but not enough to restore the lesioned dopaminergic system. J Nutr Biochem. 2016;28:140-6.
    https://www.sciencedirect.com/science/article/pii/S095528631500306X
  45. Leem E, Nam JH, Jeon MT, et al. Naringin protects the nigrostriatal dopaminergic projection through induction of GDNF in a neurotoxin model of Parkinson’s disease. J Nutr Biochem. 2014;25(7):801-6.
    https://www.sciencedirect.com/science/article/pii/S0955286314000679
  46. Jung UJ, Kim SR. Effects of naringin, a flavanone glycoside in grapefruits and citrus fruits, on the nigrostriatal dopaminergic projection in the adult brain. Neural Regen Res. 2014;9(16):1514-7.
    https://www.ncbi.nlm.nih.gov/pubmed/25317167
  47. Sachdeva AK, Kuhad A, Chopra K. Naringin ameliorates memory deficits in experimental paradigm of Alzheimer’s disease by attenuating mitochondrial dysfunction. Pharmacol Biochem Behav. 2014;127:101-10.
    https://www.sciencedirect.com/science/article/pii/S0091305714002950
  48. Golechha M, Sarangal V, Bhatia J, Chaudhry U, Saluja D, Arya DS. Naringin ameliorates pentylenetetrazol-induced seizures and associated oxidative stress, inflammation, and cognitive impairment in rats: possible mechanisms of neuroprotection. Epilepsy Behav. 2014;41:98-102.
    https://www.ncbi.nlm.nih.gov/pubmed/25461197
  49. Adebiyi OA, Adebiyi OO, Owira PM. Naringin Reduces Hyperglycemia-Induced Cardiac Fibrosis by Relieving Oxidative Stress. PLoS ONE. 2016;11(3):e0149890.
    http://journals.plos.org/plosone/article?id=10.1371/journal.pone.0149890
  50. Dhanya R, Arun KB, Nisha VM, et al. Preconditioning L6 Muscle Cells with Naringin Ameliorates Oxidative Stress and Increases Glucose Uptake. PLoS ONE. 2015;10(7):e0132429.
    http://journals.plos.org/plosone/article?id=10.1371/journal.pone.0132429
  51. Wang D, Yan J, Chen J, Wu W, Zhu X, Wang Y. Naringin Improves Neuronal Insulin Signaling, Brain Mitochondrial Function, and Cognitive Function in High-Fat Diet-Induced Obese Mice. Cell Mol Neurobiol. 2015;35(7):1061-71.
    https://www.sciencedirect.com/science/article/pii/S0925443916000053
  52. Chen F, Zhang N, Ma X, et al. Naringin Alleviates Diabetic Kidney Disease through Inhibiting Oxidative Stress and Inflammatory Reaction. PLoS ONE. 2015;10(11):e0143868.
    http://journals.plos.org/plosone/article?id=10.1371/journal.pone.0143868
  53. Sun X, Li F, Ma X, et al. The Effects of Combined Treatment with Naringin and Treadmill Exercise on Osteoporosis in Ovariectomized Rats. Sci Rep. 2015;5:13009.
    https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4531319/
  54. Anuja GI, Latha PG, Suja SR, et al. Anti-inflammatory and analgesic properties of Drynaria quercifolia (L.) J. Smith. J Ethnopharmacol. 2010;132(2):456-60.
    https://www.sciencedirect.com/science/article/pii/S0378874110005854
  55. Kandhare AD, Alam J, Patil MV, Sinha A, Bodhankar SL. Wound healing potential of naringin ointment formulation via regulating the expression of inflammatory, apoptotic and growth mediators in experimental rats. Pharm Biol. 2016;54(3):419-32.
    http://www.tandfonline.com/doi/pdf/10.3109/13880209.2015.1038755
  56. Chanet A, Milenkovic D, Deval C, et al. Naringin, the major grapefruit flavonoid, specifically affects atherosclerosis development in diet-induced hypercholesterolemia in mice. J Nutr Biochem. 2012;23(5):469-77.
    https://www.sciencedirect.com/science/article/pii/S0955286311000672
  57. Etxeberria U, De la garza AL, Martíinez JA, Milagro I. Biocompounds Attenuating the Development of Obesity and Insulin Resistance Produced by a High-fat Sucrose Diet. Nat Prod Commun. 2015;10(8):1417-20.
    https://www.ncbi.nlm.nih.gov/pubmed/26434131
  58. Nakajima VM, Madeira JV, Macedo GA, Macedo JA. Biotransformation effects on anti lipogenic activity of citrus extracts. Food Chem. 2016;197 Pt B:1046-53.
    https://www.sciencedirect.com/science/article/pii/S0308814615302545
  59. Stohs SJ, Badmaev V. A Review of Natural Stimulant and Non-stimulant Thermogenic Agents. Phytother Res. 2016;30(5):732-40.
    https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5067548/
  60. Manna K, Das U, Das D, et al. Naringin inhibits gamma radiation-induced oxidative DNA damage and inflammation, by modulating p53 and NF-κB signaling pathways in murine splenocytes. Free Radic Res. 2015;49(4):422-39.
    https://www.medscape.com/medline/abstract/25812588
  61. Ren X, Shi Y, Zhao D, et al. Naringin protects ultraviolet B-induced skin damage by regulating p38 MAPK signal pathway. J Dermatol Sci. 2016;82(2):106-14.
    https://www.sciencedirect.com/science/article/pii/S0923181115300918
  62. Takekoshi S, Nagata H, Kitatani K. Flavonoids enhance melanogenesis in human melanoma cells. Tokai J Exp Clin Med. 2014;39(3):116-21.
    https://www.ncbi.nlm.nih.gov/pubmed/25248426
  63. Jiao HY, Su WW, Li PB, et al. Therapeutic effects of naringin in a guinea pig model of ovalbumin-induced cough-variant asthma. Pulm Pharmacol Ther. 2015;33:59-65.
    https://www.ncbi.nlm.nih.gov/pubmed/26169899
  64. Casey SC, Amedei A, Aquilano K, et al. Cancer prevention and therapy through the modulation of the tumor microenvironment. Semin Cancer Biol. 2015;35 Suppl:S199-223.
    https://www.sciencedirect.com/science/article/pii/S1044579X15000152
  65. Sun Y, Gu J. [Study on effect of naringenin in inhibiting migration and invasion of breast cancer cells and its molecular mechanism]. Zhongguo Zhong Yao Za Zhi. 2015;40(6):1144-50.
    https://www.ncbi.nlm.nih.gov/pubmed/26226761
  66. Song HM, Park GH, Eo HJ, et al. Anti-Proliferative Effect of Naringenin through p38-Dependent Downregulation of Cyclin D1 in Human Colorectal Cancer Cells. Biomol Ther (Seoul). 2015;23(4):339-44.
    http://europepmc.org/articles/PMC4489828
  67. Yen HR, Liu CJ, Yeh CC. Naringenin suppresses TPA-induced tumor invasion by suppressing multiple signal transduction pathways in human hepatocellular carcinoma cells. Chem Biol Interact. 2015;235:1-9.
    https://www.sciencedirect.com/science/article/pii/S0009279715001477
  68. Maggioni D, Nicolini G, Rigolio R, et al. Myricetin and naringenin inhibit human squamous cell carcinoma proliferation and migration in vitro. Nutr Cancer. 2014;66(7):1257-67.
    https://www.ncbi.nlm.nih.gov/pubmed/25256786
  69. Liao AC, Kuo CC, Huang YC, et al. Naringenin inhibits migration of bladder cancer cells through downregulation of AKT and MMP‑ Mol Med Rep. 2014;10(3):1531-6.
    https://www.spandidos-publications.com/mmr/10/3/1531
  70. Lou C, Zhang F, Yang M, et al. Naringenin decreases invasiveness and metastasis by inhibiting TGF-β-induced epithelial to mesenchymal transition in pancreatic cancer cells. PLoS ONE. 2012;7(12).
    http://journals.plos.org/plosone/article?id=10.1371/journal.pone.0050956
  71. Bao L, Liu F, Guo HB, et al. Naringenin inhibits proliferation, migration, and invasion as well as induces apoptosis of gastric cancer SGC7901 cell line by downregulation of AKT pathway. Tumour Biol. 2016.
    http://europepmc.org/abstract/MED/26960693
  72. Song HM, Park GH, Eo HJ, Jeong JB. Naringenin-Mediated ATF3 Expression Contributes to Apoptosis in Human Colon Cancer. Biomol Ther (Seoul). 2016;24(2):140-6.
    https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4774494/
  73. Abaza MS, Orabi KY, Al-quattan E, Al-attiyah RJ. Growth inhibitory and chemo-sensitization effects of naringenin, a natural flavanone purified from Thymus vulgaris, on human breast and colorectal cancer. Cancer Cell Int. 2015;15:46.
    https://cancerci.biomedcentral.com/articles/10.1186/s12935-015-0194-0
  74. Ayob Z, Mohd bohari SP, Abd samad A, Jamil S. Cytotoxic Activities against Breast Cancer Cells of Local Justicia gendarussa Crude Extracts. Evid Based Complement Alternat Med. 2014.
    https://www.hindawi.com/journals/ecam/2014/732980/
  75. Li RF, Feng YQ, Chen JH, Ge LT, Xiao SY, Zuo XL. Naringenin suppresses K562 human leukemia cell proliferation and ameliorates Adriamycin-induced oxidative damage in polymorphonuclear leukocytes. Exp Ther Med. 2015;9(3):697-706.
    https://spandidos-publications.com/10.3892/etm.2015.2185
  76. Ahamad MS, Siddiqui S, Jafri A, Ahmad S, Afzal M, Arshad M. Induction of apoptosis and antiproliferative activity of naringenin in human epidermoid carcinoma cell through ROS generation and cell cycle arrest. PLoS ONE. 2014;9(10):e110003.
    https://www.researchgate.net/publication/267329547_Induction_of_Apoptosis_and_Antiproliferative_Activity_of_Naringenin_in_Human_Epidermoid_Carcinoma_Cell_through_ROS_Generation_and_Cell_Cycle_Arrest
  77. Arul D, Subramanian P. Naringenin (citrus flavonone) induces growth inhibition, cell cycle arrest and apoptosis in human hepatocellular carcinoma cells. Pathol Oncol Res. 2013;19(4):763-70.
    https://link.springer.com/article/10.1007/s12253-013-9641-1
  78. Ekambaram G, Rajendran P, Magesh V, Sakthisekaran D. Naringenin reduces tumor size and weight lost in N-methyl-N’-nitro-N-nitrosoguanidine-induced gastric carcinogenesis in rats. Nutr Res. 2008;28(2):106-12.
    https://www.sciencedirect.com/science/article/pii/S0271531707002837
  79. Qin L, Jin L, Lu L, et al. Naringenin reduces lung metastasis in a breast cancer resection model. Protein Cell. 2011;2(6):507-16.
    https://link.springer.com/article/10.1007/s13238-011-1056-8
  80. Sabarinathan D, Mahalakshmi P, Vanisree AJ. Naringenin, a flavanone inhibits the proliferation of cerebrally implanted C6 glioma cells in rats. Chem Biol Interact. 2011;189(1-2):26-36.
    https://www.sciencedirect.com/science/article/pii/S0009279710005740
  81. Bodduluru LN, Kasala ER, Madhana RM, et al. Naringenin ameliorates inflammation and cell proliferation in benzo(a)pyrene induced pulmonary carcinogenesis by modulating CYP1A1, NFκB and PCNA expression. Int Immunopharmacol. 2016;30:102-10.
    https://www.sciencedirect.com/science/article/pii/S1567576915302046
  82. Sabarinathan D, Mahalakshmi P, Vanisree AJ. Naringenin promote apoptosis in cerebrally implanted C6 glioma cells. Mol Cell Biochem. 2010;345(1-2):215-22.
    https://link.springer.com/article/10.1007/s11010-010-0575-6
  83. Shi D, Xu Y, Du X, et al. Co-treatment of THP-1 cells with naringenin and curcumin induces cell cycle arrest and apoptosis via numerous pathways. Mol Med Rep. 2015;12(6):8223-8.
    http://europepmc.org/abstract/MED/26496980
  84. Torricelli P, Ricci P, Provenzano B, Lentini A, Tabolacci C. Synergic effect of α-tocopherol and naringenin in transglutaminase-induced differentiation of human prostate cancer cells. Amino Acids. 2011;41(5):1207-14.
    https://link.springer.com/article/10.1007/s00726-010-0788-8
  85. Bulzomi P, Bolli A, Galluzzo P, Acconcia F, Ascenzi P, Marino M. The naringenin-induced proapoptotic effect in breast cancer cell lines holds out against a high bisphenol a background. IUBMB Life. 2012;64(8):690-6.
    http://onlinelibrary.wiley.com/doi/10.1002/iub.1049/full
  86. Kumar SP, Birundha K, Kaveri K, Devi KT. Antioxidant studies of chitosan nanoparticles containing naringenin and their cytotoxicity effects in lung cancer cells. Int J Biol Macromol. 2015;78:87-95.
    https://www.sciencedirect.com/science/article/pii/S014181301500197X
  87. Fouad AA, Albuali WH, Jresat I. Protective Effect of Naringenin against Lipopolysaccharide-Induced Acute Lung Injury in Rats. Pharmacology. 2016;97(5-6):224-32.
    https://www.karger.com/Article/Abstract/444262
  88. Mostafa Hel-S, Abd el-baset SA, Kattaia AA, Zidan RA, Al sadek MM. Efficacy of naringenin against permethrin-induced testicular toxicity in rats. Int J Exp Pathol. 2016;97(1):37-49.
    https://www.ncbi.nlm.nih.gov/pubmed/26867500
  89. Chtourou Y, Slima AB, Gdoura R, Fetoui H. Naringenin Mitigates Iron-Induced Anxiety-Like Behavioral Impairment, Mitochondrial Dysfunctions, Ectonucleotidases and Acetylcholinesterase Alteration Activities in Rat Hippocampus. Neurochem Res. 2015;40(8):1563-75.
    https://www.medscape.com/medline/abstract/26050208
  90. Sachdeva S, Pant SC, Kushwaha P, Bhargava R, Flora SJ. Sodium tungstate induced neurological alterations in rat brain regions and their response to antioxidants. Food Chem Toxicol. 2015;82:64-71.
    https://pubag.nal.usda.gov/catalog/5419697
  91. Lin JL, Wang YD, Ma Y, et al. Protective effects of naringenin eye drops on N-methyl-N-nitrosourea-induced photoreceptor cell death in rats. Int J Ophthalmol. 2014;7(3):391-6.
    http://europepmc.org/articles/PMC4067647
  92. Nahmias Y, Goldwasser J, Casali M, et al. Apolipoprotein B-dependent hepatitis C virus secretion is inhibited by the grapefruit flavonoid naringenin. Hepatology. 2008;47(5):1437-45.
    http://onlinelibrary.wiley.com/doi/10.1002/hep.22197/full
  93. Paredes A, Alzuru M, Mendez J, Rodríguez-ortega M. Anti-Sindbis activity of flavanones hesperetin and naringenin. Biol Pharm Bull. 2003;26(1):108-9.
    https://www.jstage.jst.go.jp/article/bpb/26/1/26_1_108/_article
  94. Pinho-ribeiro FA, Zarpelon AC, Fattori V, et al. Naringenin reduces inflammatory pain in mice. 2016;105:508-519.
    https://www.sciencedirect.com/science/article/pii/S002839081630051X
  95. Manchope MF, Calixto-campos C, Coelho-silva L, et al. Naringenin Inhibits Superoxide Anion-Induced Inflammatory Pain: Role of Oxidative Stress, Cytokines, Nrf-2 and the NO-cGMP-PKG-KATPChannel Signaling Pathway. PLoS ONE. 2016;11(4).
    https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4821586/
  96. Oršolić N, Goluža E, Dikić D, et al. Role of flavonoids on oxidative stress and mineral contents in the retinoic acid-induced bone loss model of rat. Eur J Nutr. 2014;53(5):1217-27.
    https://link.springer.com/article/10.1007/s00394-013-0622-7
  97. La VD, Tanabe S, Grenier D. Naringenin inhibits human osteoclastogenesis and osteoclastic bone resorption. J Periodont Res. 2009;44(2):193-8.
    http://onlinelibrary.wiley.com/doi/10.1111/j.1600-0765.2008.01107.x/abstract
  98. Ming LG, Lv X, Ma XN, et al. The prenyl group contributes to activities of phytoestrogen 8-prenynaringenin in enhancing bone formation and inhibiting bone resorption in vitro. Endocrinology. 2013;154(3):1202-14.
    http://press.endocrine.org/doi/10.1210/en.2012-2086
  99. Ming LG, Ge BF, Wang MG, Chen KM. Comparison between 8-prenylnarigenin and narigenin concerning their activities on promotion of rat bone marrow stromal cells’ osteogenic differentiation in vitro. Cell Prolif. 2012;45(6):508-15.
    http://onlinelibrary.wiley.com/doi/10.1111/j.1365-2184.2012.00844.x/abstract
  100. Amat-ur-rasool H, Ahmed M. Designing Second Generation Anti-Alzheimer Compounds as Inhibitors of Human Acetylcholinesterase: Computational Screening of Synthetic Molecules and Dietary Phytochemicals. PLoS ONE. 2015;10(9).
    http://journals.plos.org/plosone/article?id=10.1371/journal.pone.0136509
  101. Ghofrani S, Joghataei MT, Mohseni S, et al. Naringenin improves learning and memory in an Alzheimer’s disease rat model: Insights into the underlying mechanisms. Eur J Pharmacol. 2015;764:195-201.
    https://www.sciencedirect.com/science/article/pii/S0014299915301291
  102. Chtourou Y, Fetoui H, Gdoura R. Protective effects of naringenin on iron-overload-induced cerebral cortex neurotoxicity correlated with oxidative stress. Biol Trace Elem Res. 2014;158(3):376-83.
    https://link.springer.com/article/10.1007/s12011-014-9948-0
  103. Zbarsky V, Datla KP, Parkar S, Rai DK, Aruoma OI, Dexter DT. Neuroprotective properties of the natural phenolic antioxidants curcumin and naringenin but not quercetin and fisetin in a 6-OHDA model of Parkinson’s disease. Free Radic Res. 2005;39(10):1119-25.
    http://www.tandfonline.com/doi/abs/10.1080/10715760500233113?journalCode=ifra20
  104. Lim W, Song G. Naringenin-induced migration of embrynoic trophectoderm cells is mediated via PI3K/AKT and ERK1/2 MAPK signaling cascades. Mol Cell Endocrinol. 2016;428:28-37.
    https://www.sciencedirect.com/science/article/pii/S0303720716300661
  105. Yi LT, Li CF, Zhan X, et al. Involvement of monoaminergic system in the antidepressant-like effect of the flavonoid naringenin in mice. Prog Neuropsychopharmacol Biol Psychiatry. 2010;34(7):1223-8.
    https://www.sciencedirect.com/science/article/pii/S0278584610002411
  106. Yi LT, Liu BB, Li J, et al. BDNF signaling is necessary for the antidepressant-like effect of naringenin. Prog Neuropsychopharmacol Biol Psychiatry. 2014;48:135-41.
    https://www.sciencedirect.com/science/article/pii/S0278584613002182
  107. Yi LT, Li J, Li HC, et al. Antidepressant-like behavioral, neurochemical and neuroendocrine effects of naringenin in the mouse repeated tail suspension test. Prog Neuropsychopharmacol Biol Psychiatry. 2012;39(1):175-81.
    https://www.sciencedirect.com/science/article/pii/S0278584612001443
  108. Ozkaya A, Sahin Z, Dag U, Ozkaraca M. Effects of Naringenin on Oxidative Stress and Histopathological Changes in the Liver of Lead Acetate Administered Rats. J Biochem Mol Toxicol. 2016;30(5):243-8.
    http://onlinelibrary.wiley.com/doi/10.1002/jbt.21785/full
  109. Roy S, Ahmed F, Banerjee S, Saha U. Naringenin ameliorates streptozotocin-induced diabetic rat renal impairment by downregulation of TGF-β1 and IL-1 via modulation of oxidative stress correlates with decreased apoptotic events. Pharm Biol. 2016;:1-12.
    https://www.ncbi.nlm.nih.gov/pubmed/26928632
  110. Ren B, Qin W, Wu F, et al. Apigenin and naringenin regulate glucose and lipid metabolism, and ameliorate vascular dysfunction in type 2 diabetic rats. Eur J Pharmacol. 2016;773:13-23.
    https://www.sciencedirect.com/science/article/pii/S0014299916300024
  111. Bhattacharya S, Oksbjerg N, Young JF, Jeppesen PB. Caffeic acid, naringenin and quercetin enhance glucose-stimulated insulin secretion and glucose sensitivity in INS-1E cells. Diabetes Obes Metab. 2014;16(7):602-12.
    http://onlinelibrary.wiley.com/doi/10.1111/dom.12236/abstract
  112. Habauzit V, Verny MA, Milenkovic D, et al. Flavanones protect from arterial stiffness in postmenopausal women consuming grapefruit juice for 6 mo: a randomized, controlled, crossover trial. Am J Clin Nutr. 2015;102(1):66-74.
    https://www.ncbi.nlm.nih.gov/pubmed/26016866
  113. Orhan IE, Nabavi SF, Daglia M, Tenore GC, Mansouri K, Nabavi SM. Naringenin and atherosclerosis: a review of literature. Curr Pharm Biotechnol. 2015;16(3):245-51.
    https://www.ncbi.nlm.nih.gov/pubmed/25483717
  114. Zhang N, Yang Z, Yuan Y, et al. Naringenin attenuates pressure overload-induced cardiac hypertrophy. Exp Ther Med. 2015;10(6):2206-2212.
    https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4665326/
  115. Li YR, Chen DY, Chu CL, et al. Naringenin inhibits dendritic cell maturation and has therapeutic effects in a murine model of collagen-induced arthritis. J Nutr Biochem. 2015;26(12):1467-78.
    https://www.sciencedirect.com/science/article/pii/S0955286315001825
  116. Du G, Jin L, Han X, Song Z, Zhang H, Liang W. Naringenin: a potential immunomodulator for inhibiting lung fibrosis and metastasis. Cancer Res. 2009;69(7):3205-12.
    http://cancerres.aacrjournals.org/content/69/7/3205
  117. Qin L, Jin L, Lu L, et al. Naringenin reduces lung metastasis in a breast cancer resection model. Protein Cell. 2011;2(6):507-16.
    https://link.springer.com/article/10.1007/s13238-011-1056-8
  118. Nasr-bouzaiene N, Sassi A, Bedoui A, Krifa M, Chekir-ghedira L, Ghedira K. Immunomodulatory and cellular antioxidant activities of pure compounds from Teucrium ramosissimum Desf. Tumour Biol. 2015.
    https://www.researchgate.net/publication/287806907_Immunomodulatory_and_cellular_antioxidant_activities_of_pure_compounds_from_Teucrium_ramosissimum_Desf
  119. Kim JH, Lee JK. Naringenin enhances NK cell lysis activity by increasing the expression of NKG2D ligands on Burkitt’s lymphoma cells. Arch Pharm Res. 2015;38(11):2042-8.
    https://link.springer.com/article/10.1007/s12272-015-0624-5
  120. Farzaei MH, Rahimi R, Abdollahi M. The role of dietary polyphenols in the management of inflammatory bowel disease. Curr Pharm Biotechnol. 2015;16(3):196-210.
    http://www.eurekaselect.com/127791/article
  121. Al-rejaie SS, Abuohashish HM, Al-enazi MM, Al-assaf AH, Parmar MY, Ahmed MM. Protective effect of naringenin on acetic acid-induced ulcerative colitis in rats. World J Gastroenterol. 2013;19(34):5633-44.
    https://www.wjgnet.com/1007-9327/full/v19/i34/5633.htm
  122. Azuma T, Shigeshiro M, Kodama M, Tanabe S, Suzuki T. Supplemental naringenin prevents intestinal barrier defects and inflammation in colitic mice. J Nutr. 2013;143(6):827-34.
    http://jn.nutrition.org/content/143/6/827.long
  123. Chattopadhyay D, Sen S, Chatterjee R, Roy D, James J, Thirumurugan K. Context- and dose-dependent modulatory effects of naringenin on survival and development of Drosophila melanogaster. Biogerontology. 2016;17(2):383-93.
    https://www.medscape.com/medline/abstract/26520643
  124. Assini JM, Mulvihill EE, Burke AC, et al. Naringenin prevents obesity, hepatic steatosis, and glucose intolerance in male mice independent of fibroblast growth factor 21. Endocrinology. 2015;156(6):2087-102.
    https://academic.oup.com/endo/article/156/6/2087/2422904
  125. Ke JY, Cole RM, Hamad EM, et al. Citrus flavonoid, naringenin, increases locomotor activity and reduces diacylglycerol accumulation in skeletal muscle of obese ovariectomized mice. Mol Nutr Food Res. 2016;60(2):313-24.
    https://cancer.osu.edu/research-and-education/find-a-scientific-publication/citrus-flavonoid-naringenin-increases-locomotor-activity-and-reduces-diacylglycerol-accumulation-in-skeletal-muscle-of-obese-ovariectomized-mice#!
  126. Kumar S, Tiku AB. Biochemical and Molecular Mechanisms of Radioprotective Effects of Naringenin, a Phytochemical from Citrus Fruits. J Agric Food Chem. 2016;64(8):1676-85.
    http://pubs.acs.org/doi/abs/10.1021/acs.jafc.5b05067
  127. Jung SK, Ha SJ, Jung CH, et al. Naringenin targets ERK2 and suppresses UVB-induced photoaging. J Cell Mol Med. 2016;20(5):909-19.
    http://onlinelibrary.wiley.com/doi/10.1111/jcmm.12780/full
  128. Martinez RM, Pinho-ribeiro FA, Steffen VS, et al. Topical Formulation Containing Naringenin: Efficacy against Ultraviolet B Irradiation-Induced Skin Inflammation and Oxidative Stress in Mice. PLoS ONE. 2016;11(1):e0146296.
    http://journals.plos.org/plosone/article?id=10.1371/journal.pone.0146296
  129. Martinez RM, Pinho-ribeiro FA, Steffen VS, et al. Naringenin Inhibits UVB Irradiation-Induced Inflammation and Oxidative Stress in the Skin of Hairless Mice. J Nat Prod. 2015;78(7):1647-55.
    http://pubs.acs.org/doi/abs/10.1021/acs.jnatprod.5b00198
  130. Kawakami CM, Gaspar LR. Mangiferin and naringenin affect the photostability and phototoxicity of sunscreens containing avobenzone. J Photochem Photobiol B, Biol. 2015;151:239-47.
    https://www.sciencedirect.com/science/article/pii/S1011134415002638
  131. Nasr bouzaiene N, Chaabane F, Sassi A, Chekir-ghedira L, Ghedira K. Effect of apigenin-7-glucoside, genkwanin and naringenin on tyrosinase activity and melanin synthesis in B16F10 melanoma cells. Life Sci. 2016;144:80-5.
    https://www.sciencedirect.com/science/article/pii/S002432051530093X
  132. Liu Y, Wang P, Chen F, et al. Role of plant polyphenols in acrylamide formation and elimination. Food Chem. 2015;186:46-53.
    https://www.sciencedirect.com/science/article/pii/S030881461500494X
  133. Keiler AM, Dörfelt P, Chatterjee N, et al. Assessment of the effects of naringenin-type flavanones in uterus and vagina. J Steroid Biochem Mol Biol. 2015;145:49-57.
    https://www.sciencedirect.com/science/article/pii/S0960076014002349
  134. Lee CJ, Wilson L, Jordan MA, Nguyen V, Tang J, Smiyun G. Hesperidin suppressed proliferations of both human breast cancer and androgen-dependent prostate cancer cells. Phytother Res. 2010;24 Suppl 1:S15-9.
    http://onlinelibrary.wiley.com/doi/10.1002/ptr.2856/abstract
  135. Choi EJ. Hesperetin induced G1-phase cell cycle arrest in human breast cancer MCF-7 cells: involvement of CDK4 and p21. Nutr Cancer. 2007;59(1):115-9.
    https://www.ncbi.nlm.nih.gov/pubmed/17927510
  136. Bracke M, Vyncke B, Opdenakker G, Foidart JM, De pestel G, Mareel M. Effect of catechins and citrus flavonoids on invasion in vitro. Clin Exp Metastasis. 1991;9(1):13-25.
    https://link.springer.com/article/10.1007/BF01831706
  137. Banjerdpongchai R, Wudtiwai B, Khaw-on P, Rachakhom W, Duangnil N, Kongtawelert P. Hesperidin from Citrus seed induces human hepatocellular carcinoma HepG2 cell apoptosis via both mitochondrial and death receptor pathways. Tumour Biol. 2016;37(1):227-37.
    https://link.springer.com/article/10.1007/s13277-015-3774-7
  138. Nazari M, Ghorbani A, Hekmat-doost A, Jeddi-tehrani M, Zand H. Inactivation of nuclear factor-κB by citrus flavanone hesperidin contributes to apoptosis and chemo-sensitizing effect in Ramos cells. Eur J Pharmacol. 2011;650(2-3):526-33.
    https://www.sciencedirect.com/science/article/pii/S0014299910010800
  139. Park HJ, Kim MJ, Ha E, Chung JH. Apoptotic effect of hesperidin through caspase3 activation in human colon cancer cells, SNU-C4. Phytomedicine. 2008;15(1-2):147-51.
    https://www.sciencedirect.com/science/article/pii/S0944711307001857
  140. Ghorbani A, Nazari M, Jeddi-tehrani M, Zand H. The citrus flavonoid hesperidin induces p53 and inhibits NF-κB activation in order to trigger apoptosis in NALM-6 cells: involvement of PPARγ-dependent mechanism. Eur J Nutr. 2012;51(1):39-46.
    https://link.springer.com/article/10.1007/s00394-011-0187-2
  141. Adan A, Baran Y. The pleiotropic effects of fisetin and hesperetin on human acute promyelocytic leukemia cells are mediated through apoptosis, cell cycle arrest, and alterations in signaling networks. Tumour Biol. 2015;36(11):8973-84.
    https://link.springer.com/article/10.1007/s13277-015-3597-6
  142. Yumnam S, Hong GE, Raha S, et al. Mitochondrial Dysfunction and Ca(2+) Overload Contributes to Hesperidin Induced Paraptosis in Hepatoblastoma Cells, HepG2. J Cell Physiol. 2016;231(6):1261-8.
    http://onlinelibrary.wiley.com/doi/10.1002/jcp.25222/abstract
  143. Birsu cincin Z, Unlu M, Kiran B, Sinem bireller E, Baran Y, Cakmakoglu B. Anti-proliferative, apoptotic and signal transduction effects of hesperidin in non-small cell lung cancer cells. Cell Oncol (Dordr). 2015;38(3):195-204.
    https://link.springer.com/article/10.1007/s13402-015-0222-z
  144. Palit S, Kar S, Sharma G, Das PK. Hesperetin Induces Apoptosis in Breast Carcinoma by Triggering Accumulation of ROS and Activation of ASK1/JNK Pathway. J Cell Physiol. 2015;230(8):1729-39.
    https://lup.lub.lu.se/search/publication/a0f759d3-1dfa-40d0-8acd-f22d8d482f93
  145. Tanaka T, Makita H, Kawabata K, et al. Chemoprevention of azoxymethane-induced rat colon carcinogenesis by the naturally occurring flavonoids, diosmin and hesperidin. Carcinogenesis. 1997;18(5):957-65.
    https://academic.oup.com/carcin/article/18/5/957/2365109
  146. Kamaraj S, Anandakumar P, Jagan S, Ramakrishnan G, Devaki T. Modulatory effect of hesperidin on benzo(a)pyrene induced experimental lung carcinogenesis with reference to COX-2, MMP-2 and MMP-9. Eur J Pharmacol. 2010;649(1-3):320-7.
    https://www.sciencedirect.com/science/article/pii/S0014299910008988
  147. Yang M, Tanaka T, Hirose Y, Deguchi T, Mori H, Kawada Y. Chemopreventive effects of diosmin and hesperidin on N-butyl-N-(4-hydroxybutyl)nitrosamine-induced urinary-bladder carcinogenesis in male ICR mice. Int J Cancer. 1997;73(5):719-24.
    http://onlinelibrary.wiley.com/doi/10.1002/(SICI)1097-0215(19971127)73:5%3C719::AID-IJC18%3E3.0.CO;2-0/abstract
  148. Tanaka T, Makita H, Ohnishi M, et al. Chemoprevention of 4-nitroquinoline 1-oxide-induced oral carcinogenesis by dietary curcumin and hesperidin: comparison with the protective effect of beta-carotene. Cancer Res. 1994;54(17):4653-9.
    http://cancerres.aacrjournals.org/content/54/17/4653
  149. Tanaka T, Makita H, Ohnishi M, et al. Chemoprevention of 4-nitroquinoline 1-oxide-induced oral carcinogenesis in rats by flavonoids diosmin and hesperidin, each alone and in combination. Cancer Res. 1997;57(2):246-52.
    http://cancerres.aacrjournals.org/content/57/2/246.short
  150. Tanaka T, Makita H, Kawabata K, et al. Modulation of N-methyl-N-amylnitrosamine-induced rat oesophageal tumourigenesis by dietary feeding of diosmin and hesperidin, both alone and in combination. Carcinogenesis. 1997;18(4):761-9.
    https://academic.oup.com/carcin/article/18/4/761/2365093
  151. Zhang J, Wu D, Vikash, et al. Hesperetin Induces the Apoptosis of Gastric Cancer Cells via Activating Mitochondrial Pathway by Increasing Reactive Oxygen Species. Dig Dis Sci. 2015;60(10):2985-95.
    https://link.springer.com/article/10.1007/s10620-015-3696-7
  152. Saiprasad G, Chitra P, Manikandan R, Sudhandiran G. Hesperidin induces apoptosis and triggers autophagic markers through inhibition of Aurora-A mediated phosphoinositide-3-kinase/Akt/mammalian target of rapamycin and glycogen synthase kinase-3 beta signalling cascades in experimental colon carcinogenesis. Eur J Cancer. 2014;50(14):2489-507.
    https://www.sciencedirect.com/science/article/pii/S0959804914007576
  153. Gwiazdowska D, Juś K, Jasnowska-małecka J, Kluczyńska K. The impact of polyphenols on Bifidobacterium growth. Acta Biochim Pol. 2015;62(4):895-901.
    http://europepmc.org/abstract/MED/26619254
  154. Abuelsaad AS, Allam G, Al-solumani AA. Hesperidin inhibits inflammatory response induced by Aeromonas hydrophila infection and alters CD4+/CD8+ T cell ratio. Mediators Inflamm. 2014;2014:393217.
    https://www.hindawi.com/journals/mi/2014/393217/
  155. Siddiqi A, Nafees S, Rashid S, Sultana S, Saidullah B. Hesperidin ameliorates trichloroethylene-induced nephrotoxicity by abrogation of oxidative stress and apoptosis in wistar rats. Mol Cell Biochem. 2015;406(1-2):9-20.
    https://link.springer.com/article/10.1007/s11010-015-2400-8
  156. Anandan R, Subramanian P. Renal protective effect of hesperidin on gentamicin-induced acute nephrotoxicity in male Wistar albino rats. Redox Rep. 2012;17(5):219-26.
    http://www.tandfonline.com/doi/full/10.1179/1351000212Y.0000000019
  157. Sahu BD, Kuncha M, Sindhura GJ, Sistla R. Hesperidin attenuates cisplatin-induced acute renal injury by decreasing oxidative stress, inflammation and DNA Phytomedicine. 2013;20(5):453-60.
    https://www.sciencedirect.com/science/article/pii/S0944711312005089
  158. Kamel KM, Abd el-raouf OM, Metwally SA, Abd el-latif HA, El-sayed ME. Hesperidin and rutin, antioxidant citrus flavonoids, attenuate cisplatin-induced nephrotoxicity in rats. J Biochem Mol Toxicol. 2014;28(7):312-9.
    https://www.ncbi.nlm.nih.gov/pubmed/24819883
  159. Polat N, Ciftci O, Cetin A, Yılmaz T. Toxic effects of systemic cisplatin on rat eyes and the protective effect of hesperidin against this toxicity. Cutan Ocul Toxicol. 2016;35(1):1-7.
    http://www.tandfonline.com/doi/abs/10.3109/15569527.2014.999080?journalCode=icot20
  160. Kaya K, Ciftci O, Cetin A, Doğan H, Başak N. Hesperidin protects testicular and spermatological damages induced by cisplatin in rats. 2015;47(7):793-800.
    http://onlinelibrary.wiley.com/doi/10.1111/and.12332/abstract
  161. Omar HA, Mohamed WR, Arafa el-SA, et al. Hesperidin alleviates cisplatin-induced hepatotoxicity in rats without inhibiting its antitumor activity. Pharmacol Rep. 2016;68(2):349-56.
    https://www.sciencedirect.com/science/article/pii/S1734114015003242
  162. Saha RK, Takahashi T, Suzuki T. Glucosyl hesperidin prevents influenza a virus replication in vitro by inhibition of viral sialidase. Biol Pharm Bull. 2009;32(7):1188-92.
    https://www.jstage.jst.go.jp/article/bpb/32/7/32_7_1188/_article
  163. Carvalho OV, Botelho CV, Ferreira CG, et al. In vitro inhibition of canine distemper virus by flavonoids and phenolic acids: implications of structural differences for antiviral design. Res Vet Sci. 2013;95(2):717-24.
    https://www.sciencedirect.com/science/article/pii/S0034528813001392
  164. Bae EA, Han MJ, Lee M, Kim DH. In vitro inhibitory effect of some flavonoids on rotavirus infectivity. Biol Pharm Bull. 2000;23(9):1122-4.
    https://www.ncbi.nlm.nih.gov/pubmed/10993220
  165. Panasiak W, Wleklik M, Oraczewska A, Luczak M. Influence of flavonoids on combined experimental infections with EMC virus and Staphylococcus aureus in mice. Acta Microbiol Pol. 1989;38(2):185-8.
    https://www.ncbi.nlm.nih.gov/pubmed/2482663
  166. Ahmed YM, Messiha BA, Abo-saif AA. Protective Effects of Simvastatin and Hesperidin against Complete Freund’s Adjuvant-Induced Rheumatoid Arthritis in Rats. Pharmacology. 2015;96(5-6):217-25.
    https://www.karger.com/Article/Abstract/439538
  167. Li R, Cai L, Xie XF, Yang F, Li J. Hesperidin suppresses adjuvant arthritis in rats by inhibiting synoviocyte activity. Phytother Res. 2010;24 Suppl 1:S71-6.
    https://www.ncbi.nlm.nih.gov/pubmed/19585485
  168. Martin BR, Mccabe GP, Mccabe L, et al. Effect of Hesperidin With and Without a Calcium (Calcilock) Supplement on Bone Health in Postmenopausal Women. J Clin Endocrinol Metab. 2016;101(3):923-7.
    https://academic.oup.com/jcem/article/101/3/923/2804802
  169. Chiba H, Kim H, Matsumoto A, et al. Hesperidin prevents androgen deficiency-induced bone loss in male mice. Phytother Res. 2014;28(2):289-95.
    http://onlinelibrary.wiley.com/doi/10.1002/ptr.5001/abstract
  170. Habauzit V, Sacco SM, Gil-izquierdo A, et al. Differential effects of two citrus flavanones on bone quality in senescent male rats in relation to their bioavailability and metabolism. Bone. 2011;49(5):1108-16.
    https://www.sciencedirect.com/science/article/pii/S875632821101132X
  171. Tamilselvam K, Braidy N, Manivasagam T, et al. Neuroprotective effects of hesperidin, a plant flavanone, on rotenone-induced oxidative stress and apoptosis in a cellular model for Parkinson’s disease. Oxid Med Cell Longev. 2013;2013:102741.
    https://www.hindawi.com/journals/omcl/2013/102741/
  172. Khan MH, Parvez S. Hesperidin ameliorates heavy metal induced toxicity mediated by oxidative stress in brain of Wistar rats. J Trace Elem Med Biol. 2015;31:53-60.
    https://www.sciencedirect.com/science/article/pii/S0946672X15000322
  173. Wang D, Liu L, Zhu X, Wu W, Wang Y. Hesperidin alleviates cognitive impairment, mitochondrial dysfunction and oxidative stress in a mouse model of Alzheimer’s disease. Cell Mol Neurobiol. 2014;34(8):1209-21.
    https://link.springer.com/article/10.1007/s10571-014-0098-x
  174. Antunes MS, Goes AT, Boeira SP, Prigol M, Jesse CR. Protective effect of hesperidin in a model of Parkinson’s disease induced by 6-hydroxydopamine in aged mice. Nutrition. 2014;30(11-12):1415-22.
    https://www.sciencedirect.com/science/article/pii/S0899900714001853
  175. El-marasy SA, Abdallah HM, El-shenawy SM, El-khatib AS, El-shabrawy OA, Kenawy SA. Anti-depressant effect of hesperidin in diabetic rats. Can J Physiol Pharmacol. 2014;92(11):945-52.
    https://www.ncbi.nlm.nih.gov/pubmed/25358020
  176. Donato F, De gomes MG, Goes AT, et al. Hesperidin exerts antidepressant-like effects in acute and chronic treatments in mice: possible role of l-arginine-NO-cGMP pathway and BDNF levels. Brain Res Bull. 2014;104:19-26.
    https://www.sciencedirect.com/science/article/pii/S0361923014000549
  177. Cai L, Li R, Wu QQ, Wu TN. [Effect of hesperidin on behavior and HPA axis of rat model of chronic stress-induced depression]. Zhongguo Zhong Yao Za Zhi. 2013;38(2):229-33.
    https://www.ncbi.nlm.nih.gov/pubmed/23672047
  178. Sharma M, Akhtar N, Sambhav K, Shete G, Bansal AK, Sharma SS. Emerging potential of citrus flavanones as an antioxidant in diabetes and its complications. Curr Top Med Chem. 2015;15(2):187-95.
    https://www.ncbi.nlm.nih.gov/pubmed/25496274
  179. De oliveira DM, Dourado GK, Cesar TB. Hesperidin associated with continuous and interval swimming improved biochemical and oxidative biomarkers in rats. J Int Soc Sports Nutr. 2013;10:27.
    https://jissn.biomedcentral.com/articles/10.1186/1550-2783-10-27
  180. Dimpfel W. Different anticonvulsive effects of hesperidin and its aglycone hesperetin on electrical activity in the rat hippocampus in-vitro. J Pharm Pharmacol. 2006;58(3):375-9.
    http://onlinelibrary.wiley.com/doi/10.1211/jpp.58.3.0012/abstract
  181. Zanotti simoes dourado GK, De abreu ribeiro LC, Zeppone carlos I, Borges césar T. Orange juice and hesperidin promote differential innate immune response in macrophages ex vivo. Int J Vitam Nutr Res. 2013;83(3):162-7.
    http://europepmc.org/abstract/MED/24846905
  182. Kamboh AA, Hang SQ, Khan MA, Zhu WY. In vivo immunomodulatory effects of plant flavonoids in lipopolysaccharide-challenged broilers. Animal. 2016;:1-7.
    https://www.researchgate.net/publication/301330649_In_vivo_immunomodulatory_effects_of_plant_flavonoids_in_lipopolysaccharide-challenged_broilers
  183. Amiot MJ, Riva C, Vinet A. Effects of dietary polyphenols on metabolic syndrome features in humans: a systematic review. Obes Rev. 2016.
    http://onlinelibrary.wiley.com/doi/10.1111/obr.12409/full
  184. Assini JM, Mulvihill EE, Huff MW. Citrus flavonoids and lipid metabolism. Curr Opin Lipidol. 2013;24(1):34-40.
    https://www.ncbi.nlm.nih.gov/pubmed/23254473
  185. Pradeep K, Ko KC, Choi MH, Kang JA, Chung YJ, Park SH. Protective effect of hesperidin, a citrus flavanoglycone, against γ-radiation-induced tissue damage in Sprague-Dawley rats. J Med Food. 2012;15(5):419-27.
    https://www.ncbi.nlm.nih.gov/pubmed/22404572
  186. Martinez RM, Pinho-ribeiro FA, Steffen VS, et al. Topical formulation containing hesperidin methyl chalcone inhibits skin oxidative stress and inflammation induced by ultraviolet B irradiation. Photochem Photobiol Sci. 2016;15(4):554-63.
    http://pubs.rsc.org/en/content/articlelanding/2016/pp/c5pp00467e
  187. Madduma hewage SR, Piao MJ, Kang KA, et al. Hesperidin Attenuates Ultraviolet B-Induced Apoptosis by Mitigating Oxidative Stress in Human Keratinocytes. Biomol Ther (Seoul). 2016;24(3):312-9.
    https://www.ncbi.nlm.nih.gov/pubmed/26797112
  188. Said UZ, Saada HN, Abd-alla MS, Elsayed ME, Amin AM. Hesperidin attenuates brain biochemical changes of irradiated rats. Int J Radiat Biol. 2012;88(8):613-8.
    http://www.tandfonline.com/doi/abs/10.3109/09553002.2012.694008?journalCode=irab20
  189. Lee YR, Jung JH, Kim HS. Hesperidin partially restores impaired immune and nutritional function in irradiated mice. J Med Food. 2011;14(5):475-82.
    https://www.ncbi.nlm.nih.gov/pubmed/21434774
  190. Giannini I, Amato A, Basso L, et al. Flavonoids mixture (diosmin, troxerutin, hesperidin) in the treatment of acute hemorrhoidal disease: a prospective, randomized, triple-blind, controlled trial. Tech Coloproctol. 2015;19(6):339-45.
    https://www.ncbi.nlm.nih.gov/pubmed/25893991
  191. Man G, Mauro TM, Zhai Y, et al. Topical hesperidin enhances epidermal function in an aged murine model. J Invest Dermatol. 2015;135(4):1184-7.
    https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4366273/
  192. Usach I, Taléns-visconti R, Magraner-pardo L, Peris JE. Hesperetin induces melanin production in adult human epidermal melanocytes. Food Chem Toxicol. 2015;80:80-4.
    https://www.sciencedirect.com/science/article/pii/S0278691515000630
  193. Kurata Y, Fukushima S, Hagiwara A, Ito H, Ogawa K, Ito N. Carcinogenicity study of methyl hesperidin in B6C3F1 mice. Food Chem Toxicol. 1990;28(9):613-8.
    https://www.sciencedirect.com/science/article/pii/027869159090168M
  194. Carpenter KJ. The discovery of vitamin C. Ann Nutr Metab. 2012;61(3):259-64.
    https://www.ncbi.nlm.nih.gov/pubmed/23183299
  195. Padayatty SJ, Levine M. Vitamin C: the known, the unknown, and Goldilocks. Oral Dis. 2016;
    http://onlinelibrary.wiley.com/doi/10.1111/odi.12446/abstract
  196. Shaw JH, Phillips PH, Elvehjem CA. Acute and chronic ascorbic acid deficiencies in the rhesus monkey. Nutr. 1945; vol 29. No. 6:365-372.
    https://www.researchgate.net/publication/237306029_ACUTE_AND_CHEONIC_ASCORBIC_ACID_DEFICIENCIES_IN_THE_RHESUS_MONKEY_1
  197. Mastrangelo D, Massai L, Lo coco F, et al. Cytotoxic effects of high concentrations of sodium ascorbate on human myeloid cell lines. Ann Hematol. 2015;94(11):1807-16.
    https://link.springer.com/article/10.1007/s00277-015-2464-2
  198. Sertkaya A, Wong HH, Jessup A, Beleche T. Key cost drivers of pharmaceutical clinical trials in the United States. Sage Journals. 2016; vol 13, issue 2.
    https://www.ncbi.nlm.nih.gov/pubmed/26908540
  199. PDQ Cancer Information Summaries – High-dose Vitamin C. 2015. [Available] http://www.ncbi.nlm.nih.gov/books/NBK127724/#CDR0000742253__1 [May 11, 2016].
  200. Venturelli S, Sinnberg TW, Niessner H, Busch C. Molecular mechanisms of pharmacological doses of ascorbate on cancer cells. Wien Med Wochenschr. 2015;165(11-12):251-7.
    https://link.springer.com/article/10.1007/s10354-015-0356-7
  201. Cameron E. Vitamin C and cancer: an overview. Int J Vitam Nutr Res Suppl. 1982;23:115-27.
    https://www.ncbi.nlm.nih.gov/pubmed/6180998/
  202. Malavolti M, Malagoli C, Fiorentini C, et al. Association between dietary vitamin C and risk of cutaneous melanoma in a population of Northern Italy. Int J Vitam Nutr Res. 2013;83(5):291-8.
    https://moh-it.pure.elsevier.com/en/publications/association-between-dietary-vitamin-c-and-risk-of-cutaneous-melan
  203. Vance TM, Wang Y, Su LJ, et al. Dietary Total Antioxidant Capacity is Inversely Associated with Prostate Cancer Aggressiveness in a Population-Based Study. Nutr Cancer. 2016;68(2):214-24.
    http://www.tandfonline.com/doi/abs/10.1080/01635581.2016.1134596
  204. Kong P, Cai Q, Geng Q, et al. Vitamin intake reduce the risk of gastric cancer: meta-analysis and systematic review of randomized and observational studies. PLoS ONE. 2014;9(12):e116060.
    http://journals.plos.org/plosone/article?id=10.1371/journal.pone.0116060
  205. Benade L, Howard T, Burk D. Synergistic killing of Ehrlich ascites carcinoma cells by ascorbate and 3-amino-1,2,4,-triazole. Oncology. 1969;23(1):33-43.
    https://www.ncbi.nlm.nih.gov/pubmed/5774953
  206. Pires AS, Marques CR, Encarnação JC, et al. Ascorbic acid and colon cancer: an oxidative stimulus to cell death depending on cell profile. Eur J Cell Biol. 2016;
    https://www.sciencedirect.com/science/article/pii/S0171933516300401
  207. Uetaki M, Tabata S, Nakasuka F, Soga T, Tomita M. Metabolomic alterations in human cancer cells by vitamin C-induced oxidative stress. Sci Rep. 2015;5:13896.
    https://www.researchgate.net/publication/281636045_Metabolomic_alterations_in_human_cancer_cells_by_Vitamin_C-induced_oxidative_stress
  208. Serrano OK, Parrow NL, Violet PC, et al. Antitumor effect of pharmacologic ascorbate in the B16 murine melanoma model. Free Radic Biol Med. 2015;87:193-203.
    https://www.sciencedirect.com/science/article/pii/S0891584915003007
  209. Mastrangelo D, Massai L, Lo coco F, et al. Cytotoxic effects of high concentrations of sodium ascorbate on human myeloid cell lines. Ann Hematol. 2015;94(11):1807-16.
    https://link.springer.com/article/10.1007/s00277-015-2464-2
  210. Chen N, Yin S, Song X, Fan L, Hu H. Vitamin B₂ Sensitizes Cancer Cells to Vitamin-C-Induced Cell Death via Modulation of Akt and Bad Phosphorylation. J Agric Food Chem. 2015;63(30):6739-48.
    https://www.ncbi.nlm.nih.gov/pubmed/26165392
  211. Campbell EJ, Vissers MC, Bozonet S, Dyer A, Robinson BA, Dachs GU. Restoring physiological levels of ascorbate slows tumor growth and moderates HIF-1 pathway activity in Gulo(-/-) mice. Cancer Med. 2015;4(2):303-14.
    http://onlinelibrary.wiley.com/doi/10.1002/cam4.349/full
  212. Chen Q, Espey MG, Sun AY, et al. Pharmacologic doses of ascorbate act as a prooxidant and decrease growth of aggressive tumor xenografts in mice. Proc Natl Acad Sci USA. 2008;105(32):11105-9.
    https://www.ncbi.nlm.nih.gov/pubmed/18678913
  213. Takemura Y, Satoh M, Satoh K, Hamada H, Sekido Y, Kubota S. High dose of ascorbic acid induces cell death in mesothelioma cells. Biochem Biophys Res Commun. 2010;394(2):249-53.
    https://www.sciencedirect.com/science/article/pii/S0006291X10002123
  214. Yun J, Mullarky E, Lu C, et al. Vitamin C selectively kills KRAS and BRAF mutant colorectal cancer cells by targeting GAPDH. Science. 2015;350(6266):1391-6.
    https://www.ncbi.nlm.nih.gov/pubmed/26541605
  215. Yeom CH, Lee G, Park JH, et al. High dose concentration administration of ascorbic acid inhibits tumor growth in BALB/C mice implanted with sarcoma 180 cancer cells via the restriction of angiogenesis. J Transl Med. 2009;7:70.
    http://europepmc.org/articles/PMC2732919
  216. Casciari JJ, Riordan HD, Miranda-massari JR, Gonzalez MJ. Effects of high dose ascorbate administration on L-10 tumor growth in guinea pigs. P R Health Sci J. 2005;24(2):145-50.
    http://prhsj.rcm.upr.edu/index.php/prhsj/article/view/384
  217. Roomi MW, Cha J, Kalinovsky T, Roomi N, Niedzwiecki A, Rath M. Effect of a nutrient mixture on the localization of extracellular matrix proteins in HeLa human cervical cancer xenografts in female nude mice. Exp Ther Med. 2015;10(3):901-906.
    https://www.drrathresearch.org/attachments/Cancer/Cervical%20Cancer/HeLa-2-pub.pdf
  218. Ambattu LA, Rekha MR. Collagen synthesis promoting pullulan-PEI-ascorbic acid conjugate as an efficient anti-cancer gene delivery vector. Carbohydr Polym. 2015;126:52-61.
    https://www.sciencedirect.com/science/article/pii/S014486171500226X
  219. Güney G, Kutlu HM, Genç L. Preparation and characterization of ascorbic acid loaded solid lipid nanoparticles and investigation of their apoptotic effects. Colloids Surf B Biointerfaces. 2014;121:270-80.
    https://www.sciencedirect.com/science/article/pii/S0927776514002343
  220. Gustafson CB, Yang C, Dickson KM, et al. Epigenetic reprogramming of melanoma cells by vitamin C treatment. Clin Epigenetics. 2015;7(1):51.
    https://clinicalepigeneticsjournal.biomedcentral.com/articles/10.1186/s13148-015-0087-z
  221. Robinson AB, Hunsberger A, Westall FC. Suppression of squamous cell carcinoma in hairless mice by dietary nutrient variation. Mech Ageing Dev. 1994;76(2-3):201-14.
    https://www.sciencedirect.com/science/article/pii/0047637494915946
  222. Cameron E, Pauling L. Supplemental ascorbate in the supportive treatment of cancer: Prolongation of survival times in terminal human cancer. Proc Natl Acad Sci USA. 1976;73(10):3685-9.
    https://www.ncbi.nlm.nih.gov/pubmed/1068480
  223. Raymond YC, Glenda CS, Meng LK. Effects of High Doses of Vitamin C on Cancer Patients in Singapore: Nine Cases. Integr Cancer Ther. 2015;
    http://journals.sagepub.com/doi/pdf/10.1177/1534735415622010
  224. Vollbracht C, Schneider B, Leendert V, Weiss G, Auerbach L, Beuth J. Intravenous vitamin C administration improves quality of life in breast cancer patients during chemo-/radiotherapy and aftercare: results of a retrospective, multicentre, epidemiological cohort study in Germany. In Vivo. 2011;25(6):983-90.
    http://iv.iiarjournals.org/content/25/6/983.long
  225. Yeom CH, Jung GC, Song KJ. Changes of terminal cancer patients’ health-related quality of life after high dose vitamin C administration. J Korean Med Sci. 2007;22(1):7-11.
    https://www.ncbi.nlm.nih.gov/pubmed/17297243
  226. Mikirova N, Casciari J, Rogers A, Taylor P. Effect of high-dose intravenous vitamin C on inflammation in cancer patients. J Transl Med. 2012;10:189.
    https://www.ncbi.nlm.nih.gov/pubmed/22963460
  227. Mateen S, Moin S, Khan AQ, Zafar A, Fatima N. Increased Reactive Oxygen Species Formation and Oxidative Stress in Rheumatoid Arthritis. PLoS ONE. 2016;11(4):e0152925.
    https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4820274/
  228. Mohamed R, Shivaprasad HV, Jameel NM, Shekar MA, Vishwanath BS. Neutralization of local toxicity induced by vipera russelli phospholipase A2 by lipophilic derivative of ascorbic acid. Curr Top Med Chem. 2011;11(20):2531-9.
    https://www.ncbi.nlm.nih.gov/pubmed/21682683
  229. Laing MD. A cure for mushroom poisoning. S Afr Med J. 1984;65(15):590.
    https://www.ncbi.nlm.nih.gov/pubmed/6200941
  230. Pavlovic V, Cekic S, Kamenov B, Ciric M, Krtinic D. The Effect of Ascorbic Acid on Mancozeb-Induced Toxicity in Rat Thymocytes. Folia Biol (Praha). 2015;61(3):116-23.
    https://www.researchgate.net/publication/281819121_The_Effect_of_Ascorbic_Acid_on_Mancozeb-Induced_Toxicity_in_Rat_Thymocytes
  231. Ozmen O. Endosulfan splenic pathology and amelioration by vitamin C in New Zealand rabbit. J Immunotoxicol. 2015;:1-6.
    http://www.tandfonline.com/doi/pdf/10.3109/1547691X.2015.1095825
  232. Guo W, Huen K, Park JS, et al. Vitamin C intervention may lower the levels of persistent organic pollutants in blood of healthy women – A pilot study. Food Chem Toxicol. 2016;92:197-204.
    https://www.sciencedirect.com/science/article/pii/S0278691516301120
  233. Roderique JD, Josef CS, Newcomb AH, Reynolds PS, Somera LG, Spiess BD. Preclinical evaluation of injectable reduced hydroxocobalamin as an antidote to acute carbon monoxide poisoning. J Trauma Acute Care Surg. 2015;79(4 Suppl 2):S116-20.
    http://europepmc.org/abstract/MED/26406423
  234. Abe S, Tanaka Y, Fujise N, et al. An antioxidative nutrient-rich enteral diet attenuates lethal activity and oxidative stress induced by lipopolysaccharide in mice. JPEN J Parenter Enteral Nutr. 2007;31(3):181-7.
    http://journals.sagepub.com/doi/abs/10.1177/0148607107031003181
  235. Berger MM, Oudemans-van straaten HM. Vitamin C supplementation in the critically ill patient. Curr Opin Clin Nutr Metab Care. 2015;18(2):193-201.
    https://www.ncbi.nlm.nih.gov/pubmed/25635594
  236. Arslan M, Sezen SC, Turgut HC, et al. Vitamin C ameliorates high dose Dexmedetomidine induced liver injury. Bratisl Lek Listy. 2016;117(1):36-40.
    https://www.ncbi.nlm.nih.gov/pubmed/26810168
  237. Farombi EO, Onyema OO. Monosodium glutamate-induced oxidative damage and genotoxicity in the rat: modulatory role of vitamin C, vitamin E and quercetin. Hum Exp Toxicol. 2006;25(5):251-9.
    https://www.ncbi.nlm.nih.gov/pubmed/16758767
  238. Mozhdeganloo Z, Jafari AM, Koohi MK, Heidarpour M. Methylmercury-induced oxidative stress in rainbow trout (Oncorhynchus mykiss) liver: ameliorating effect of vitamin C. Biol Trace Elem Res. 2015;165(1):103-9.
    https://link.springer.com/article/10.1007/s12011-015-0241-7
  239. Gulec M, Gurel A, Armutcu F. Vitamin E protects against oxidative damage caused by formaldehyde in the liver and plasma of rats. Mol Cell Biochem. 2006;290(1-2):61-7.
    https://www.ncbi.nlm.nih.gov/pubmed/16937016
  240. Rezvanjoo B, Rashidi S, Jouyban A, Beheshtiha SH, Samini M. Effects of vitamin C and melatonin on cysteamine-induced duodenal ulcer in a cholestatic rat model: A controlled experimental study. Curr Ther Res Clin Exp. 2010;71(5):322-30.
    https://www.sciencedirect.com/science/article/pii/S0011393X10000779
  241. Kim SR, Ha YM, Kim YM, et al. Ascorbic acid reduces HMGB1 secretion in lipopolysaccharide-activated RAW 264.7 cells and improves survival rate in septic mice by activation of Nrf2/HO-1 signals. Biochem Pharmacol. 2015;95(4):279-89.
    https://www.sciencedirect.com/science/article/pii/S0006295215002002
  242. Tokuda Y, Miura N, Kobayashi M, et al. Ascorbic acid deficiency increases endotoxin influx to portal blood and liver inflammatory gene expressions in ODS rats. 2015;31(2):373-9.
    https://www.sciencedirect.com/science/article/pii/S0899900714003487
  243. Fisher BJ, Kraskauskas D, Martin EJ, et al. Attenuation of sepsis-induced organ injury in mice by vitamin C. JPEN J Parenter Enteral Nutr. 2014;38(7):825-39.
    http://journals.sagepub.com/doi/abs/10.1177/0148607113497760
  244. Abhilash PA, Harikrishnan R, Indira M. Ascorbic acid suppresses endotoxemia and NF-κB signaling cascade in alcoholic liver fibrosis in guinea pigs: a mechanistic approach. Toxicol Appl Pharmacol. 2014;274(2):215-24.
    https://www.sciencedirect.com/science/article/pii/S0041008X13004912
  245. Lowes DA, Webster NR, Galley HF. Dehydroascorbic acid as pre-conditioner: protection from lipopolysaccharide induced mitochondrial damage. Free Radic Res. 2010;44(3):283-92.
    https://www.ncbi.nlm.nih.gov/pubmed/20166893
  246. Fisher BJ, Seropian IM, Kraskauskas D, et al. Ascorbic acid attenuates lipopolysaccharide-induced acute lung injury. Crit Care Med. 2011;39(6):1454-60.
    https://www.ncbi.nlm.nih.gov/pubmed/21358394
  247. Mckinnon RL, Lidington D, Tyml K. Ascorbate inhibits reduced arteriolar conducted vasoconstriction in septic mouse cremaster muscle. Microcirculation. 2007;14(7):697-707.
    http://onlinelibrary.wiley.com/doi/10.1080/10739680701410389/abstract
  248. Kanter M, Coskun O, Armutcu F, Uz YH, Kizilay G. Protective effects of vitamin C, alone or in combination with vitamin A, on endotoxin-induced oxidative renal tissue damage in rats. Tohoku J Exp Med. 2005;206(2):155-62.
    https://www.ncbi.nlm.nih.gov/pubmed/15888972
  249. Chen MF, Yang CM, Su CM, Hu ML. Vitamin C protects against cisplatin-induced nephrotoxicity and damage without reducing its effectiveness in C57BL/6 mice xenografted with Lewis lung carcinoma. Nutr Cancer. 2014;66(7):1085-91.
    http://www.tandfonline.com/doi/abs/10.1080/01635581.2014.948211
  250. Al-asmari AK, Khan AQ, Al-masri N. Mitigation of 5-fluorouracil-induced liver damage in rats by vitamin C via targeting redox-sensitive transcription factors. Hum Exp Toxicol. 2016;
    http://journals.sagepub.com/doi/abs/10.1177/0960327115626583?journalCode=hetb
  251. Peng LJ, Lu DX, Qi RB, Zhang T, Wang Z, Sun Y. [Therapeutic effect of intravenous high-dose vitamin C on implanted hepatoma in rats]. Nan Fang Yi Ke Da Xue Xue Bao. 2009;29(2):264-6.
    https://www.ncbi.nlm.nih.gov/pubmed/19246295
  252. Cheng LL, Liu YY, Li B, Li SY, Ran PX. [An in vitro study on the pharmacological ascorbate treatment of influenza virus]. Zhonghua Jie He He Hu Xi Za Zhi. 2012;35(7):520-3.
    https://www.pubfacts.com/detail/22931805/An-in-vitro-study-on-the-pharmacological-ascorbate-treatment-of-influenza-virus
  253. Wintergerst ES, Maggini S, Hornig DH. Immune-enhancing role of vitamin C and zinc and effect on clinical conditions. Ann Nutr Metab. 2006;50(2):85-94.
    https://www.karger.com/Article/Abstract/90495
  254. Harakeh S, Jariwalla RJ. Comparative study of the anti-HIV activities of ascorbate and thiol-containing reducing agents in chronically HIV-infected cells. Am J Clin Nutr. 1991;54(6 Suppl):1231S-1235S.
    http://ajcn.nutrition.org/content/54/6/1231S.short?cited-by=yesl54/6/1231Sr54/6/1231S
  255. Dalton WL. Massive Doses of Vitamin C in the Treatment of Viral Diseases. Journal of the Indiana State Medical Association. 1962.
    https://www.ncbi.nlm.nih.gov/pubmed/13883259
  256. Riordan Clinic. High-dose Intravenous Vitamin C as a Successful Treatment of Viral Infections. [Available] https://riordanclinic.org/2014/02/high-dose-intravenous-vitamin-c-as-a-successful-treatment-of-viral-infections [May 11, 2016].
  257. Mikirova N, Hunninghake R. Effect of high dose vitamin C on Epstein-Barr viral infection. Med Sci Monit. 2014;20:725-32.
    https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4015650/
  258. Jahan K, Ahmad K, Ali MA. Effect of ascorbic acid in the treatment of tetanus. Bangladesh Med Res Counc Bull. 1984;10(1):24-8.
    https://www.ncbi.nlm.nih.gov/pubmed/6466264
  259. Hemilä H, Koivula TT. Vitamin C for preventing and treating tetanus. Cochrane Database Syst Rev. 2008;(2):CD006665.
    https://www.ncbi.nlm.nih.gov/pubmed/24226506
  260. Gonzalez MJ, Miranda-massari JR, Berdiel MJ, et al. High Dose Intraveneous Vitamin C and Chikungunya Fever: A Case Report. J Orthomol Med. 2014;29(4):154-156.
    http://europepmc.org/articles/PMC4335641
  261. Chin KY, Ima-nirwana S. Vitamin C and Bone Health: Evidence from Cell, Animal and Human Studies. Curr Drug Targets. 2015;
    http://europepmc.org/abstract/MED/26343111
  262. Kim YA, Kim KM, Lim S, et al. Favorable effect of dietary vitamin C on bone mineral density in postmenopausal women (KNHANES IV, 2009): discrepancies regarding skeletal sites, age, and vitamin D status. Osteoporos Int. 2015;26(9):2329-37.
    http://press.endocrine.org/doi/abs/10.1210/endo-meetings.2014.BCHVD.16.SAT-0263
  263. Hart A, Cota A, Makhdom A, Harvey EJ. The Role of Vitamin C in Orthopedic Trauma and Bone Health. Am J Orthop. 2015;44(7):306-11.
    https://www.ncbi.nlm.nih.gov/pubmed/26161758
  264. Sun LL, Li BL, Xie HL, et al. Associations between the dietary intake of antioxidant nutrients and the risk of hip fracture in elderly Chinese: a case-control study. Br J Nutr. 2014;112(10):1706-14.
    https://www.ncbi.nlm.nih.gov/pubmed/25287150
  265. Zhu LL, Cao J, Sun M, et al. Vitamin C prevents hypogonadal bone loss. PLoS ONE. 2012;7(10):e47058.
    http://journals.plos.org/plosone/article?id=10.1371/journal.pone.0047058
  266. Torbergsen AC, Watne LO, Wyller TB, et al. Micronutrients and the risk of hip fracture: Case-control study. Clin Nutr. 2015.
    https://www.sciencedirect.com/science/article/pii/S0261561415003520
  267. Huang YN, Yang LY, Wang JY, Lai CC, Chiu CT, Wang JY. L-Ascorbate Protects Against Methamphetamine-Induced Neurotoxicity of Cortical Cells via Inhibiting Oxidative Stress, Autophagy, and Apoptosis. Mol Neurobiol. 2016.
    https://link.springer.com/article/10.1007/s12035-015-9561-z
  268. Ozkan F, Gündüz SG, Berköz M, Hunt AO, Yalın S. The protective role of ascorbic acid (vitamin C) against chlorpyrifos-induced oxidative stress in Oreochromis niloticus. Fish Physiol Biochem. 2012;38(3):635-43.
    https://link.springer.com/article/10.1007/s10695-011-9544-6
  269. Altuntas I, Delibas N, Sutcu R. The effects of organophosphate insecticide methidathion on lipid peroxidation and anti-oxidant enzymes in rat erythrocytes: role of vitamins E and C. Hum Exp Toxicol. 2002;21(12):681-5.
    http://journals.sagepub.com/doi/abs/10.1191/0960327102ht304oa?journalCode=hetb
  270. Shah SA, Yoon GH, Kim HO, Kim MO. Vitamin C neuroprotection against dose-dependent glutamate-induced neurodegeneration in the postnatal brain. Neurochem Res. 2015;40(5):875-84.
    https://www.ncbi.nlm.nih.gov/pubmed/25701025
  271. Warner TA, Kang JQ, Kennard JA, Harrison FE. Low brain ascorbic acid increases susceptibility to seizures in mouse models of decreased brain ascorbic acid transport and Alzheimer’s disease. Epilepsy Res. 2015;110:20-5.
    https://www.sciencedirect.com/science/article/pii/S0920121114003337
  272. Ide K, Yamada H, Umegaki K, et al. Lymphocyte vitamin C levels as potential biomarker for progression of Parkinson’s disease. Nutrition. 2015;31(2):406-8.
    https://www.ncbi.nlm.nih.gov/pubmed/25592020
  273. Schjoldager JG, Paidi MD, Lindblad MM, et al. Maternal vitamin C deficiency during pregnancy results in transient fetal and placental growth retardation in guinea pigs. Eur J Nutr. 2015;54(4):667-76.
    https://www.ncbi.nlm.nih.gov/pubmed/25472559
  274. Rafiee B, Morowvat MH, Rahimi-ghalati N. Comparing the Effectiveness of Dietary Vitamin C and Exercise Interventions on Fertility Parameters in Normal Obese Men. Urol J. 2016;13(2):2635-9.
    https://www.researchgate.net/publication/301354228_Comparing_the_Effectiveness_of_Dietary_Vitamin_C_and_Exercise_Interventions_on_Fertility_Parameters_in_Normal_Obese_Men
  275. Vijayprasad S, Bb G, Bb N. Effect of vitamin C on male fertility in rats subjected to forced swimming stress. J Clin Diagn Res. 2014;8(7):HC05-8.
    https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4149087/
  276. De oliveira IJ, De souza VV, Motta V, Da-silva SL. Effects of Oral Vitamin C Supplementation on Anxiety in Students: A Double-Blind, Randomized, Placebo-Controlled Trial. Pak J Biol Sci. 2015;18(1):11-8.
    https://www.ncbi.nlm.nih.gov/pubmed/26353411
  277. Ebuehi OA, Ogedegbe RA, Ebuehi OM. Oral administration of vitamin C and vitamin E ameliorates lead-induced hepatotoxicity and oxidative stress in the rat brain. Nig Q J Hosp Med. 2012;22(2):85-90.
    https://www.readbyqxmd.com/read/23175903/oral-administration-of-vitamin-c-and-vitamin-e-ameliorates-lead-induced-hepatotoxicity-and-oxidative-stress-in-the-rat-brain
  278. Tabatabaei-malazy O, Nikfar S, Larijani B, Abdollahi M. Influence of ascorbic acid supplementation on type 2 diabetes mellitus in observational and randomized controlled trials; a systematic review with meta-analysis. J Pharm Pharm Sci. 2014;17(4):554-82.
    https://journals.library.ualberta.ca/jpps/index.php/JPPS/article/view/23408
  279. Maged AM, Torky H, Fouad MA, et al. Role of antioxidants in gestational diabetes mellitus and relation to fetal outcome: a randomized controlled trial. J Matern Fetal Neonatal Med. 2016;:1-6.
    http://www.tandfonline.com/doi/abs/10.3109/14767058.2016.1154526?journalCode=ijmf20
  280. Mason SA, Della gatta PA, Snow RJ, Russell AP, Wadley GD. Ascorbic acid supplementation improves skeletal muscle oxidative stress and insulin sensitivity in people with type 2 diabetes: Findings of a randomized controlled study. Free Radic Biol Med. 2016;93:227-38.
    https://www.sciencedirect.com/science/article/pii/S0891584916000071
  281. Kim HJ, Song W, Jin EH, et al. Combined Low-Intensity Exercise and Ascorbic Acid Attenuates Kainic Acid-Induced Seizure and Oxidative Stress in Mice. Neurochem Res. 2016;41(5):1035-41.
    https://link.springer.com/article/10.1007/s11064-015-1789-5
  282. Tutkun E, Arslan G, Soslu R, Ayyildiz M, Agar E. Long-term ascorbic acid administration causes anticonvulsant activity during moderate and long-duration swimming exercise in experimental epilepsy. Acta Neurobiol Exp (Wars). 2015;75(2):192-9.
    https://www.researchgate.net/publication/281817469_Long-term_ascorbic_acid_administration_causes_anticonvulsant_activity_during_moderate_and_long-duration_swimming_exercise_in_experimental_epilepsy
  283. Richards JC, Crecelius AR, Larson DG, Dinenno FA. Acute ascorbic acid ingestion increases skeletal muscle blood flow and oxygen consumption via local vasodilation during graded handgrip exercise in older adults. Am J Physiol Heart Circ Physiol. 2015;309(2):H360-8.
    http://www.physiology.org/doi/abs/10.1152/ajpheart.00209.2015
  284. Chayasirisobhon S. Efficacy of Pinus radiata bark extract and vitamin C combination product as a prophylactic therapy for recalcitrant migraine and long-term results. Acta Neurol Taiwan. 2013;22(1):13-21.
    https://www.ncbi.nlm.nih.gov/pubmed/23479241
  285. Kim JE, Cho HS, Yang HS, et al. Depletion of ascorbic acid impairs NK cell activity against ovarian cancer in a mouse model. Immunobiology. 2012;217(9):873-81.
    https://www.sciencedirect.com/science/article/pii/S0171298511002865
  286. Li K, Wang J, Shi M, et al. Prescription consisting of Vitamin C and Baicalin inhibits tumor growth by enhancing the antioxidant capacity in vivo. J BUON. 2015;20(5):1368-72.
    http://europepmc.org/abstract/MED/26537087
  287. Maggini S, Wenzlaff S, Hornig D. Essential role of vitamin C and zinc in child immunity and health. J Int Med Res. 2010;38(2):386-414.
    http://journals.sagepub.com/doi/pdf/10.1177/147323001003800203
  288. Ströhle A, Hahn A. [Vitamin C and immune function]. Med Monatsschr Pharm. 2009;32(2):49-54.
    https://www.ncbi.nlm.nih.gov/pubmed/19263912
  289. Huijskens MJ, Walczak M, Sarkar S, et al. Ascorbic acid promotes proliferation of natural killer cell populations in culture systems applicable for natural killer cell therapy. Cytotherapy. 2015;17(5):613-20.
    https://www.sciencedirect.com/science/article/pii/S1465324915000183
  290. Toliopoulos IK, Simos YV, Daskalou TA, Verginadis II, Evangelou AM, Karkabounas SC. Inhibition of platelet aggregation and immunomodulation of NK lymphocytes by administration of ascorbic acid. Indian J Exp Biol. 2011;49(12):904-8.
    https://www.pubfacts.com/detail/22403863/Inhibition-of-platelet-aggregation-and-immunomodulation-of-NK-lymphocytes-by-administration-of-ascor
  291. Atasever B, Ertan NZ, Erdem-kuruca S, Karakas Z. In vitro effects of vitamin C and selenium on NK activity of patients with beta-thalassemia major. Pediatr Hematol Oncol. 2006;23(3):187-97.
    https://www.ncbi.nlm.nih.gov/pubmed/16517535
  292. Kim JE, Cho HS, Yang HS, et al. Depletion of ascorbic acid impairs NK cell activity against ovarian cancer in a mouse model. Immunobiology. 2012;217(9):873-81.
    https://www.sciencedirect.com/science/article/pii/S0171298511002865
  293. Choi MK, Song HJ, Paek YJ, Lee HJ. Gender differences in the relationship between vitamin C and abdominal obesity. Int J Vitam Nutr Res. 2013;83(6):377-84.
    https://econtent.hogrefe.com/doi/abs/10.1024/0300-9831/a000179
  294. Adaramoye O, Ogungbenro B, Anyaegbu O, Fafunso M. Protective effects of extracts of Vernonia amygdalina, Hibiscus sabdariffa and vitamin C against radiation-induced liver damage in rats. J Radiat Res. 2008;49(2):123-31.
    https://www.ncbi.nlm.nih.gov/pubmed/18250564
  295. Vasilyeval IN, Bespalov VG. [Release of Extracellular DNA after Administration of Radioprotective Combination of α-Tocopherol and Ascorbic Acid]. Radiats Biol Radioecol. 2015;55(5):495-500.
    https://www.ncbi.nlm.nih.gov/pubmed/26863779
  296. Rostami A, Moosavi SA, Dianat moghadam H, Bolookat ER. Micronuclei Assessment of The Radioprotective Effects of Melatonin and Vitamin C in Human Lymphocytes. Cell J. 2016;18(1):46-51.
    https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4819385/
  297. Mortazavi SM, Rahimi S, Mosleh-shirazi MA, et al. A Comparative Study on the Life-Saving Radioprotective Effects of Vitamins A, E, C and Over-the-Counter Multivitamins. J Biomed Phys Eng. 2015;5(2):59-66.
    http://europepmc.org/articles/PMC4479387
  298. Domina EA, Pylypchuk OP, Mikhailenko VM. Destabilization of human cell genome under the combined effect of radiation and ascorbic acid. Exp Oncol. 2014;36(4):236-40.
    https://www.ncbi.nlm.nih.gov/pubmed/25537216
  299. Fujii Y, Kato TA, Ueno A, Kubota N, Fujimori A, Okayasu R. Ascorbic acid gives different protective effects in human cells exposed to X-rays and heavy ions. Mutat Res. 2010;699(1-2):58-61.
    https://www.sciencedirect.com/science/article/pii/S1383571810001269
  300. Mhaidat NM, Alzoubi KH, Khabour OF, Tashtoush NH, Banihani SA, Abdul-razzak KK. Exploring the effect of vitamin C on sleep deprivation induced memory impairment. Brain Res Bull. 2015;113:41-7.
    https://www.sciencedirect.com/science/article/pii/S0361923015000325
  301. Mohammed BM, Fisher BJ, Kraskauskas D, et al. Vitamin C promotes wound healing through novel pleiotropic mechanisms. Int Wound J. 2015;
    http://onlinelibrary.wiley.com/doi/10.1111/iwj.12484/full
  302. Murray EL. Burning mouth syndrome response to high-dose vitamin C. Headache. 2014;54(1):169.
    http://onlinelibrary.wiley.com/doi/10.1111/head.12209/abstract
  303. Dawood MA, Koshio S, Ishikawa M, Yokoyama S. Immune responses and stress resistance in red sea bream, Pagrus major, after oral administration of heat-killed Lactobacillus plantarum and vitamin C. Fish Shellfish Immunol. 2016;54:266-275.
    https://www.ncbi.nlm.nih.gov/pubmed/27095173
  304. Sadeghpour A, Alizadehasl A, Kyavar M, et al. Impact of vitamin C supplementation on post-cardiac surgery ICU and hospital length of stay. Anesth Pain Med. 2015;5(1):e25337.
    http://europepmc.org/articles/PMC4350190
  305. Yang M, Barak OF, Dujic Z, et al. Ascorbic acid supplementation diminishes microparticle elevations and neutrophil activation following SCUBA diving. Am J Physiol Regul Integr Comp Physiol. 2015;309(4):R338-44.
    http://www.greenmedinfo.com/article/ascorbic-acid-supplementation-diminishes-microparticle-elevations-and
  306. Besse JL, Gadeyne S, Galand-desmé S, Lerat JL, Moyen B. Effect of vitamin C on prevention of complex regional pain syndrome type I in foot and ankle surgery. Foot Ankle Surg. 2009;15(4):179-82.
    https://www.sciencedirect.com/science/article/pii/S1268773109000265
  307. Lane DJ, Richardson DR. The active role of vitamin C in mammalian iron metabolism: much more than just enhanced iron absorption!. Free Radic Biol Med. 2014;75:69-83.
    https://www.sciencedirect.com/science/article/pii/S0891584914003220
  308. May JM, Qu ZC, Mendiratta S. Role of ascorbic acid in transferrin-independent reduction and uptake of iron by U-937 cells. Biochem Pharmacol. 1999;57(11):1275-82.
    https://www.sciencedirect.com/science/article/pii/S0006295299000404
  309. Lane DJ, Chikhani S, Richardson V, Richardson DR. Transferrin iron uptake is stimulated by ascorbate via an intracellular reductive mechanism. Biochim Biophys Acta. 2013;1833(6):1527-41.
    https://www.sciencedirect.com/science/article/pii/S0167488913000682
  310. Mojić M, Bogdanović pristov J, Maksimović-ivanić D, et al. Extracellular iron diminishes anticancer effects of vitamin C: an in vitro study. Sci Rep. 2014;4:5955.
    https://www.nature.com/articles/srep05955
  311. Angelucci E, Pilo F. Management of iron overload before, during, and after hematopoietic stem cell transplantation for thalassemia major. Ann N Y Acad Sci. 2016;
    http://onlinelibrary.wiley.com/doi/10.1111/nyas.13027/abstract
  312. Grenier D, Huot MP, Mayrand D. Iron-chelating activity of tetracyclines and its impact on the susceptibility of Actinobacillus actinomycetemcomitans to these antibiotics. Antimicrob Agents Chemother. 2000;44(3):763-6.
    http://aac.asm.org/content/44/3/763.full
  313. National Institutes of Health. (2008). Vitamin C Injections Slow Tumor Growth in Mice. [Available] https://www.nih.gov/news-events/news-releases/vitamin-c-injections-slow-tumor-growth-mice  May 11, 2016].
  314. Hickey, S., & Saul, A. W. (2008). Vitamin C: The real story: The remarkable and controversial healing factor. Laguna Beach, CA: Basic Health Publications.
    http://amzn.to/2Dp5Ir9
  315. Jackson CL, Dreaden TM, Theobald LK, et al. Pectin induces apoptosis in human prostate cancer cells: correlation of apoptotic function with pectin structure. Glycobiology. 2007;17(8):805-19.
    http://paperity.org/p/40612166/pectin-induces-apoptosis-in-human-prostate-cancer-cells-correlation-of-apoptotic-function
  316. Zhang L, Ye X, Xue SJ, et al. Effect of high-intensity ultrasound on the physicochemical properties and nanostructure of citrus pectin. J Sci Food Agric. 2013;93(8):2028-36.
    http://onlinelibrary.wiley.com/doi/10.1002/jsfa.6011/abstract
  317. Leclere L, Fransolet M, Cambier P, et al. Identification of a cytotoxic molecule in heat-modified citrus pectin. Carbohydr Polym. 2016;137:39-51.
    https://www.sciencedirect.com/science/article/pii/S0144861715010358
  318. Glinsky VV, Raz A. Modified citrus pectin anti-metastatic properties: one bullet, multiple targets. Carbohydr Res. 2009;344(14):1788-91.
    https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2782490/
  319. Huang ZL, Liu HY. [Expression of galectin-3 in liver metastasis of colon cancer and the inhibitory effect of modified citrus pectin]. Nan Fang Yi Ke Da Xue Xue Bao. 2008;28(8):1358-61.
    https://www.medscape.com/medline/abstract/18753060
  320. Hsieh TC, Wu JM. Changes in cell growth, cyclin/kinase, endogenous phosphoproteins and nm23 gene expression in human prostatic JCA-1 cells treated with modified citrus pectin. Biochem Mol Biol Int. 1995;37(5):833-41.
    https://www.ncbi.nlm.nih.gov/pubmed/8624488
  321. Leclere L, Fransolet M, Cote F, et al. Heat-modified citrus pectin induces apoptosis-like cell death and autophagy in HepG2 and A549 cancer cells. PLoS ONE. 2015;10(3):e0115831.
    http://journals.plos.org/plosone/article?id=10.1371/journal.pone.0115831
  322. Yan J, Katz A. PectaSol-C modified citrus pectin induces apoptosis and inhibition of proliferation in human and mouse androgen-dependent and- independent prostate cancer cells. Integr Cancer Ther. 2010;9(2):197-203.
    https://www.ncbi.nlm.nih.gov/pubmed/20462856
  323. Hao M, Yuan X, Cheng H, et al. Comparative studies on the anti-tumor activities of high temperature- and pH-modified citrus pectins. Food Funct. 2013;4(6):960-71.
    http://pubs.rsc.org/en/content/articlelanding/2013/fo/c3fo30350k#!
  324. Jiang J, Eliaz I, Sliva D. Synergistic and additive effects of modified citrus pectin with two polybotanical compounds, in the suppression of invasive behavior of human breast and prostate cancer cells. Integr Cancer Ther. 2013;12(2):145-52.
    https://www.ncbi.nlm.nih.gov/pubmed/22532035
  325. Wang Y, Nangia-makker P, Balan V, Hogan V, Raz A. Calpain activation through galectin-3 inhibition sensitizes prostate cancer cells to cisplatin treatment. Cell Death Dis. 2010;1:e101.
    https://www.nature.com/articles/cddis201079
  326. Hossein G, Keshavarz M, Ahmadi S, Naderi N. Synergistic effects of PectaSol-C modified citrus pectin an inhibitor of Galectin-3 and paclitaxel on apoptosis of human SKOV-3 ovarian cancer cells. Asian Pac J Cancer Prev. 2013;14(12):7561-8.
    https://www.ncbi.nlm.nih.gov/pubmed/24460334
  327. Inohara H, Raz A. Effects of natural complex carbohydrate (citrus pectin) on murine melanoma cell properties related to galectin-3 functions. Glycoconj J. 1994;11(6):527-32.
    https://www.researchgate.net/publication/15502020_Effects_of_natural_complex_carbohydrate_citrus_pectin_on_murine_melanoma_cell_properties_related_to_galectin_3_function
  328. Nangia-makker P, Hogan V, Honjo Y, et al. Inhibition of human cancer cell growth and metastasis in nude mice by oral intake of modified citrus pectin. J Natl Cancer Inst. 2002;94(24):1854-62.
    https://www.ncbi.nlm.nih.gov/pubmed/12488479
  329. Pienta KJ, Naik H, Akhtar A, et al. Inhibition of spontaneous metastasis in a rat prostate cancer model by oral administration of modified citrus pectin. J Natl Cancer Inst. 1995;87(5):348-53.
    https://academic.oup.com/jnci/article-abstract/87/5/348/916058
  330. Platt D, Raz A. Modulation of the lung colonization of B16-F1 melanoma cells by citrus pectin. J Natl Cancer Inst. 1992;84(6):438-42.
    https://academic.oup.com/jnci/article-abstract/84/6/438/1019001?redirectedFrom=fulltext
  331. Liu HY, Huang ZL, Yang GH, Lu WQ, Yu NR. Inhibitory effect of modified citrus pectin on liver metastases in a mouse colon cancer model. World J Gastroenterol. 2008;14(48):7386-91.
    http://europepmc.org/articles/PMC2778124
  332. Hayashi A, Gillen AC, Lott JR. Effects of daily oral administration of quercetin chalcone and modified citrus pectin on implanted colon-25 tumor growth in Balb-c mice. Altern Med Rev. 2000;5(6):546-52.
    http://www.chiro.org/nutrition/ABSTRACTS/Effects_of_Daily_Oral.shtml
  333. Ma Z, Han Q, Wang X, Ai Z, Zheng Y. Galectin-3 Inhibition Is Associated with Neuropathic Pain Attenuation after Peripheral Nerve Injury. PLoS ONE. 2016;11(2):e0148792.
    https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4752273/
  334. Abu-elsaad NM, Elkashef WF. Modified citrus pectin stops progression of liver fibrosis by inhibiting galectin-3 and inducing apoptosis of stellate cells. Can J Physiol Pharmacol. 2016;94(5):554-62.
    https://www.medscape.com/medline/abstract/27010252
  335. Kolatsi-joannou M, Price KL, Winyard PJ, Long DA. Modified citrus pectin reduces galectin-3 expression and disease severity in experimental acute kidney injury. PLoS ONE. 2011;6(4):e18683.
    http://journals.plos.org/plosone/article?id=10.1371/journal.pone.0018683
  336. Chen CH, Sheu MT, Chen TF, et al. Suppression of endotoxin-induced proinflammatory responses by citrus pectin through blocking LPS signaling pathways. Biochem Pharmacol. 2006;72(8):1001-9.
    https://www.sciencedirect.com/science/article/pii/S0006295206003984
  337. Arad U, Madar-balakirski N, Angel-korman A, et al. Galectin-3 is a sensor-regulator of toll-like receptor pathways in synovial fibroblasts. Cytokine. 2015;73(1):30-5.
    https://www.sciencedirect.com/science/article/pii/S1043466615000204
  338. Eliaz I, Hotchkiss AT, Fishman ML, Rode D. The effect of modified citrus pectin on urinary excretion of toxic elements. Phytother Res. 2006;20(10):859-64.
    http://onlinelibrary.wiley.com/doi/10.1002/ptr.1953/abstract
  339. Zhao ZY, Liang L, Fan X, et al. The role of modified citrus pectin as an effective chelator of lead in children hospitalized with toxic lead levels. Altern Ther Health Med. 2008;14(4):34-8.
    https://www.ncbi.nlm.nih.gov/pubmed/18616067
  340. Eliaz I, Weil E, Wilk B. Integrative medicine and the role of modified citrus pectin/alginates in heavy metal chelation and detoxification–five case reports. Forsch Komplementmed. 2007;14(6):358-64.
    https://www.ncbi.nlm.nih.gov/pubmed/18219211
  341. Vergaro G, Prud’homme M, Fazal L, et al. Inhibition of Galectin-3 Pathway Prevents Isoproterenol-Induced Left Ventricular Dysfunction and Fibrosis in Mice. Hypertension. 2016;67(3):606-12.
    http://hyper.ahajournals.org/content/67/3/606
  342. Martínez-martínez E, López-Ándres N, Jurado-lópez R, et al. Galectin-3 Participates in Cardiovascular Remodeling Associated With Obesity. Hypertension. 2015;66(5):961-9.
    https://www.researchgate.net/publication/281635758_Galectin-3_Participates_in_Cardiovascular_Remodeling_Associated_With_Obesity
  343. Martínez-martínez E, Calvier L, Fernández-celis A, et al. Galectin-3 blockade inhibits cardiac inflammation and fibrosis in experimental hyperaldosteronism and hypertension. Hypertension. 2015;66(4):767-75.
    http://hyper.ahajournals.org/content/early/2015/08/03/HYPERTENSIONAHA.115.05876
  344. Calvier L, Martinez-martinez E, Miana M, et al. The impact of galectin-3 inhibition on aldosterone-induced cardiac and renal injuries. JACC Heart Fail. 2015;3(1):59-67.
    https://www.sciencedirect.com/science/article/pii/S2213177914003886
  345. Mackinnon AC, Liu X, Hadoke PW, Miller MR, Newby DE, Sethi T. Inhibition of galectin-3 reduces atherosclerosis in apolipoprotein E-deficient mice. Glycobiology. 2013;23(6):654-63.
    https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3641797/
  346. Ramachandran C, Wilk BJ, Hotchkiss A, Chau H, Eliaz I, Melnick SJ. Activation of human T-helper/inducer cell, T-cytotoxic cell, B-cell, and natural killer (NK)-cells and induction of natural killer cell activity against K562 chronic myeloid leukemia cells with modified citrus pectin. BMC Complement Altern Med. 2011;11:59.
    https://bmccomplementalternmed.biomedcentral.com/articles/10.1186/1472-6882-11-59
  347. Martínez-martínez E, Calvier L, Rossignol P, et al. Galectin-3 inhibition prevents adipose tissue remodelling in obesity. Int J Obes (Lond). 2016;
    https://www.medscape.com/medline/abstract/26853916
  348. Dourado GK, Stanilka JM, Percival SS, Cesar TB. Chemopreventive Actions of Blond and Red-Fleshed Sweet Orange Juice on the Loucy Leukemia Cell Line. Asian Pac J Cancer Prev. 2015;16(15):6491-9.
    https://www.ncbi.nlm.nih.gov/pubmed/26434864/
  349. So FV, Guthrie N, Chambers AF, Moussa M, Carroll KK. Inhibition of human breast cancer cell proliferation and delay of mammary tumorigenesis by flavonoids and citrus juices. Nutr Cancer. 1996;26(2):167-81.
    http://www.tandfonline.com/doi/abs/10.1080/01635589609514473
  350. Tanaka T, Kohno H, Murakami M, et al. Suppression of azoxymethane-induced colon carcinogenesis in male F344 rats by mandarin juices rich in beta-cryptoxanthin and hesperidin. Int J Cancer. 2000;88(1):146-50.
    http://onlinelibrary.wiley.com/doi/10.1002/1097-0215(20001001)88:1%3C146::AID-IJC23%3E3.0.CO;2-I/full
  351. Miyagi Y, Om AS, Chee KM, Bennink MR. Inhibition of azoxymethane-induced colon cancer by orange juice. Nutr Cancer. 2000;36(2):224-9.
    https://www.ncbi.nlm.nih.gov/pubmed/10890034
  352. Tanaka T, Tanaka T, Tanaka M, Kuno T. Cancer chemoprevention by citrus pulp and juices containing high amounts of β-cryptoxanthin and hesperidin. J Biomed Biotechnol. 2012;2012:516981.
    https://www.hindawi.com/journals/bmri/2012/516981/ref/
  353. Oikeh EI, Omoregie ES, Oviasogie FE, Oriakhi K. Phytochemical, antimicrobial, and antioxidant activities of different citrus juice concentrates. Food Sci Nutr. 2016;4(1):103-9.
    https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4708628/
  354. Buscemi S, Rosafio G, Arcoleo G, et al. Effects of red orange juice intake on endothelial function and inflammatory markers in adult subjects with increased cardiovascular risk. Am J Clin Nutr. 2012;95(5):1089-95.
    https://www.ncbi.nlm.nih.gov/pubmed/22492368
  355. Ko SH, Choi SW, Ye SK, Cho BL, Kim HS, Chung MH. Comparison of the antioxidant activities of nine different fruits in human plasma. J Med Food. 2005;8(1):41-6.
    https://www.ncbi.nlm.nih.gov/pubmed/15857208
  356. Constans J, Bennetau-pelissero C, Martin JF, et al. Marked antioxidant effect of orange juice intake and its phytomicronutrients in a preliminary randomized cross-over trial on mild hypercholesterolemic men. Clin Nutr. 2015;34(6):1093-100.
    https://www.researchgate.net/publication/270053281_Marked_antioxidant_effect_of_orange_juice_intake_and_its_phytomicronutrients_in_a_preliminary_randomized_cross-over_trial_on_mild_hypercholesterolemic_men
  357. Lee SG, Yang M, Wang Y, et al. Impact of orange juice consumption on bone health of the U.S. population in the national health and nutrition examination survey 2003-2006. J Med Food. 2014;17(10):1142-50.
    https://www.ncbi.nlm.nih.gov/pubmed/22433835
  358. Deyhim F, Garica K, Lopez E, et al. Citrus juice modulates bone strength in male senescent rat model of osteoporosis. Nutrition. 2006;22(5):559-63.
    https://www.sciencedirect.com/science/article/pii/S0899900705003795
  359. Pittaluga M, Sgadari A, Tavazzi B, et al. Exercise-induced oxidative stress in elderly subjects: the effect of red orange supplementation on the biochemical and cellular response to a single bout of intense physical activity. Free Radic Res. 2013;47(3):202-11.
    https://www.ncbi.nlm.nih.gov/pubmed/23297807
  360. Aptekmann NP, Cesar TB. Orange juice improved lipid profile and blood lactate of overweight middle-aged women subjected to aerobic training. Maturitas. 2010;67(4):343-7.
    https://www.sciencedirect.com/science/article/pii/S0378512210003075
  361. Escudero-lópez B, Berná G, Ortega Á, et al. Consumption of orange fermented beverage reduces cardiovascular risk factors in healthy mice. Food Chem Toxicol. 2015;78:78-85.
    https://www.ncbi.nlm.nih.gov/pubmed/25666657
  362. Zanotti simoes dourado GK, De abreu ribeiro LC, Zeppone carlos I, Borges césar T. Orange juice and hesperidin promote differential innate immune response in macrophages ex vivo. Int J Vitam Nutr Res. 2013;83(3):162-7.
    http://europepmc.org/abstract/MED/24846905
  363. Wang Y, Lloyd B, Yang M, et al. Impact of orange juice consumption on macronutrient and energy intakes and body composition in the US population. Public Health Nutr. 2012;15(12):2220-7.
    https://www.ncbi.nlm.nih.gov/pubmed/22433835
  364. O’neil CE, Nicklas TA, Rampersaud GC, Fulgoni VL. 100% orange juice consumption is associated with better diet quality, improved nutrient adequacy, decreased risk for obesity, and improved biomarkers of health in adults: National Health and Nutrition Examination Survey, 2003-2006. Nutr J. 2012;11:107.
    https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3545988/
  365. Salamone F, Li volti G, Titta L, et al. Moro orange juice prevents fatty liver in mice. World J Gastroenterol. 2012;18(29):3862-8.
    https://www.wjgnet.com/1007-9327/full/v18/i29/3862.htm
  366. Azik M. Phytochemicals in citrus. Florida Department of Citrus. 2010. Available: http://fdocgrower.com/wp-content/uploads/2010/11/Flavonoids_Orange.pdf [February 5, 2017].
  367. Better Health Publishing. New Research: Modified Citrus Pectin – A potent anti-cancer therapy. PR Newswire. 2013.
    https://www.newswise.com/articles/new-research-modified-citrus-pectin-a-potent-anti-cancer-therapy
  368. Stone I. On the genetic etiology of scurvy. Acta Genet Med Gemellol (Roma). 1966;15(4):345-50.
    https://www.seanet.com/~alexs/ascorbate/196x/stone-i-acta_genet_med_et_gemell-1966-v15-p345.htm
  369. Jiao Y, Wilkinson J, Di X, et al. Curcumin, a cancer chemopreventive and chemotherapeutic agent, is a biologically active iron chelator. Blood. 2009;113(2):462-9.
    http://www.bloodjournal.org/content/113/2/462