Abstract
Good health and rapid progress depend on an optimal dose of nicotinamide. Too little meat triggers the neurodegenerative condition pellagra and tolerance of symbionts such as tuberculosis (TB), risking dysbioses and impaired resistance to acute infections. Nicotinamide deficiency is an overlooked diagnosis in poor cereal-dependant economies masquerading as ‘environmental enteropathy’ or physical and cognitive stunting. Too much meat (and supplements) may precipitate immune intolerance and autoimmune and allergic disease, with relative infertility and longevity, via the tryptophan-nicotinamide pathway. This switch favours a dearth of regulatory T (Treg) and an excess of T helper cells. High nicotinamide intake is implicated in cancer and Parkinson’s disease. Pro-fertility genes, evolved to counteract high-nicotinamide-induced infertility, may now be risk factors for degenerative disease. Moderation of the dose of nicotinamide could prevent some common diseases and personalised doses at times of stress or, depending on genetic background or age, may treat some other conditions.
Keywords: Tregs, pellagra, environmental enteropathy, TB, hypervitaminosis B3, immune intolerance, antagonistic pleiotropy, thrifty phenotype, disposable soma, cancer, Parkinson’s disease, Flynn effect, IQ, dementia
It is as if man had been appointed managing director of the biggest business of all, the business of evolution . . . the sooner he realizes it, the better.
Julian Huxley
Introduction
Moving down the food chain to a more plant-based diet with less nicotinamide encouraged fertility over the last 30 000 years or more.1 This led to population booms with benefits, but the Neolithic was also the crucible of disease and inter-group violence over resources. Signalling molecules from diet and microbiome, such as serotonin, played a part in settling down and domestication.2–8 Domestication and diet is fundamental to our recent evolution (even dogs changed their diet to domesticate).9–14 A pro-fertility diet and a pro-family communal child rearing culture saved us, we think, from extinction with ornamentation religion and the arts being survival and mating mechanisms, not ‘icing on the cake’, as was enlightened pro-social thinking, language, and writing.15–49
Cereal cultivation moved down first from the ‘hilly flanks’ to Mesopotamian riverside alluvial plains that allowed animal domestication but typically still needed steppe pastoralists to specialise in meat production with surpluses to trade for cereals.50–63 History can be seen in the light of a drive for an omnivorous diet whether trades, raids, or (civil) wars; many social relationships, belief systems, and institutions may be built on this essential infrastructure needed for reproduction. Overall progress was made when amalgamations occurred between agrarian farmers and pastoralists or where geography allowed mixed farming and a balanced diet in the first place.64–68 A high-meat high-cuisine diet however was often the preserve of the clever ruling classes and more recently the middle classes that expanded on ‘wheat and beef’ in the wealthier and usually Anglophone countries.69,70 This desire for meat continues against ecological opposition and climate concerns but may have good biological reasons.71,72
Nicotinamide adenine dinucleotide (NAD) can be synthesised from tryptophan via the kynurenine ‘immune tolerance’ pathway, but the preferred source is dietary nicotinamide mainly derived from animal products (Figure 1). Nicotinamide has a detoxification pathway via nicotinamide N-methyltransferase (NNMT) that links to methyl metabolism. Nicotinamide adenine dinucleotide consumers control metabolism and NAD sensors drive the quest for food and construction of a NAD world. There are important interactions with the immune system, but it is complex with some currently irreconcilable effects including on disease models depending on dose, metabolite, and even route of administration so we have had to simplify to support our hypothesis: although some contradictions may pertain to mixing ultimate and proximate causation and a system where resting immunologic state and secondary or even tertiary homeostatic reactions cannot always be easily separated.
Figure 1.
Nicotinamide switch. Higher doses shift the immune system from tolerance of infection to intolerance of antigens with consequences for both disease and fertility. Many modern therapies try to rebalance the immune system but moderation of the nicotinamide dose might have prevented the problem. AHR indicates aryl hydrocarbon receptor; BD, Behçet disease; IL, interleukin; MS, multiple sclerosis; NAD, nicotinamide adenine dinucleotide; RA, rheumatoid arthritis; SLE, systemic lupus erythematosus; TGF, transforming growth factor; TRYP, Tryptophan.
Our history is full of famines that did not affect fertility in the direction anticipated, except when extreme. We explain high fertility on poor diets by an interaction with tryptophan catabolism. Tolerance of the allogeneic foetus occurs by controlling indoleamine 2,3-dioxygenase (IDO) that also affects microbial survival and symbiont acceptance. Kynurenines modulate T cells.73–78 This pathway is conditioned by nicotinamide and is perhaps the basis for our constitution that, with our collaborative, sexual, and social natures, forms the mainspring for successful civilisations.79,80 Mismatched diets perhaps cause delayed demographic and exaggerated disease transitions, and friction. Dietary friction between sexes has been dated to the invention of ploughs relegating women to secondary producers of food (although women may, in fact, prefer a lower meat diet to aid reproduction).81–83
Meat as a Main Nicotinamide Source
Meat was key to our evolution with extensive meat sharing but also aggression with hustling and wars to obtain it, or the wherewithal to stock-breed whether land, water, or fossil fuels. Meat has, like cereals, been revered in our early cultures and gods, often with animal or human sacrifice, and hunting was the main subject of cave drawings. Meat has a divided literature with emotive titles such as ‘The Hunting Apes’ or ‘The Meat Crisis’.84–86 There is evidence for a genuine ‘meat hunger’ and that meat is not all about violence for violence’s sake, or status, or sexual preferment: vegetarian movements demonstrate the need for balance.87
Steppes up to the Plate
Reversion to hunter-gathering once an agricultural society was rare – it did occur on climatic edges (such as Norse Greenland) but more tellingly where populations were short of meat and prone to pellagra, as in the Americas, suggesting that the benefit-to-risk ratio of a cereal diet could be a close call and sometimes reverse.88,89 At the end of the Roman empire, settled agriculturalists defected to the pastoralist Huns suggesting that the drive for meat can contribute to the demise of empires. Many early trading arrangements were centred on pastoralist to agriculturalist ‘meat for cereal’ deals and trading routes that originated as herding pathways, such as the Silk Road. Meat intake has long been linked with success as individuals, as tribes, or as countries. Much of African history can be seen in this light with many other examples found in all other continents.90
Empires and Economic Divergences: Meat Eaters Win
Columbian and other exchanges and conquests: diet and ‘virgin soil’ disease
The Columbian exchange was an example of meat-eating Europeans conquering a maize-based low-meat culture with the winners having better technological brains and a better constitution to fight many infectious diseases. On other occasions, where immunity can develop, diseases can protect against ‘virgin’ colonialists as seen with yellow fever and malaria, but those with the better constitution still usually win.91–97 Animal diseases can alter dynamics – rinderpest or sleeping sickness in cattle aided the colonial acquisition of Africa and coincided with outbreaks of pellagra in the local population. It has been surprisingly difficult during our history to achieve even subsistence-level balanced diets so getting ahead of the meat curve may have been crucial for success – with more conventional explanations being secondary often necessary, but not sufficient, developments98–116 (Figure 2).
Figure 2.
A well-balanced diet is the base from which all else follows. Formulae for success had emphasised the necessity of the higher tiers, although without much agreement. Superstructure is however important as positive feedback loops further secure a high-quality diet.
Post Black Death
Most historical crises and cycles were famine followed by plague or war then a baby boom, then repeat. The more unusual beneficial long-term effects of the Black Death are attributed to increased pastoralism and availability of meat with higher wages from a reduced workforce. Agricultural developments helped as they benefitted the nitrogen cycle through crop rotation, ploughing and feeding animals in winter, and more use of animal by-products such as manure117,118 (Figure 3). This benign period petered out in the early 18th century due to climate change from volcanic activity, poor harvests, wars (impeding use of imports), and epizootic diseases of cattle. This decline in diet quality that became virtually vegetarian coincided with a spurt of population growth due to increases in fertility (as happened in the Neolithic).119–139 Even rich peers suffered so one can imagine how much life deteriorated for the poor making the increase in population even more remarkable140–161 (Figure 4). This state of affairs was mitigated later in the mid-19th-century United Kingdom by meat imports and by revised Corn Laws that enabled the price of grain to fall along with imperialistic acquisitions to avoid crises with civil war and food revolts. The concept (and German argument for their expansion) of ‘Lebenstraum’ – farming space – is generalisable with ‘Landrush’ phenomenon explaining many aspects of many European empires’ behaviour. Sometimes, flexing between agrarian and pastoralism is done peaceably but even within economies can cause friction as with the Scottish ‘clearances’ needed to feed meat to cities.162–164
Figure 3.
After the Black Death (triggered by famines), the population remained remarkably stable as did the supply of meat helped by increased wages. This period is generally agreed to have been a take-off for the industrial revolution.
Figure 4.

After the Black Death, the meat supply remained affordable and in step with the price of grain until around 1650. Then in the ‘little ice-age’ (perhaps triggered by population collapse in the new world) with harvest failures, and wars, meat became very expensive and with the exception of the wealthy the population became virtually vegetarian. Consistent with our hypothesis, fertility and population took off as did TB in the period just before the United Kingdom’s demographic transition around 1850 and the decline of TB as meat became more available, largely thanks to the wealth to pay for imports. TB indicates tuberculosis.
Lessons from America: common denominator is pellagra
America’s ‘King Cotton’ states and industrial North in the 19th century form another link with pellagra. The poorer and weaker South (home to pellagra) lost the civil war, losing both men and even more cattle. The North industrialised with many Yankee inventions and better wages that emerged ‘out of thin air’.165 In effect, a poor-meat diet and pellagra held back a modern economy for over 50 years166–169 (Figure 5). Fertility declined faster and earlier in the richer New England states and data support high fertility rates in slaves, short of starvation170–174 (Figure 6). As happened in the north of Italy, nicotinamide deficiency delays or stalls the switch to modernity however defined.175–177 A classic demographic transition as happened in the United Kingdom from 1850 correlates, by contrast, with rising meat and therefore nicotinamide levels (Figure 7).
Figure 5.

The poor diet in pellagra-ridden American southern states delayed economic progress, despite being the source of the international cotton industry.
Figure 6.

Comparison of birth rates between the industrial North and the South of the United States. Southern states prone to pellagra maintained high fertility for a lot longer as did the pellagra-prone province around Venice a century earlier – both examples lengthening their demographic transitions.
Figure 7.
Fertility rates fell after the death rate and as meat intake rose in the United Kingdom’s demographic transition. This is the opposite of what happened in the pellagra-prone provinces of Italy and the United States.
There has been debate over economic divergences between England and Europe or East and West. All relate to higher wages or natural resources allowing a higher meat intake. Nicotinamide adenine dinucleotide supply is the crucial variable. The first ‘luxury’ above subsistence level is meat. We now live in an ‘Age of Extremes’ without fully recognising that a balanced diet is at the base of progress or that NAD homeostasis is not only the master variable but also the master narrative.178–184
Lessons From Asia: ‘Land to the Tillers’ and Triumph of Gardening
Over the last 75 years, ‘tiger’ economies led by Japan, Taiwan, and South Korea transformed themselves reaping a demographic dividend from a healthy youthful population. China and India are following after false starts – such as the ‘Great Leap Forward’ pulling farmers off the land for ill-conceived industrial projects causing famine – or coercive family planning. Others are behind such as the Soviet bloc, North Korea, Cambodia, and Papua, New Guinea.185–195 Land and agrarian reform whether internally or externally driven (as happened in Japan) with reversion to small-holding mixed farming and later encouragement of export-led manufacture all improving diet was key to success. The tension between rural and town is striking with the latter being motivated to keep food and meat prices low: support for rural peasants in the form of land reform drives local innovation and efficiency and seems to work. Later, corporate agricultural arrangements can make sense once countries can import quality produce such as meat – policies that subsidise or import cheap or free cereals may however be disastrous in the long term.
Avoiding Malthusian Traps
Success and prosperity relate to avoiding Malthusian traps set up by calorific surpluses leading to population booms that bust. England and later America and now China all achieved this on the back of a higher nicotinamide diet that suppressed fertility and allowed better brains and longer lives. Malthus was perhaps right when discussing cereal-dependant economies but could not factor in food quality particularly if that relates not only to the ‘food necessary for existence’ but also to his second postulate that human passion between the sexes ‘would remain in its present state’.196–199 If societies get ahead on the meat curve, either after a Malthusian crash or from economic success, then lower fertility and better brains drive technological and economic progress in positive feedback cycles.200–220 It will be ironic if meat equity could have avoided such high global populations and high meat and cattle needs that are a major contributor to deforestation and ‘green-house’ gases through the use of fertilisers, water, and fossil fuels. Technological advances, often invoked to show that Malthus was wrong, could be temporary and ultimately cataclysmic fixes unless technology changes its biases towards optimal meat intake rather than more and more cereals delaying demographic transitions221–226 (Figure 8).
Figure 8.
Conventional demographic transition joined to the Neolithic transition. Lower meat drove the Neolithic, whereas an increase in the meat/cereal ratio drove recent transitions. Natural increase = excess of birth after deaths. NAD indicates nicotinamide adenine dinucleotide.
Nicotinamide, Gut Microbiome, and Tuberculosis
Nutritional symbioses include organisms that are dangerous by reputation, such as tuberculosis (TB). These symbionts support poor diets as does the gut microbiome but become dysbiotic if the diet becomes very poor. ‘Latent’ TB is metabolically active using host-derived cholesterol in exchange for nicotinic acid (Figure 9). Tuberculosis rarely evolves to evade the immune system (unlike the ‘arms races’ of pathogens): hosts may be tolerant for good metabolic reasons but seem perfectly capable of sterilising granulomas when they choose.227–237 The role of nutrition in activating latent TB has long been implicated and the harvest of deaths from TB when under dietary and other stresses often noted.
Figure 9.

Koch’s postulates need revision for nutritional symbiotic relationships. Symbionts, such as TB, enhance the supply of nicotinamide when the diet is poor but become dysbiotic if the diet becomes extremely poor. Improving diet, a preventive in the early stages, may no longer be enough to reverse pathology later. TB indicates tuberculosis.
Gut symbioses favour complex carbohydrate busters: even the oligosaccharide concentrations in breast milk affect the infant microbiome and make a contribution to nicotinamide levels (ruminants rely on their microbiome to supply vitamin B; Figure 10). Helminth interactions show extensive use of the tryptophan-NAD pathway that might benefit the host and NAD relationships extend to malaria and the host’s genetic responses. With malaria, an ecological approach is necessary as agricultural static water encourages Anopheles larvae, which grow on maize-specific pollen and then are more likely to be dichlorodiphenyltrichloroethane (DDT) resistant, and the adult malaria vector bites humans where there are few other animals to target.238–244 Emergent diseases in general are all more likely to evolve or become dysbiotic in poor ecological circumstances.
Figure 10.
Many pathogens import niacin. TB (and some gut microbes) can export nicotinic acid. On a high-nicotinamide diet, both classic pathogens and symbionts are less virulent or dysbiotic. NAD indicates nicotinamide adenine dinucleotide; TB, tuberculosis;
Tryptophan Metabolism and the Immune System
‘Host-directed therapies’ that enhance immunity through normalising NAD-consumer and energy-related sensors, such as mammalian target of rapamycin (mTOR) and AMP-activated protein kinase (AMPK) signalling rediscover the importance of nicotinamide. Metabolic regulation, over and above bioenergetic and biosynthetic demands of T cell differentiation, of immune responses works often through simple compounds, for instance, short-chain fatty acids (SCFAs) or glutamate metabolites programming T cell fates and the ratio of T helper 17 (Th17) and induced regulatory T cells (Tregs). These are conditioned by tryptophan status and co-evolved together in placental mammals to enable reproduction: functionally these pathways ameliorate autoimmune encephalomyelitis, the model for MS, and other pathogenic Th17-mediated autoimmune disease.8,245–273
Role of Tregs
Nicotinamide insufficiency activates the ‘de novo’ pathway. This leads to dysbioses, poor defences against pathogens, and ‘autocarnivory’ with organ damage. Activation predisposes to immune tolerance through the production of more Treg but less Th17 cells.274–284 Regulation of T cells has been linked with leprosy and TB (nicotinamide is antibiotic) and MS, myasthenia, and rheumatoid arthritis. Tregs that only exist in the periphery in placental mammals cause, shown by transfer experiments or rare mutations, other autoimmune diseases including oophoritis. Tregs with Th17 cells, even though they have a common developmental path, form an immune fulcrum governing tolerance to self and non-self and pro/anti-inflammation and B cell antibody responses and even (muscle) stem cell regeneration and tumour control. High levels of specific Tregs (but low Th17) with their anti-inflammatory cytokines and effect on dendritic cells discourage elimination of TB and other organisms, but low levels encourage ‘rogue’-specific self-reactive T cells and a spectrum of autoimmune diseases and allergies.285–290
Nicotinamide Switch Explains the Hygiene Hypothesis
This system therefore has checkpoints that connect nicotinamide with metabolism and innate and adaptive immunity, specifically the balance of Tregs and other T cell populations. This forms the ‘nicotinamide switch’ controlling the inflammatory response from activation to tolerance important to the latest versions of the ‘hygiene hypothesis’.291–299 In states of affluence, we ‘miss’ an evolved dependence on nutritional symbionts that are now surplus to requirements and are therefore ‘absent’. Even the foetus is exposed to this new environment with ‘maternal immune activation’ working through excess Th17 cells and interleukin-17 and a dearth of Tregs. At the other end of the nutritional spectrum, the IDO pathway will shut down when there is not even enough of its substrate tryptophan – and that will cause frank pellagra and complete immune and dysbiotic disarray and neurodegeneration – but notably no autoimmune disease.300,301
Indoleamine 2,3-dioxygenase is also a critical mediator of autoantibody production from B cells and a pathogenic driver of organ-specific autoimmune disease alongside its role in immune privilege, whether for the foetus or immune evasion for symbionts and cancers, and has an impact on mood by diverting tryptophan away from serotonin and tryptamine synthesis.302
High nicotinamide in diet therefore has the overall consequence of inducing immune intolerance (accepting some contradictions in the literature) and that may be behind the epidemic of autoimmune and allergic disease but perhaps also cancers and neuropsychiatric ills including autism and carbohydrate-induced obesity. A summary of the main pathways of nicotinamide metabolism is shown in Figures 11 and 12.
Figure 11.

NAD(H) recycles (not shown) in redox and dehydrogenase reactions and supplies mitochondria to generate ATP. Here we show consumption reactions and salvage pathways that conserve the supply of nicotinamide. When the dietary supply is poor, the ‘de novo’ pathway needs a dietary supply of tryptophan. ATP indicates adenosine triphosphate; NA, nicotinamide; NAD, nicotinamide adenine dinucleotide; NAD(H), nicotinamide adenine dinucleotide plus hydrogen; NAM, nicotinamide; NAMPT, nicotinamide phosphoribosyltransferase; NMN, nicotinamide mononucleotide; NMNAT, nicotinamide mononucleotide adenylyltransferase; NNMT, nicotinamide N-methyltransferase; NR, nicotinamide-riboside; PARP, poly(ADP-ribose) polymerase.
Figure 12.

Structure of nicotinamide showing its detoxification pathway that consumes methyl groups and produces N-methyl-nicotinamide that is metabolically active but then excreted. MNAM indicates N1-methylnicotinamide; NAM, nicotinamide; NNMT, nicotinamide N-methyltransferase; SAH, S-adenosylhomocysteine; SAM, S-adenosylmethionine.
Meat Elites
Americans and Europeans consume 150 kg of meat per annum alongside 250 kg of milk and eggs. The poorest eat negligible amounts of animal products. This is ironic as meat sharing was a defining feature of hunter-gatherer days, but later cattle ownership was the original form of capitalism that drove stratification and ‘meat elites’. Cash handouts to the poor would lead to a reasonable meat ration but is opposed currently for reasons that include cost and environmental concerns.
Too Little and too Much Meat – TB or Cancer and Autoimmunity
In recent times, these extremes have been tested with poor outcomes for billions. There is a long history of concern that too much meat causes cancer and an even longer history of advocating meat/milk supplements for the poor. One example – ‘Zomotherapy’ (zomos = meat broth) – was advocated by Nobel Laureates Charles Richet and Renee Dubos who also suggested skim milk for targeting TB (as did the sanatorium movement); this was later implemented as milk supplements and school meals with documented health benefits. Even earlier, Roget (of Thesaurus fame) in 1799 had published ‘Observations on the non-prevalence of consumption among Butchers and Fishermen’ – who, of course, had preferential access to their own produce.
Williams documented in 1908 falling TB rates with rising cancer rates and correlated them with meat intake in the United Kingdom and we replicated this finding (Figure 13). Rapid increases in TB rates have been documented on multiple occasions when diet deteriorates: some are recent, for instance, in the Russian ‘katastroika’ (around 1990) TB rates doubled in less than 5 years and life expectancy fell as the meat market collapsed. There is a further history of support for more meat for working people: for example, in the 19th-century United Kingdom there were calls for an ‘Industrial ration’, to be supplied by ‘killed and chilled’ corn-fed imports; and later for a ‘Colonial’ diet based on pioneering work done early in 20th-century Kenya showing the importance of animal products was proposed; and more recently rations (that included meat, bacon butter, and milk) were implemented to overcome the very poor meat intakes in the inter-war and world war years.303–306 Positive meat transitions from improved economics and these interventions are correlated with periods when health, height, and IQ increase and ‘modern minds are forged’.
Figure 13.
Striking time linked correlations between the fall in TB and the rise of cancer at a time when meat intake doubled. The same was true of the rise, particularly after 1900, in allergic and autoimmune diseases, as well as Parkinson’s disease. TB indicates tuberculosis.
Diseases Disappear and Appear: Risk of Plague
Both TB and leprosy disappear when and where nicotinamide dose in diet increases – nicotinamide is, after all, the original ‘antibiotic’ for both organisms.307,308 Malaria can also ‘disappear’ and of interest nicotinamide has anti-malarial activity as it does for other parasites. However, we should not be fooled by this ‘Mirage of Health’ given the recent grim comeback of ‘medieval’ pestilences and near apocalypses, such as plague in Madagascar.309–318 Recent warnings have come concerning future plagues using the parallel of the 1918 Spanish Flu pandemic that hit hard when metabolic requirements were high in the young and diets were poor. Diet is the commonest cause of impaired resistance to a wide variety of organisms including measles and smallpox epidemics, particularly on ‘virgin soils’ where populations have no previous exposure. Such warnings emphasise air travel, the ‘global village’, and antibiotic resistance but do not always emphasise the real microbe mutant magnets of poor diet and general squalor allowing the emergence of disease that are then a danger to rich and poor alike.
Earlier global crises, discussed already, were triggered by the weather (a lesson about dangers of climate change) and poor harvests from lost summers. There were widespread revolts between haves and have-nots as populations exploded then collapsed with descriptions of pellagra within the famines; ‘blackened faces like ovens’ in prematurely aged children among widespread poor behaviour, followed by plagues, and the rise of TB. Recovering nations revolutionised their agriculture away from cereal dependence with more pastureland and a mixed diet aided by the mass emigration to America and the first welfare states.
Circumstances where people live in ‘barnyard’ circumstances point now to crucibles of plagues in Asia or Africa – but it is worth remembering that the Flu epidemic 1918 - that killed more than both world wars combined – originated in pellagra-prone Kansas. The Cuban experience shows that poor-income countries can have effective health care systems coping with an epidemic of nutritional disease with widespread vitamin supplements.319–321 Amartya Sen, the Nobel Laureate, once said ‘I wonder whether there is any way of making poverty infectious – if so, I am certain its elimination would be remarkably rapid’ – dangers from poverty are, in fact, infectious (including violence) and that is one important lesson from the history of pellagra.
Nicotinamide and Better Brains
Pellagra also causes brain atrophy. Atrophy due to poor diet can be prevented as shown by the Flynn effect. Improved diet being necessary to improve learning (and teaching) before better schooling can build on a stronger cognitive base and lead to economic progress further improving diet and education. Improvement in IQ allows better brain reserves to combat ageing and that may explain the recent decline in the incidence of dementia in rich countries.322–327
Longevity: ‘Mens Sana in Corpore Sano’
There are links between nicotinamide and longevity. The observation is not controversial as nicotinamide has been explored as an anti-ageing compound in organisms from yeasts to worms to man. Proximate reasons relate to a better constitution, DNA repairs and reductions in virulence of pathogens and dangerous symbionts.328–335
A Refresh and Call to Action
Nicotinamide deficiency is unmeasured and underdiagnosed
The Columbian exchange brought maize to the Old World as it has the advantage of being easy to grow in difficult hydrological circumstances and has big returns per grain planted. Pellagra and subclinical nicotinamide deficiency is a risk (particularly when cultural traditions of mixed farming and special cooking are not exported) and had consequences first in the Americas but now in Asia and Africa. Pellagra still exists but is rarely formally diagnosed, prevented, or treated masquerading as ‘environmental enteropathy’ or general ill health and poor cognition – there is no easy biochemical test. Social breakdown can also be a feature as a (Marxian) ‘metabolic rift’. History is repeating itself as even in the pellagra epidemics cases were missed as the symptoms are protean and vague and seasonal with remissions. As a form of sunburn, black skins are resistant even evolved for and make it harder to spot. In the southern states of America, pellagra was rife and long before the official epidemic was endemic in the slave population and among poor whites ‘antebellum’ (and may have been a determinant of Confederate defeat) and instead called ‘black tongue’ or typhoid or a (genetic)negro ‘disease’.336
Nicotinamide: all is in the dose
A continuing role for missing symbionts is supported by evidence that re-introducing parasitic infection protects against allergic disease. This does not imply that they are metabolically needed when diet improves but emphasises their role in educating the immune system and that their absence causes problems, at least for a generation or two.
Nicotinamide at low doses is anti-cancer and neuroprotective but at a higher dose it is carcinogenic or neurotoxic. An optimal dose is even described in stem cell models as the ‘Fountain of Youth’. Some genetic and toxic and anoxic diseases respond if NAD is raised through diet or enzyme manipulation. A beneficial effect of nicotinamide on perinatal asphyxia or trauma alongside a range of developmental conditions has been demonstrated.337 Diseases that are NAD sensitive cover a variety of phenotypes and proposed mechanisms of neurotoxicity whether mitochondrial, proteotoxic, oxidative stress, or excitotoxic and whether cell body or axonal degeneration.338–356
Nicotinamide toxicity is common
High dosage in diet with induction of NNMT indicates that there might be a hypervitaminosis state with a wide phenotype – that includes the metabolic syndrome, some cancers, Parkinson, schizophrenia, and autism with double-edged sword relationships with dose. Depending on the dynamics of enzyme induction, if the dose is not maintained throughout life, then nicotinamide deficiency could occur on an apparently normal diet with increased catabolism confusing epidemiological studies.
Obesity and cancer
NNMT may be a target for obesity using novel anti-sense technology. High nicotinamide in diet could be toxic by a number of means and even homeostatic attempts to remove it with high NNMT levels in inflamed or pre-cancerous tissue could have long-term dangers357–373 (Figure 14).
Figure 14.
High nicotinamide in diet has consequences. The switch from infections to inflammation and autoimmunity can be explained by several overlapping mechanisms as can relative infertility and longevity alongside the metabolic syndrome and cancer. MNAM indicates N1-methylnicotinamide; NMN, nicotinamide mononucleotide; NNMT, nicotinamide N-methyltransferase; SAM, S-adenosylmethionine; PD, Parkinson’s disease.
Parkinson’s disease
Nicotinamide N-methyltransferase is raised in the brains of Parkinson’s disease (PD) patients as is N-methyl-nicotinamide excretion contributing to an argument, backed by epidemiological evidence that incidence had risen in rich high-meat-eating countries but was low in previous pellagra states, that nicotinamide could cause dopaminergic toxicity as an MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine)-like molecule. High-dose nicotinamide has shown toxicity in a proteasomal toxicity model of PD even though neuroprotective prophylactically in MPTP models. Nicotinamide is an important morphogen encouraging neuronal differentiation towards dopaminergic cells at moderate but not high doses.374 Furthermore, NNMT has been shown to interfere, by consuming methyl groups, with DNA methylation and autophagy that controls quality of proteins and organelles, as shown by toxins or PD mutations, but if excessive can cause cell death. There is epidemiological support for PD being a disease of affluence: China had an incidence of one fifth of rich nations but this is closing rapidly as their meat intake increases. The argument that this rise in incidence is all due to an ageing population is complex if higher nicotinamide dose is driving longevity – PD then being a side-effect of the cause of better ageing, rather than due to ageing per se375–386 (Figure 15).
Figure 15.
Nicotinamide dose matters from conception to cradle to grave. PD is a good example. An optimal dose induces NNMT and supplies NAD to mitochondria and NAD consumers and is enough to regulate DNA methylation and stimulate autophagy to keep organelles in good repair. Too much (or too little) nicotinamide and all fails, exacerbated by genetic mutations that affect autophagy known to be important in PD. MPTP indicates 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; NAD, nicotinamide adenine dinucleotide; NNMT, nicotinamide N-methyltransferase; PARP, poly(ADP-ribose) polymerase; PD, Parkinson’s disease; SAM, S-adenosylmethionine.
International fertility: redux and review
Many have mentioned cereal diets increasing fertility by increasing carbohydrates, or indirect effects such as enabling early weaning, but have not discussed the tryptophan pathway. We showed correlations between meat intake and improved longevity and declining fertility in the United Kingdom during 1850 to 1950 and argued that averting a population collapse was helped by revisions of the Corn Laws and the ‘ecological windfall’ of using money from (cotton) exports to import meat often from colonies. Meat famines at home were mitigated by agricultural and hunting arrangements abroad – to the detriment of local populations (now the Third World) but allowing painless demographic transitions at home.
France, for instance, where maize was banned for human consumption, barely had a demographic transition: whereas low-meat eaters such as China, India, and Japan had their transition in the mid-20th century slowly and painfully with large population booms much later than Europe – cereal-dependant modern Africa fails to complete the transition. These observations are consistent with population booms on American maize, between 1750 and 1850. Looking to poor diet for high fertility makes a welcome relief from blaming poor genes or race as in the United States where eugenists believed that pellagrins were ‘feeble-minded’ and ‘shiftless’ multi-generationally and should enter legal sterilisation programmes with marriage and immigration prohibitions: casting a long shadow to later genocidal and immigration policies.
Analyses of declining birth rates first among the wealthy discuss conscious contraception or a preference for careers or a reluctance of women to produce ‘cannon fodder’ but overlook diet as a potential factor. Education, emancipation, and birth control may however be the dividend building on a food-dependant demography and higher human capital. Increased fertility on a more vegetarian diet and decreased fertility on a high-meat diet go back to hunter-gatherers such as the Khoisan people in South Africa who were out-reproduced by Bantu agriculturalists. Meat reduces and poor vegetarian diets increase fertility. This may explain (and been evolved for) baby booms after famines and why low-meat/high-cereal societies have population booms and ‘Malthusian’ corrections. Evidence from other animal populations that pumping in calories (‘paradox of enrichment’) leads to population instability will make sense once tryptophan metabolism is factored into their interpretation. Optimal meat/nicotinamide may allow for painless demographic transitions with sustainable environments and populations. As a corollary, fertility declines may reverse when extremes of high meat intake moderate.
‘Antagonistic Pleiotropy’, ‘Disposable Soma’, ‘Thrifty’ Genes and Phenotypes
In possible ‘proof of concept’ twists, the early fertility crises reversed by a more plant-based diet in the Palaeolithic left marks on our genome. Intriguingly, these pro-fertility genes, such as apolipoprotein E4 (APOE-4), interact with infections with resistance to diarrhoeal illness predisposed to by pellagra. Mutations spread at the time of fertility and infectious stress now showing up as risk factors for late-onset non-communicable diseases such as cancer and neurodegeneration.387–392 Many mutations involve NAD metabolism and DNA repair suggesting that they evolved at times when nicotinamide homeostasis was out of kilter – but could now be helped by altering the dose of nicotinamide by individual genome and depending on age.393–398 Another trade-off between fertility and healthy ageing is the ‘disposable soma’ theory whereby reproduction is metabolically favoured over repairs – this trade-off (with immunosuppression leading to greater fecundity but more infection) fades away during epidemiological transitions with a more carnivorous diet, and experimentally with increased ‘autocarnivory’.390,399–408 ‘Thrifty’ genotypes and phenotypes can also be brought into this discussion as they may be a manifestation of ‘r’ selection for quantity over quality at a price with late costs, such as the metabolic syndrome, being perhaps avoidable by fairer nicotinamide sourcing throughout and across lives.400,409–414
Conclusions
Nicotinamide is critical to ‘evolution in four dimensions’ as it affects genomic, epigenomic, behavioural, and symbolic/cultural inheritance.415–419 Nicotinamide resonates between developmental and phenotypic plasticity and a niche-constructed ecologically inherited ‘NAD’ world. Nicotinamide, buffered by the microbiome, allowed and selected hominid lineages to evolve into anatomically modern man and then a fertility crisis 30 000 to 40 000 years ago in Europe (earlier in southern Africa and Asia) drove a pro-fertility-plant-based diet with mating brains and cultural artefacts that we call civilisation.420,421
As Huxley implied, we should now direct our evolution by ensuring an appropriate diet for all as an entitlement. The flourishing of humankind’s culture in the Mesolithic and the later economic and artistic and scientific ‘take-off’ involved diet that is an independent variable for fertility, health, and brain power. Iatrogenic climate change and other worries about the future may sort themselves with higher human capital but lower numbers of people.422–427
Moving up then down the food chain led to the ‘Ultra-social conquest of Earth’ but now we can aim for individual quality. We no longer need poverty to encourage fertility and should guarantee a ‘goldilocks’ diet for all as a human right and out of self-interest to avoid plagues and wars. A well-balanced diet would underpin progress everywhere so that ‘No One’ country is dominant and return to our more egalitarian meat-sharing past.428,429 Active intervention in population control has not been that successful suggesting that we need to look at fundamental biological and dietary controls given that ‘Demography is Destiny’.430–432
Food sovereignty puts an emphasis on food quality by political and scientific means known as ‘Physiocracy’ that can be traced back to mixed farming enthusiasts, such as Virgil or Cato, and Hippocratic dietary regimens.433–449 Hippocratic regimens are only slightly different from current dietary advice. Becoming vegans in a ‘meat retreat’ is not the answer to good health, animal rights, population control, climate change, or loss of biodiversity. Meat hunger is for good biological reason and Engel’s law repeatedly shows that poor individuals (and nations) eat more meat if allowed. However, there is a limit and we propose a hypervitaminosis B3 with an equally broad phenotype to pellagra. This is the ‘Wisdom of the Body’ pertaining to NAD homeostasis. We should avoid ending, like other empires that collapsed, because we ignored historical intelligence and did not appreciate fully that progress depends on a balanced diet and sharing meat across societies and nations.450–456
Footnotes
Funding:The authors disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This study was funded by QEHB Charity, Birmingham, UK.
Declaration of conflicting interests:The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
Author Contributions: AW and LJH wrote the manuscript, approved and submitted the final draft.
ORCID iD: Lisa J Hill
https://orcid.org/0000-0001-8431-7029
References
- 1. Clutton-Brock J, Museum NH. A Natural History of Domesticated Mammals. Cambridge, UK: Cambridge University Press; 1999. [Google Scholar]
- 2. Cohen LJ, Esterhazy D, Kim S-H, et al. Commensal bacteria make GPCR ligands that mimic human signalling molecules. Nature. 2017;549:48–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Crockett MJ, Clark L, Smillie L, Robbins TW. The effects of acute tryptophan depletion on costly information sampling: impulsivity or aversive processing? Psychopharmacology. 2012;219:587–597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Jenkins TA, Nguyen JC, Polglaze KE, Bertrand PP. Influence of tryptophan and serotonin on mood and cognition with a possible role of the gut-brain axis. Nutrients. 2016;8:56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Rogers SM, Ott SR. Differential activation of serotonergic neurons during short-and long-term gregarization of desert locusts. Proc Biol Sci. 2015;282:20142062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Toyomaki A, Koga M, Okada E, et al. The relationship between a low grain intake dietary pattern and impulsive behaviors in middle-aged Japanese people. PLoS ONE. 2017;12:e0181057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Worbe Y, Savulich G, Voon V, Fernandez-Egea E, Robbins TW. Serotonin depletion induces ‘waiting impulsivity’ on the human four-choice serial reaction time task: cross-species translational significance. Neuropsychopharmacology. 2014;39:1519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Zhu X, Han Y, Du J, Liu R, Jin K, Yi W. Microbiota-gut-brain axis and the central nervous system. Oncotarget. 2017;8:53829–53838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Fagan B. The Intimate Bond: How Animals Shaped Human History. New York, NY: Bloomsbury; 2015. [Google Scholar]
- 10. Gray J. Straw Dogs: Thoughts on Humans and Other Animals. London, England: Granta Publications; 2015. [Google Scholar]
- 11. Morey D. Dogs: Domestication and the Development of a Social Bond. Cambridge, UK: Cambridge University Press; 2010. [Google Scholar]
- 12. Harris DR. Agriculture, cultivation and domestication: exploring the conceptual framework of early food production. In: Denham TP, Iriarte J, Vrydaghs L, eds. Rethinking Agriculture: Archaeological and Ethnoarchaeological Perspectives. Walnut Creek, CA: Left Coast; 2007:16–35. [Google Scholar]
- 13. Francis RC. Domesticated: Evolution in a Man-made World. New York, NY: W. W. Norton & Company; 2016. [Google Scholar]
- 14. Zumla A, Maeurer M, Chakaya J, et al. Towards host-directed therapies for tuberculosis. Nat Rev Drug Discov. 2015;14:511–512. [DOI] [PubMed] [Google Scholar]
- 15. Durham WH. Coevolution: Genes, Culture, and Human Diversity. Stanford, CA: Stanford University Press; 1991. [Google Scholar]
- 16. Pinker S. The Better Angels of Our Nature: Why Violence Has Declined. London, England: Penguin Books; 2012. [Google Scholar]
- 17. Pulliam HR, Dunford C. Programmed to Learn: An Essay on the Evolution of Culture. New York, NY: Columbia University Press; 1980. [Google Scholar]
- 18. Ashby W. Design for a Brain: The Origin of Adaptive Behaviour. Berlin, Germany: Springer Science+Business Media; 2013. [Google Scholar]
- 19. Cronin H. The Ant and the Peacock: Altruism and Sexual Selection From Darwin to Today. Cambridge, UK: Cambridge University Press; 1993. [Google Scholar]
- 20. Dixson AF. Sexual Selection and the Origins of Human Mating Systems. Oxford, UK: Oxford University Press; 2009. [Google Scholar]
- 21. Ghiselin MT. The Economy of Nature and the Evolution of Sex. Berkeley, CA: University of California Press; 1974. [Google Scholar]
- 22. Hoquet T. Current Perspectives on Sexual Selection: What’s Left After Darwin? Dordrecht, The Netherlands: Springer; 2015. [Google Scholar]
- 23. Ingold T. The Appropriation of Nature: Essays on Human Ecology and Social Relations. Iowa City, IA: University of Iowa Press; 1987. [Google Scholar]
- 24. Jochim MA. Strategies for Survival: Cultural Behavior in an Ecological Context. Amsterdam, The Netherlands: Elsevier Science; 2013. [Google Scholar]
- 25. Miller G. The Mating Mind: How Sexual Choice Shaped the Evolution of Human Nature. London, England: Vintage Books; 2001. [Google Scholar]
- 26. Taylor T. The Prehistory of Sex: Four Million Years of Human Sexual Culture. London, England: Fourth Estate; 1997. [Google Scholar]
- 27. Bahn P, Lorblanchet M, Soulages P. The First Artists: In Search of the World’s Oldest Art. London, England: Thames & Hudson; 2017. [Google Scholar]
- 28. Henrich J. The Secret of Our Success: How Culture Is Driving Human Evolution, Domesticating Our Species, and Making Us Smarter. Princeton, NJ: Princeton University Press; 2015. [Google Scholar]
- 29. Tanner NM. On Becoming Human. Cambridge, UK: Cambridge University Press; 1981. [Google Scholar]
- 30. Ruhlen M. The Origin of Language: Tracing the Evolution of the Mother Tongue. Hoboken, NJ: Wiley; 1996. [Google Scholar]
- 31. Boyer P. Minds Make Societies: How Cognition Explains the World Humans Create. New Haven, CT: Yale University Press; 2018. [Google Scholar]
- 32. Bahn P. Cave Art: A Guide to the Decorated Ice Age Caves of Europe. London, England: Frances Lincoln; 2012. [Google Scholar]
- 33. Ryan C, Jeth C. Sex at Dawn: The Prehistoric Origins of Modern Sexuality. Melbourne, VIC, Australia: Scribe Publications; 2011. [Google Scholar]
- 34. Chadwick W. Women, Art, and Society. London, England: Thames & Hudson; 1990. [Google Scholar]
- 35. Clutton-Brock T. Sexual selection in males and females. Science. 2007;318:1882–1885. [DOI] [PubMed] [Google Scholar]
- 36. Clutton-Brock TH. The Evolution of Parental Care. Princeton, NJ: Princeton University Press; 1991. [Google Scholar]
- 37. Dunbar RIM, Knight C, Power C. The Evolution of Culture: An Interdisciplinary View. New Brunswick, NJ: Rutgers University Press; 1999. [Google Scholar]
- 38. Fisher HE. The Sex Contract: The Evolution of Human Behavior. New York, NY: William Morrow; 1982:97. [Google Scholar]
- 39. Gowdy J, Krall L. The economic origins of ultrasociality. Behav Brain Sci. 2016;39:e92. [DOI] [PubMed] [Google Scholar]
- 40. Hauser MD. The Evolution of Communication. Cambridge, MA: The MIT Press; 1996. [Google Scholar]
- 41. Jacobs LF. Sexual selection and the brain. Trend Ecol Evol. 1996;11:82–86. [DOI] [PubMed] [Google Scholar]
- 42. Margulis L, Sagan D. Mystery Dance: On the Evolution of Human Sexuality. New York, NY: Summit Books; 1991. [Google Scholar]
- 43. Smith JM. Sexual selection, handicaps and true fitness. J Theoret Biol. 1985;115:1–8. [DOI] [PubMed] [Google Scholar]
- 44. Aiken N. The Biological Origins of Art. Westport, CT: Praeger; 1998. [Google Scholar]
- 45. Williams AC, Hill LJ. Nicotinamide’s ups and downs: consequences for fertility, development, longevity and diseases of poverty and affluence. Int J Tryptophan Res. 2018;11:1178646918802289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Henrich JP. Foundations of Human Sociality: Economic Experiments and Ethnographic Evidence from Fifteen Small-Scale Societies. Oxford: Oxford University Press; 2004. [Google Scholar]
- 47. Nowak M, Highfield R. SuperCooperators: Altruism, Evolution, and Why We Need Each Other to Succeed. New York, NY: Free Press; 2012. [Google Scholar]
- 48. Ridley M. It took 10,000 generations to make an iPhone. The Times. https://www.thetimes.co.uk/edition/comment/it-took-10-000-generations-to-make-an-iphone-8phfggzqz. Up-dated 2017.
- 49. Judson O. Dr Tatiana’s Sex Advice to All Creation: Definitive Guide to the Evolutionary Biology of Sex. New York, NY: Random House; 2014. [Google Scholar]
- 50. Adams RMC. Land Behind Baghdad: A History of Settlement on the Diyala Plains. Chicago, IL: University of Chicago Press; 1965. [Google Scholar]
- 51. Barkow JH, Cosmides L, Tooby J. The Adapted Mind: Evolutionary Psychology and the Generation of Culture. New York, NY: Oxford University Press; 1995. [Google Scholar]
- 52. Despres LA. Ethnicity and Resource Competition in Plural Societies. Berlin, Germany: De Gruyter; 1975. [Google Scholar]
- 53. Etcoff N. Survival of the Prettiest: The Science of Beauty. New York, NY: Knopf Doubleday; 2011. [Google Scholar]
- 54. Gregg SA. Foragers and Farmers: Population Interaction and Agricultural Expansion in Prehistoric Europe. Chicago, IL: University of Chicago Press; 1988. [Google Scholar]
- 55. Marcuse H. Eros and Civilization. Abingdon, UK: Routledge; 2012. [Google Scholar]
- 56. Nissen HJ, Lutzeier E. The Early History of the Ancient Near East, 9000–2000 B.C. Chicago, IL: University of Chicago Press; 2011. [Google Scholar]
- 57. Symons D. Beauty is in the adaptations of the beholder: the evolutionary psychology of human female sexual attractiveness. In: Abramson PR, Pinkerton SD, eds. Sexual Nature, Sexual Culture. Chicago, IL: University of Chicago Press; 1995:80–118. [Google Scholar]
- 58. Tauger MB. Agriculture in World History. Abingdon, UK: Taylor & Francis; 2013. [Google Scholar]
- 59. Thornhill R, Grammer K. The body and face of woman: one ornament that signals quality? Evol Human Behavior. 1999;20:105–120. [Google Scholar]
- 60. Childe VG. The Dawn of European Civilization. Abingdon, UK: Taylor & Francis; 2013. [Google Scholar]
- 61. Braidwood RJ, Braidwood L. Jarmo: a village of early farmers in Iraq. Antiquity. 1950;24:189–195. [Google Scholar]
- 62. Redman CL. The Rise of Civilization: From Early Farmers to Urban Society in the Ancient Near East. San Francisco, CA: WH Freeman; 1978. [Google Scholar]
- 63. Mazoyer M, Roudart L, Membrez JH. A History of World Agriculture: From the Neolithic Age to the Current Crisis. New York, NY: Monthly Review Press; 2006. [Google Scholar]
- 64. Pollock S. Feasts, funerals, and fast food in early Mesopotamian states. In: Bray TL, ed. The Archaeology and Politics of Food and Feasting in Early States and Empires. Boston, MA: Springer; 2003:17–38. [Google Scholar]
- 65. Bellwood P. First Islanders: Prehistory and Human Migration in Island Southeast Asia. Hoboken, NJ: Wiley; 2017. [Google Scholar]
- 66. Bollig M, Schnegg M, Wotzka HP. Pastoralism in Africa: Past, Present and Future. New York, NY: Berghahn Books; 2013. [Google Scholar]
- 67. Hakenbeck SE, Evans J, Chapman H, Fothi E. Practising pastoralism in an agricultural environment: an isotopic analysis of the impact of the Hunnic incursions on Pannonian populations. PLoS ONE. 2017;12:e0173079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Salzman PC. Pastoralists: Equality, Hierarchy, and the State. Abingdon, UK: Taylor & Francis; 2018. [Google Scholar]
- 69. Bottéro J. The Oldest Cuisine in the World: Cooking in Mesopotamia. Chicago, IL: University of Chicago Press; 2004. [Google Scholar]
- 70. Goody J. Cooking, Cuisine and Class: A Study in Comparative Sociology. Cambridge, UK: Cambridge University Press; 1982. [Google Scholar]
- 71. Belich J. Replenishing the Earth: The Settler Revolution and the Rise of the Angloworld. Oxford, UK: Oxford University Press; 2011. [Google Scholar]
- 72. Kiple KF. A Movable Feast: Ten Millennia of Food Globalization. Cambridge, UK: Cambridge University Press; 2007. [Google Scholar]
- 73. Grohmann U, Fallarino F, Puccetti P. Tolerance, DCs and tryptophan: much ado about IDO. Trends Immunol. 2003;24:242–248. [DOI] [PubMed] [Google Scholar]
- 74. Grohmann U, Puccetti P. The coevolution of IDO1 and AhR in the emergence of regulatory T-cells in mammals. Front Immunol. 2015;6:58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Barbi J, Pardoll D, Pan F. Metabolic control of the Treg/Th17 axis. Immunol Rev. 2013;252:52–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Bessede A, Gargaro M, Pallotta MT, et al. Aryl hydrocarbon receptor control of a disease tolerance defence pathway. Nature. 2014;511:184–190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Liston A. Regulatory T Cells in Health and Disease. Amsterdam, The Netherlands: Elsevier Science; 2015. [Google Scholar]
- 78. Smith PM, Howitt MR, Panikov N, et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science. 2013;341:569–573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Huntington E. Mainsprings of Civilization. New York, NY: New American Library; Mentor Books; 1959. [Google Scholar]
- 80. Noble W, Davidson I, Hobbs D. Human Evolution, Language and Mind: A Psychological and Archaeological Inquiry. Cambridge, UK: Cambridge University Press; 1996. [Google Scholar]
- 81. Adams CJ. The Sexual Politics of Meat - 25th Anniversary Edition: A Feminist-Vegetarian Critical Theory. New York, NY: Bloomsbury; 2015. [Google Scholar]
- 82. Alesina A, Giuliano P, Nunn N. On the origins of gender roles: women and the plough. Quarter J Economic. 2013;128:469–530. [Google Scholar]
- 83. Harris S. What Have Plants Ever Done for Us? Western Civilization in Fifty Plants. Oxford, UK: Bodleian Library; 2015. [Google Scholar]
- 84. Evans-Pritchard EE. The Nuer. Oxford, UK: Clarendon Press; 1940. [Google Scholar]
- 85. Smil V. Should We Eat Meat? Evolution and Consequences of Modern Carnivory. Hoboken, NJ: John Wiley & Sons; 2013. [Google Scholar]
- 86. Stanford CB. The Hunting Apes: Meat Eating and the Origins of Human Behavior. Princeton, NJ: Princeton University Press; 1999. [Google Scholar]
- 87. Spencer C. The Heretic’s Feast: A History of Vegetarianism. Lebanon, NH: University Press of New England; 1996. [Google Scholar]
- 88. Larsen CS. Bioarchaeology: Interpreting Behavior from the Human Skeleton. Cambridge, UK: Cambridge University Press; 2015. [Google Scholar]
- 89. Moltke I, Fumagalli M, Korneliussen TS, et al. Uncovering the genetic history of the present-day Greenlandic population. Am J Hum Genet. 2015;96:54–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Hall M. Farmers, Kings, and Traders: The People of Southern Africa, 200-1860. Chicago, IL: University of Chicago Press; 1990. [Google Scholar]
- 91. McGuire MK, Meehan CL, McGuire MA, et al. What’s normal? Oligosaccharide concentrations and profiles in milk produced by healthy women vary geographically. Am J Clin Nutr. 2017;105:1086–1100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. McNeill JR. Mosquito Empires: Ecology and War in the Greater Caribbean, 1620–1914. Cambridge, UK: Cambridge University Press; 2010. [Google Scholar]
- 93. Beaver MW. Population, infant mortality and milk. Popul Stud (Camb). 1973;27:243–254. [PubMed] [Google Scholar]
- 94. Hudson WH. Far Away and Long Ago. Auckland, New Zealand: Floating Press; 2017. [Google Scholar]
- 95. Linklater A. Owning the Earth: The Transforming History of Land Ownership. New York, NY: Bloomsbury; 2014. [Google Scholar]
- 96. Walling P. Till the Cows Come Home: The Story of Our Eternal Dependence. London, England: Atlantic Books; 2018. [Google Scholar]
- 97. Verano JW, Ubelaker DH. Disease and demography in the Americas. J Field Archaeol. 1992;22:119–121. [Google Scholar]
- 98. Evans H, Buckland G, Lefer D. They Made America: From the Steam Engine to the Search Engine: Two Centuries of Innovators. Boston, MA: Little, Brown and Company; 2009. [Google Scholar]
- 99. Johnson P. The Birth of the Modern: World Society 1815–1830. London, England: Orion; 2013. [Google Scholar]
- 100. Olson M. The Rise and Decline of Nations: Economic Growth, Stagflation, and Social Rigidities. New Haven, CT: Yale University Press; 2008. [Google Scholar]
- 101. Phelps ES. Mass Flourishing: How Grassroots Innovation Created Jobs, Challenge, and Change. Princeton, NJ: Princeton University Press; 2013. [Google Scholar]
- 102. Wells DA. Recent Economic Changes, and Their Effect on the Production and Distribution of Wealth and the Well-being of Society. Boston, MA: D. Appleton; 1899. [Google Scholar]
- 103. Crafts NF. Some dimensions of the ‘quality of life’ during the British industrial revolution. Econom History Rev. 1997;50:617–639. [Google Scholar]
- 104. Aberth J. From the Brink of the Apocalypse: Confronting Famine, War, Plague and Death in the Later Middle Ages. Abingdon, UK: Taylor & Francis; 2013. [Google Scholar]
- 105. Allen RC. Global Economic History: A Very Short Introduction. Oxford, UK: Oxford University Press; 2011. [Google Scholar]
- 106. Armitage D, Subrahmanyam S. The Age of Revolutions in Global Context, C. 1760–1840. Basingstoke, UK: Palgrave Macmillan; 2009. [Google Scholar]
- 107. Darwin J. After Tamerlane: The Rise and Fall of Global Empires, 1400–2000. London, England: Penguin Books; 2007. [Google Scholar]
- 108. Dugan S, Dugan D. The Day the World Took Off: The Roots of the Industrial Revolution. London, England: Channel 4 Books; 2000. [Google Scholar]
- 109. Engerman SL, Gallman RE. Long-term Factors in American Economic Growth. Chicago, IL: University of Chicago Press; 2007. [Google Scholar]
- 110. Epstein SR. Freedom and Growth: The Rise of States and Markets in Europe, 1300–1750. Abingdon, UK: Taylor & Francis; 2002. [Google Scholar]
- 111. Findlay R, O’Rourke KH. Power and Plenty: Trade, War, and the World Economy in the Second Millennium. Princeton, NJ: Princeton University Press; 2009. [Google Scholar]
- 112. Gerth K. As China Goes, So Goes the World: How Chinese Consumers Are Transforming Everything. New York, NY: Farrar, Straus and Giroux; 2010. [Google Scholar]
- 113. Jones E. The European Miracle: Environments, Economies and Geopolitics in the History of Europe and Asia. Cambridge, UK: Cambridge University Press; 2003. [Google Scholar]
- 114. Mokyr J. The Lever of Riches: Technological Creativity and Economic Progress. Oxford, UK: Oxford University Press; 1992. [Google Scholar]
- 115. van Bath BHS. The Agrarian History of Western Europe, A.D. 500–1850. London, England: Edward Arnold; 1963. [Google Scholar]
- 116. Rostow WW. The Stages of Economic Growth: A Non-communist Manifesto. 1st ed Eastford, CT: Martino Fine Books; 2017. [Google Scholar]
- 117. Allen RC. The nitrogen hypothesis and the English agricultural revolution: a biological analysis. J Econom History. 2008;68:182–210. [Google Scholar]
- 118. Borsch SJ. The Black Death in Egypt and England: A Comparative Study. Austin, TX: University of Texas Press; 2009. [Google Scholar]
- 119. Clark G. A Farewell to Alms: A Brief Economic History of the World. Princeton, NJ: Princeton University Press; 2008. [Google Scholar]
- 120. Joyce P. Visions of the People: Industrial England and the Question of Class, C.1848–1914. Cambridge, UK: Cambridge University Press; 1994. [Google Scholar]
- 121. Rule J. The Labouring Classes in Early Industrial England, 1750–1850. Abingdon, UK: Taylor & Francis; 2014. [Google Scholar]
- 122. Wrightson K. A Social History of England, 1500–1750. Cambridge, UK: Cambridge University Press; 2017. [Google Scholar]
- 123. Clark G, Huberman M, Lindert PH. A British food puzzle, 1770–1850. Econom History Rev. 1995;48:215–237. [Google Scholar]
- 124. Galloway PR. Basic patterns in annual variations in fertility, nuptiality, mortality, and prices in pre-industrial Europe. Popul Stud (Camb). 1988;42:275–303. [DOI] [PubMed] [Google Scholar]
- 125. Komlos J. Shrinking in a growing economy? The mystery of physical stature during the industrial revolution. J Econom History. 1998;58:779–802. [Google Scholar]
- 126. Mokyr J. The Enlightened Economy: An Economic History of Britain 1700–1850. New Haven, CT: Yale University Press; 2012. [Google Scholar]
- 127. O’Rourke KH. The European grain invasion, 1870–1913. J Econom History. 1997;57:775–801. [Google Scholar]
- 128. Overton M. Agricultural Revolution in England: The Transformation of the Agrarian Economy 1500–1850. Cambridge, UK: Cambridge University Press; 1996. [Google Scholar]
- 129. Persson KG. Grain Markets in Europe, 1500–1900: Integration and Deregulation. Cambridge, UK: Cambridge University Press; 1999. [Google Scholar]
- 130. Post JD. The mortality crises of the early 1770s and European demographic trends. J Interdiscip History. 1990;21:29–62. [Google Scholar]
- 131. Bregman R. Utopia for Realists: And How We Can Get There. New York, NY: Bloomsbury; 2018. [Google Scholar]
- 132. Burnett J. Plenty and Want: A Social History of Food in England from 1815 to the Present Day. Abingdon, UK: Routledge; 2013. [Google Scholar]
- 133. Davis M. Late Victorian Holocausts: El Niño Famines and the Making of the Third World. Abingdon, UK: Taylor & Francis; 2002. [Google Scholar]
- 134. Hilton B. A Mad, Bad, and Dangerous People? England 1783–1846. Oxford, UK: Oxford University Press; 2008. [Google Scholar]
- 135. Pickett K, Wilkinson R. The Spirit Level: Why Equality is Better for Everyone. London, England: Penguin Books; 2010. [Google Scholar]
- 136. Evans J. Emigrants: Why the English Sailed to the New World. London, England: Orion; 2017. [Google Scholar]
- 137. Fagan B. The Little Ice Age: How Climate Made History 1300–1850. New York, NY: Basic Books; 2001. [Google Scholar]
- 138. Fogel RW. The Escape from Hunger and Premature Death, 1700–2100: Europe, America, and the Third World. Cambridge, UK: Cambridge University Press; 2004. [Google Scholar]
- 139. Parker G. Global Crisis: War, Climate Change and Catastrophe in the Seventeenth Century. New Haven, CT: Yale University Press; 2017. [Google Scholar]
- 140. Bayly CA. Remaking the Modern World 1900–2015: Global Connections and Comparisons. Hoboken, NJ: Wiley; 2018. [Google Scholar]
- 141. Brands HW. American Colossus: The Triumph of Capitalism, 1865–1900. New York, NY: Anchor Books; 2011. [Google Scholar]
- 142. De Vries J. The Industrious Revolution: Consumer Behavior and the Household Economy, 1650 to the Present. Cambridge, UK: Cambridge University Press; 2008. [Google Scholar]
- 143. Dyer C. Standards of Living in the Later Middle Ages: Social Change in England C.1200–1520. Cambridge, UK: Cambridge University Press; 1989. [Google Scholar]
- 144. Engerman SL, Genovese ED. Race and Slavery in the Western Hemisphere: Quantitative Studies. Princeton, NJ: Princeton University Press; 1981. [Google Scholar]
- 145. Floud R, Floud PR, Gregory A, et al. Height, Health and History: Nutritional Status in the United Kingdom, 1750–1980. Cambridge, UK: Cambridge University Press; 1990. [Google Scholar]
- 146. Glass DV, Eversley DEC. Population in History: Essays in Historical Demography. London, England: Aldine; 1965. [Google Scholar]
- 147. Goldthwaite RA. The Building of Renaissance Florence: An Economic and Social History. Baltimore, MD: Johns Hopkins University Press; 1982. [Google Scholar]
- 148. Greenspan A, Wooldridge A. Capitalism in America: A History. London, England: Penguin Books; 2018. [Google Scholar]
- 149. Hollingsworth TH. Historical Demography. Cambridge, UK: Cambridge University Press; 1976. [Google Scholar]
- 150. Klein HS. A Population History of the United States. Cambridge, UK: Cambridge University Press; 2012. [Google Scholar]
- 151. Lopez RS. The Commercial Revolution of the Middle Ages, 950–1350. Cambridge, UK: Cambridge University Press; 1976. [Google Scholar]
- 152. McDougall WA. Throes of Democracy: The American Civil War Era, 1829–1877. New York, NY: HarperCollins; 2009. [Google Scholar]
- 153. McKendrick N, Plumb JH. The Birth of a Consumer Society: The Commercialization of Eighteenth-Century England. Brighton, UK: Edward Everett Root; 2018. [Google Scholar]
- 154. McPherson JM. The Battle Cry of Freedom: The Civil War Era. Lawrence, KS: University of Kansas Press; 1988. [Google Scholar]
- 155. Ogilvie S, Cerman M. European Proto-industrialization: An Introductory Handbook. Cambridge, UK: Cambridge University Press; 1996. [Google Scholar]
- 156. Schwarz LD, De Vries J, Smith R, Johnson P, De Vries PHEJ, Wrightson K. London in the Age of Industrialisation: Entrepreneurs, Labour Force and Living Conditions, 1700–1850. Cambridge, UK: Cambridge University Press; 1992. [Google Scholar]
- 157. Sharpe J. Early Modern England: A Social History 1550–1760. Oxford, UK: Oxford University Press; 1997. [Google Scholar]
- 158. Van Zanden JL. The Long Road to the Industrial Revolution: The European Economy in a Global Perspective, 1000–1800. Leiden, The Netherlands: Brill; 2009. [Google Scholar]
- 159. Wrigley EA, Schofield RS, Schofield R, Lee R. The Population History of England 1541–1871. Cambridge, UK: Cambridge University Press; 1989. [Google Scholar]
- 160. Drayton RH. Nature’s Government: Science, Imperial Britain, and the ‘Improvement’ of the World. New Haven, CT: Yale University Press; 2000. [Google Scholar]
- 161. Harlow VT. The founding of the Second British Empire, 1763–1793. Harlow, UK: Longmans; 1952. [Google Scholar]
- 162. Devine TM. The Scottish Clearances: A History of the Dispossessed, 1600–1900. London, England: Penguin Books; 2018. [Google Scholar]
- 163. Bayly CA. The Birth of the Modern World, 1780–1914. Hoboken, NJ: Wiley; 2004. [Google Scholar]
- 164. Perren R. Taste, Trade and Technology: The Development of the International Meat Industry Since 1840. Abingdon, UK: Taylor & Francis; 2017. [Google Scholar]
- 165. Kulikoff A. From British Peasants to Colonial American Farmers. Chapel Hill, NC: University of North Carolina Press; 2014. [Google Scholar]
- 166. Beckert S. Empire of Cotton: A Global History. New York, NY: Knopf Doubleday; 2014. [Google Scholar]
- 167. Burnard T. Planters, Merchants, and Slaves: Plantation Societies in British America, 1650–1820. Chicago, IL: University of Chicago Press; 2015. [Google Scholar]
- 168. Howe DW. What Hath God Wrought: The Transformation of America, 1815–1848. Oxford, UK: Oxford University Press; 2007. [Google Scholar]
- 169. Jacob MC. Scientific Culture and the Making of the Industrial West. Oxford, UK: Oxford University Press; 1997. [Google Scholar]
- 170. Brady LM. War Upon the Land: Military Strategy and the Transformation of Southern Landscapes During the American Civil War. Athens, GA: University of Georgia Press; 2012. [Google Scholar]
- 171. Fiege M, Cronon W. The Republic of Nature: An Environmental History of the United States. Washington, DC: University of Washington Press; 2012. [Google Scholar]
- 172. Kiple KF, King VH. Another Dimension to the Black Diaspora: Diet, Disease and Racism. Cambridge, UK: Cambridge University Press; 2003. [Google Scholar]
- 173. Whitaker JW. Feedlot Empire: Beef Cattle Feeding in Illinois and Iowa 1840–1900. Iowa City, IA: Iowa State University Press; 1975. [Google Scholar]
- 174. White R. The Republic for Which it Stands: The United States During Reconstruction and the Gilded Age, 1865–1896. Oxford, UK: Oxford University Press; 2017. [Google Scholar]
- 175. Livi-Bacci M. Fertility, nutrition, and pellagra: Italy during the vital revolution. J Interdiscip Hist. 1986;16:431–454. [PubMed] [Google Scholar]
- 176. Heiser C. Seed to Civilization: The Story of Food. Cambridge, MA: Harvard University Press; 2013. [Google Scholar]
- 177. Rixson D. The History of Meat Trading. Nottingham, UK: Nottingham University Press; 2000. [Google Scholar]
- 178. Crafts NF. Industrial revolution in England and France: some thoughts on the question, ‘Why was England first?’ Econom History Rev. 1977;30:429–441. [Google Scholar]
- 179. Rosenberg NL, Birdzell LE., Jr. How the West Grew Rich: The Economic Transformation of the Industrial World. New York, NY: Basic Books; 2008. [Google Scholar]
- 180. Williams AC, Hill LJ. Meat and nicotinamide: a causal role in human evolution, history, and demographics. Int J Tryptophan Res. 2017;10:1178646917704661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181. Studer R. The Great Divergence Reconsidered. Cambridge, UK: Cambridge University Press; 2015. [Google Scholar]
- 182. Henley D. Asia-Africa Development Divergence: A Question of Intent. London, England: Zed Books; 2015. [Google Scholar]
- 183. Chubaty AM, Ma BO, Stein RW, et al. On the evolution of omnivory in a community context. Ecol Evol. 2014;4:251–265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184. Sebastian D. The evolution and maintenance of omnivory: dynamic constraints and the role of food quality. Ecology. 2003;84:2557–2567. [Google Scholar]
- 185. Amsden AH. Escape From Empire: The Developing World’s Journey Through Heaven and Hell. Cambridge, MA: The MIT Press; 2009. [Google Scholar]
- 186. Dore R. Land Reform in Japan. New York, NY: Bloomsbury; 2013. [Google Scholar]
- 187. Herring RJ. Land to the Tiller: The Political Economy of Agrarian Reform in South Asia. New Haven, CT: Yale University Press; 1983. [Google Scholar]
- 188. Kroeber AR. China’s Economy: What Everyone Needs to Know®. Oxford, UK: Oxford University Press; 2016. [Google Scholar]
- 189. Kuo SWY, Fei JCH, Ranis G. The Taiwan Success Story: Rapid Growth with Improved Distribution in the Republic of China, 1952–1979. Boulder, CO: Westview Press; 1981. [Google Scholar]
- 190. Studwell J. How Asia Works: Success and Failure in the World’s Most Dynamic Region. New York, NY: Grove Atlantic; 2013. [Google Scholar]
- 191. Lehmann D. Peasants, Landlords, and Governments: Agrarian Reform in the Third World. Teaneck, NJ: Holmes & Meier; 1974. [Google Scholar]
- 192. Li K-T. Economic Transformation of Taiwan, ROC. London, England: Shepheard-Walwyn; 1988. [Google Scholar]
- 193. Lipton M. Why Poor People Stay Poor: Urban Bias in World Development. London, England: Avebury; 1989. [Google Scholar]
- 194. Lipton M. Land Reform in Developing Countries: Property Rights and Property Wrongs. Abingdon, UK: Taylor & Francis; 2009. [Google Scholar]
- 195. Minami R. The Economic Development of Japan: A Quantitative Study. Berlin, Germany: Springer; 1986. [Google Scholar]
- 196. Smith K. The Malthusian Controversy. Abingdon, UK: Taylor & Francis; 2013. [Google Scholar]
- 197. Spengler JJ. Was Malthus right? South Econom J. 1966;33:17–34. [Google Scholar]
- 198. Abuhammad A. Cholesterol metabolism: a potential therapeutic target in Mycobacteria. Br J Pharmacol. 2017;174:2194–2208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199. Brace PT, Tezera LB, Bielecka MK, et al. Mycobacterium tuberculosis subverts negative regulatory pathways in human macrophages to drive immunopathology. PLoS Pathog. 2017;13:e1006367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200. Casterline J. Demographic transition. In: Demeny P, McNicoll G, eds. Encyclopedia of Population. New York, NY: Macmillan; 2003:210–216. [Google Scholar]
- 201. Cleland J, Wilson C. Demand theories of the fertility transition: an iconoclastic view. Populat Stud. 1987;41:5–30. [Google Scholar]
- 202. Duda P, Zrzavy J. Human population history revealed by a supertree approach. Sci Rep. 2016;6:29890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203. Gaskins AJ, Chavarro JE. Diet and fertility: a review. Am J Obstet Gynecol. 2017;218:379–389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204. Kim HL, Ratan A, Perry GH, Montenegro A, Miller W, Schuster SC. Khoisan hunter-gatherers have been the largest population throughout most of modern-human demographic history. Nat Commun. 2014;5:5692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205. Lawson DW, Borgerhoff Mulder M. The offspring quantity-quality trade-off and human fertility variation. Philos Trans R Soc Lond B Biol Sci. 2016;371:20150145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206. Lee CT, Puleston CO, Tuljapurkar S. Population and prehistory III: food-dependent demography in variable environments. Theor Popul Biol. 2009;76:179–188. [DOI] [PubMed] [Google Scholar]
- 207. Lee CT, Tuljapurkar S. Population and prehistory I: food-dependent population growth in constant environments. Theor Popul Biol. 2008;73:473–482. [DOI] [PubMed] [Google Scholar]
- 208. van de Kaa DJ. The idea of a second demographic transition in industrialized countries. Birth. 2002;35:45. [Google Scholar]
- 209. Zaidi B, Morgan SP. The second demographic transition: a review and appraisal. Ann Rev Sociol. 2017;43:473–492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210. Cipolla CM. Before the Industrial Revolution: European Society and Economy 1000–1700. Abingdon, UK: Taylor & Francis; 2004. [Google Scholar]
- 211. Macfarlane A. The Savage Wars of Peace: England, Japan and the Malthusian Trap. Abingdon, UK: Palgrave Macmillan; 2002. [Google Scholar]
- 212. Ammerman AJ, Cavalli-Sforza LL. The Neolithic Transition and the Genetics of Populations in Europe. Princeton, NJ: Princeton University Press; 2014. [Google Scholar]
- 213. Bocquet-Appel JP, Bar-Yosef O. The Neolithic Demographic Transition and Its Consequences. Dordrecht, The Netherlands: Springer; 2008. [Google Scholar]
- 214. Cassen R. India, Population, Economy, Society. Teaneck, NJ: Holmes & Meier; 1978. [Google Scholar]
- 215. Griffith GT. Population Problems of the Age of Malthus. Cambridge, UK: Cambridge University Press; 2010. [Google Scholar]
- 216. Langer WL. American foods and Europe’s population growth 1750–1850. J Soc History. 1975;8:51–66. [Google Scholar]
- 217. Soloway RA. Birth Control and the Population Question in England, 1877–1930. Chapel Hill, NC: University of North Carolina Press; 2011. [Google Scholar]
- 218. Soloway RA. Demography and Degeneration: Eugenics and the Declining Birthrate in Twentieth-Century Britain. Chapel Hill, NC: University of North Carolina Press; 2014. [Google Scholar]
- 219. Turchin P. Complex Population Dynamics: A Theoretical/Empirical Synthesis (MPB-35). Princeton, NJ: Princeton University Press; 2013. [Google Scholar]
- 220. Ward RH, Weiss KM. The Demographic Evolution of Human Populations. Cambridge, MA: Academic Press; 1976. [Google Scholar]
- 221. Afeiche MC, Gaskins AJ, Williams PL, et al. Processed meat intake is unfavorably and fish intake favorably associated with semen quality indicators among men attending a fertility clinic. J Nutr. 2014;144:1091–1098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222. Burger O, DeLong JP. What if fertility decline is not permanent? The need for an evolutionarily informed approach to understanding low fertility. Phil Trans R Soc B. 2016;371:20150157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223. Garruti G, De Palo R, De Angelisc M. Weighing the impact of diet and lifestyle on female reproductive function. Curr Med Chem. 2017;24:1–9. [DOI] [PubMed] [Google Scholar]
- 224. Hohos NM, Skaznik-Wikiel ME. High fat diet and female fertility. Endocrinology. 2017;158:2407–2419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225. Lee R. The outlook for population growth. Science. 2011;333:569–573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226. Myrskyla M, Kohler HP, Billari FC. Advances in development reverse fertility declines. Nature. 2009;460:741. [DOI] [PubMed] [Google Scholar]
- 227. Cobbett L. The decline of tuberculosis and the increase in its mortality during the war. J Hyg (Lond). 1930;30:79–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228. Comas I, Hailu E, Kiros T, et al. Population genomics of Mycobacterium tuberculosis in Ethiopia contradicts the virgin soil hypothesis for human tuberculosis in Sub-Saharan Africa. Curr Biol. 2015;25:3260–3266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229. Coscolla M, Copin R, Sutherland J, et al. M. tuberculosis T cell epitope analysis reveals paucity of antigenic variation and identifies rare variable TB antigens. Cell Host Microbe. 2015;18:538–548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230. McKeown T, Record R, Turner R. An interpretation of the decline of mortality in England and Wales during the twentieth century. Popul Stud (Camb). 1975;29:391–422. [PubMed] [Google Scholar]
- 231. Ouellet H, Johnston JB, de Montellano PR. Cholesterol catabolism as a therapeutic target in Mycobacterium tuberculosis. Trends Microbiol. 2011;19:530–539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232. Szreter S. The importance of social intervention in Britain’s mortality decline c. 1850–1914: a re-interpretation of the role of public health. Soc History Med. 1988;1:1–38. [Google Scholar]
- 233. Tripp L, Sawchuk LA. Insights into secular trends of respiratory tuberculosis: the 20th century Maltese experience. PLoS ONE. 2017;12:e0183296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234. Cheng CY, Gutierrez NM, Marzuki MB, et al. Host sirtuin 1 regulates mycobacterial immunopathogenesis and represents a therapeutic target against tuberculosis. Sci Immunol. 2017;2:eaaj1789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235. O’Garra A, Redford PS, McNab FW, Bloom CI, Wilkinson RJ, Berry MP. The immune response in tuberculosis. Ann Rev Immunol. 2013;31:475–527. [DOI] [PubMed] [Google Scholar]
- 236. Wallis RS, Hafner R. Advancing host-directed therapy for tuberculosis. Nat Rev Immunol. 2015;15:255. [DOI] [PubMed] [Google Scholar]
- 237. Beddoes T. Essay on the Causes, Early Signs, and Prevention of Pulmonary Consumption: For the Use of Parents and Preceptors. London, England; Bristol, UK: Longman and Rees; W. Sheppard; 1799. [Google Scholar]
- 238. Carter LM, Pollitt LC, Wilson LG, Reece SE. Ecological influences on the behaviour and fertility of malaria parasites. Malar J. 2016;15:220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239. Kebede A, McCann JC, Kiszewski AE, Ye-Ebiyo Y. New evidence of the effects of agro-ecologic change on malaria transmission. Am J Trop Med Hyg. 2005;73:676–680. [PubMed] [Google Scholar]
- 240. Njagi EN, Bender DA. Schistosoma mansoni: effects on tryptophan metabolism in mice. Exp Parasitol. 1990;70:43–54. [DOI] [PubMed] [Google Scholar]
- 241. Oliver SV, Brooke BD. The effect of larval nutritional deprivation on the life history and DDT resistance phenotype in laboratory strains of the malaria vector Anopheles arabiensis. Malar J. 2013;12:44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242. Wondwosen B, Hill SR, Birgersson G, Seyoum E, Tekie H, Ignell R. A (maize) ing attraction: gravid Anopheles arabiensis are attracted and oviposit in response to maize pollen odours. Malar J. 2017;16:39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243. Qiu S, Fan X, Yang Y, et al. Schistosoma japonicum infection downregulates house dust mite-induced allergic airway inflammation in mice. PLoS ONE. 2017;12:e0179565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244. Tcherniuk SO, Chesnokova O, Oleinikov IV, Oleinikov AV. Nicotinamide inhibits the growth of P. falciparum and enhances the antimalarial effect of artemisinin, chloroquine and pyrimethamine. Mol Biochem Parasitol. 2017;216:14–20. [DOI] [PubMed] [Google Scholar]
- 245. Hayashi T, Mo J-H, Gong X, et al. 3-Hydroxyanthranilic acid inhibits PDK1 activation and suppresses experimental asthma by inducing T cell apoptosis. Proc Natl Acad Sci U S A. 2007;104:18619–18624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246. Wu H, Gong J, Liu Y. Indoleamine 2, 3-dioxygenase regulation of immune response (Review). Mol Med Rep. 2018;17:4867–4873. [DOI] [PubMed] [Google Scholar]
- 247. Andersen MH, ed. Anti-regulatory T cells. Semin Immunopathol. 2017;39:317–326. [DOI] [PubMed] [Google Scholar]
- 248. Rodriguez CBH, Vasudevan A, Elkhal A. Aspects of tryptophan and nicotinamide adenine dinucleotide in immunity: a new twist in an old tale. Int J Tryptophan Res. 2017;10:1178646917713491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249. Bobosha K, Wilson L, van Meijgaarden KE, et al. T-cell regulation in lepromatous leprosy. Plos Negl Trop Dis. 2014;8:e2773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250. Danikowski K, Jayaraman S, Prabhakar B. Regulatory T cells in multiple sclerosis and myasthenia gravis. J Neuroinflammation. 2017;14:117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251. de Araujo EF, Feriotti C, Galdino NAL, Preite NW, Calich VLG, Loures FV. The IDO*AhR axis controls Th17/Treg immunity in a pulmonary model of fungal infection. Front Immunol. 2017;8:880. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252. Gaelings L, Soderholm S, Bugai A, et al. Regulation of kynurenine biosynthesis during influenza virus infection. FEBS J. 2017;284:222–236. [DOI] [PubMed] [Google Scholar]
- 253. Grant RS, Passey R, Matanovic G, Smythe G, Kapoor V. Evidence for increased de novo synthesis of NAD in immune-activated RAW264. Arch Biochem Biophys. 1999;372:1–7. [DOI] [PubMed] [Google Scholar]
- 254. Kugelberg E. Tryptophan triggers tranquillity. Nat Rev Immunol. 2016;16:338–339. [DOI] [PubMed] [Google Scholar]
- 255. Quintana FJ, Sherr DH. Aryl hydrocarbon receptor control of adaptive immunity. Pharmacol Rev. 2013;65:1148–1161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256. Stockinger B, Di Meglio P, Gialitakis M, Duarte JH. The aryl hydrocarbon receptor: multitasking in the immune system. Annu Rev Immunol. 2014;32:403–432. [DOI] [PubMed] [Google Scholar]
- 257. Wang Q, Liu D, Song P, Zou M-H. Tryptophan-kynurenine pathway is dysregulated in inflammation, and immune activation. Front Biosci (Landmark Ed). 2015;20:1116–1143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258. Xu T, Stewart KM, Wang X, et al. Metabolic control of TH17 and induced Treg cell balance by an epigenetic mechanism. Nature. 2017;548:228–233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259. Yu L, Zhang Y, Zhao F. Expression of indoleamine 2, 3-dioxygenase in pregnant mice correlates with CD4+ CD25+ Foxp3+ T regulatory cells. Eur Rev Med Pharmacol Sci. 2017;21:1722–1728. [PubMed] [Google Scholar]
- 260. Samstein RM, Josefowicz SZ, Arvey A, Treuting PM, Rudensky AY. Extrathymic generation of regulatory T cells in placental mammals mitigates maternal-fetal conflict. Cell. 2012;150:29–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261. Berer K, Gerdes LA, Cekanaviciute E, et al. Gut microbiota from multiple sclerosis patients enables spontaneous autoimmune encephalomyelitis in mice. Proc Natl Acad Sci U S A. 2017;114:10719–10724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262. Caballero-Villarraso J, Galvan A, Escribano B, Túnez I. Interrelationships among gut microbiota and host: paradigms, role in neurodegenerative diseases and future prospects. CNS Neurol Disord Drug Targets. 2017;16:945–964. [DOI] [PubMed] [Google Scholar]
- 263. Hindson J. Multiple sclerosis: a possible link between multiple sclerosis and gut microbiota. Nat Rev Neurol. 2017;13:705. [DOI] [PubMed] [Google Scholar]
- 264. Pimentel D, Marcia H, Pimentel MS. Food, Energy, and Society. 3rd ed. Boca Raton, FL: CRC Press; 2007. [Google Scholar]
- 265. Ban Y, Chang Y, Dong B, Kong B, Qu X. Indoleamine 2, 3-dioxygenase levels at the normal and recurrent spontaneous abortion fetal–maternal interface. J Int Med Res. 2013;41:1135–1149. [DOI] [PubMed] [Google Scholar]
- 266. Kudo Y, Boyd C, Sargent IL, Redman CW. Decreased tryptophan catabolism by placental indoleamine 2, 3-dioxygenase in preeclampsia. Am J Obstet Gynecol. 2003;188:719–726. [DOI] [PubMed] [Google Scholar]
- 267. Somerset DA, Zheng Y, Kilby MD, Sansom DM, Drayson MT. Normal human pregnancy is associated with an elevation in the immune suppressive CD25+ CD4+ regulatory T-cell subset. Immunology. 2004;112:38–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268. Yeung AW, Terentis AC, King NJ, Thomas SR. Role of indoleamine 2, 3-dioxygenase in health and disease. Clin Sci (Lond). 2015;129:601–672. [DOI] [PubMed] [Google Scholar]
- 269. Zong S, Li C, Luo C, et al. Dysregulated expression of IDO may cause unexplained recurrent spontaneous abortion through suppression of trophoblast cell proliferation and migration. Sci Rep. 2016;6:19916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 270. Itoh A, Ridgway WM. Targeting innate immunity to downmodulate adaptive immunity and reverse type 1 diabetes. Immunotargets Ther. 2017;6:31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271. Kwon H-S, Lim HW, Wu J, Schnolzer M, Verdin E, Ott M. Three novel acetylation sites in the Foxp3 transcription factor regulate the suppressive activity of regulatory T cells. J Immunol. 2012;188:2712–2721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272. Lim HW, Kang SG, Ryu JK, et al. SIRT1 deacetylates RORγt and enhances Th17 cell generation. J Exp Med. 2015;212:607–617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 273. van Loosdregt J, Vercoulen Y, Guichelaar T, et al. Regulation of Treg functionality by acetylation-mediated Foxp3 protein stabilization. Blood. 2010;115:965–974. [DOI] [PubMed] [Google Scholar]
- 274. Kipnis E, Dessein R. Bacterial modulation of Tregs/Th17 in intestinal disease: a balancing act. Inflamm Bowel Dis. 2012;18:1389–1390. [DOI] [PubMed] [Google Scholar]
- 275. Salazar F, Awuah D, Negm OH, Shakib F, Ghaemmaghami AM. The role of indoleamine 2, 3-dioxygenase-aryl hydrocarbon receptor pathway in the TLR4-induced tolerogenic phenotype in human DCs. Sci Rep. 2017;7:43337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 276. Allan SE, Passerini L, Bacchetta R, et al. The role of 2 FOXP3 isoforms in the generation of human CD4+ Tregs. J Clin Invest. 2005;115:3276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277. Chen X, Oppenheim JJ. Th17 cells and Tregs: unlikely allies. J Leukoc Biol. 2014;95:723–731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278. Mezrich JD, Fechner JH, Zhang X, Johnson BP, Burlingham WJ, Bradfield CA. An interaction between kynurenine and the aryl hydrocarbon receptor can generate regulatory T cells. J Immunol. 2010;185:3190–3198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 279. Palomares O, Akdis M, Martin-Fontecha M, Akdis CA. Mechanisms of immune regulation in allergic diseases: the role of regulatory T and B cells. Immunol Rev. 2017;278:219–236. [DOI] [PubMed] [Google Scholar]
- 280. Pan F, Fan H, Lu L, Liu Z, Jiang S. The yin and yang of signaling in Tregs and TH17 cells. Sci Signal. 2011;4:mr4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281. Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol. 1995;155:1151–1164. [PubMed] [Google Scholar]
- 282. Sakaguchi S, Takahashi T, Nishizuka Y. Study on cellular events in post-thymectomy autoimmune oophoritis in mice. II. Requirement of Lyt-1 cells in normal female mice for the prevention of oophoritis. J Exp Med. 1982;156:1577–1586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283. Sakaguchi S, Takahashi T, Nishizuka Y. Study on cellular events in postthymectomy autoimmune oophoritis in mice. I. Requirement of Lyt-1 effector cells for oocytes damage after adoptive transfer. J Exp Med. 1982;156:1565–1576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 284. Tang Q, Vincenti F. Transplant trials with Tregs: perils and promises. J Clin Invest. 2017;127:2505–2512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 285. Beetham W, Jr, Fischer S, Schrohenloher R. Tryptophan metabolite excretion in connective tissue diseases demonstrating a difference between rheumatoid spondylitis and rheumatoid arthritis. Proc Soc Exp Biol Med. 1964;117:756–759. [DOI] [PubMed] [Google Scholar]
- 286. Versini M, Jeandel PY, Bashi T, Bizzaro G, Blank M, Shoenfeld Y. Unraveling the Hygiene Hypothesis of helminthes and autoimmunity: origins, pathophysiology, and clinical applications. BMC Med. 2015;13:81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 287. Tullius SG, Biefer HRC, Li S, et al. NAD+ protects against EAE by regulating CD4+ T-cell differentiation. Nat Commun. 2014;5:5101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 288. Ibrahim T, Przybyl L, Harmon AC, et al. Proliferation of endogenous regulatory T cells improve the pathophysiology associated with placental ischaemia of pregnancy. Am J Reprod Immunol. 2017;78:12724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 289. Halim L, Romano M, McGregor R, et al. An atlas of human regulatory T helper-like cells reveals features of Th2-like Tregs that support a tumorigenic environment. Cell Rep. 2017;20:757–770. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 290. Hayakawa S, Ohno N, Okada S, Kobayashi M. Significant augmentation of regulatory T cell numbers occurs during the early neonatal period. Clin Exp Immunol. 2017;190:268–279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 291. Briggs N, Weatherhead J, Sastry KJ, Hotez PJ. The hygiene hypothesis and its inconvenient truths about helminth infections. PLoS Negl Trop Dis. 2016;10:e0004944. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 292. Caraballo L. The tropics, helminth infections and hygiene hypotheses. Expert Rev Clin Immunol. 2018;14:99–102. [DOI] [PubMed] [Google Scholar]
- 293. Gerrard JW, Geddes CA, Reggin PL, Gerrard CD, Horne S. Serum IgE levels in white and Metis communities in Saskatchewan. Ann Allergy. 1976;37:91–100. [PubMed] [Google Scholar]
- 294. Greenwood BM. Autoimmune disease and parasitic infections in Nigerians. Lancet. 1968;2:380–382. [DOI] [PubMed] [Google Scholar]
- 295. Lambrecht BN, Hammad H. The immunology of the allergy epidemic and the hygiene hypothesis. Nat Immunol. 2017;18:1076–1083. [DOI] [PubMed] [Google Scholar]
- 296. Rook GA, Martinelli R, Brunet LR. Innate immune responses to mycobacteria and the downregulation of atopic responses. Curr Opin Allergy Clin Immunol. 2003;3:337–342. [DOI] [PubMed] [Google Scholar]
- 297. Santiago HC, Nutman TB. Human helminths and allergic disease: the hygiene hypothesis and beyond. Am J Trop Med Hyg. 2016;95:746–753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 298. Strachan DP. Hay fever, hygiene, and household size. BMJ. 1989;299:1259–1260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 299. Foster KR, Schluter J, Coyte KZ, Rakoff-Nahoum S. The evolution of the host microbiome as an ecosystem on a leash. Nature. 2017;548:43–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 300. Platts-Mills TA. The allergy epidemics: 1870–2010. J Allergy Clin Immunol. 2015;136:3–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 301. Afzal M, Kuipers OP, Shafeeq S. Niacin-mediated gene expression and role of NiaR as a transcriptional repressor of niaX, nadC, and pnuC in Streptococcus pneumoniae. Front Cell Infect Microbiol. 2017;7:70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 302. Townsend MJ, Monroe JG, Chan AC. B-cell targeted therapies in human autoimmune diseases: an updated perspective. Immunol Rev. 2010;237:264–283. [DOI] [PubMed] [Google Scholar]
- 303. Boyd-Orr JB, Gilks JL. Studies of Nutrition: The Physique and Health of Two African Tribes. Richmond, UK: H.M. Stationery Office; 1931. [Google Scholar]
- 304. McCarrison R. Nutrition and National Health: Being the Cantor Lectures Delivered Before The Royal Society of Arts 1936. London, England: Faber and Faber; 1944. [Google Scholar]
- 305. Collingham L. The Hungry Empire: How Britain’s Quest for Food Shaped the Modern World. New York, NY: Random House; 2017. [Google Scholar]
- 306. Stone R. Public enemy number one. Science. 2013;340:422–425. [DOI] [PubMed] [Google Scholar]
- 307. Gontzea I. Nutrition and Anti-infectious Defence. Basel, Switzerland: Karger; 1974. [Google Scholar]
- 308. Scrimshaw NS, Taylor CE, Gordon JE; World Health Organization. Interactions of Nutrition and Infection. Geneva, Switzerland: World Health Organization; 1968. [PubMed] [Google Scholar]
- 309. Dubos RJ. Mirage of Health: Utopias, Progress, and Biological Change. New Brunswick, NJ: Rutgers University Press; 1987. [Google Scholar]
- 310. Barry JM. The site of origin of the 1918 influenza pandemic and its public health implications. J Transl Med. 2004;2:3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 311. Dubos RJ, Dubos J. The White Plague: Tuberculosis, Man, and Society. New Brunswick, NJ: Rutgers University Press; 1952. [Google Scholar]
- 312. Editors CR, River C. The 1918 Spanish Flu Pandemic: The History and Legacy of the Worlds Deadliest Influenza Outbreak. Scotts Valley, CA: CreateSpace; 2017. [Google Scholar]
- 313. Garrett L. The Coming Plague: Newly Emerging Diseases in a World out of Balance. New York, NY: Farrar, Straus and Giroux; 1994. [Google Scholar]
- 314. Lederberg J, Shope RE, Oaks SC; Committee on Emerging Microbial Threats to Health. Emerging Infections: Microbial Threats to Health in the United States. Washington, DC: National Academies Press; 1992. [PubMed] [Google Scholar]
- 315. Kmietowicz Z. Pneumonic plague outbreak hits cities in Madagascar. BMJ. 2017;359:j4595. [DOI] [PubMed] [Google Scholar]
- 316. Krause RM; National Foundation for Infectious Diseases. The Restless Tide: The Persistent Challenge of the Microbial World. Bethesda, MD: National Foundation for Infectious Diseases; 1981. [Google Scholar]
- 317. Rawcliffe C. Leprosy in Medieval England. Lowestoft, UK: Boydell Press; 2009. [Google Scholar]
- 318. von Seidlein L, Sack D, Azman AS, Ivers LC, Lopez AL, Deen JL. Cholera outbreak in Yemen. Lancet Gastroenterol Hepatol. 2017;2:777. [DOI] [PubMed] [Google Scholar]
- 319. Delisle HF. Poverty: the double burden of malnutrition in mothers and the intergenerational impact. Ann N Y Acad Sci. 2008;1136:172–184. [DOI] [PubMed] [Google Scholar]
- 320. Harrison GG. Public health interventions to combat micronutrient deficiencies. Public Health Reviews. 2010;32:256–266. [Google Scholar]
- 321. Worboys M. The discovery of colonial malnutrition between the wars. In: Arnold D, ed. Imperial Medicine and Indigenous Societies. Manchester, UK: Manchester University Press; 1988:208–225. [Google Scholar]
- 322. Clare L, Wu Y-T, Teale JC, et al. Potentially modifiable lifestyle factors, cognitive reserve, and cognitive function in later life: a cross-sectional study. PLoS Med. 2017;14:e1002259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 323. Jones DS, Greene JA. Is dementia in decline? Historical trends and future trajectories. N Engl J Med. 2016;374:507–509. [DOI] [PubMed] [Google Scholar]
- 324. Ritchie S. Intelligence: All That Matters. London, England: Hodder & Stoughton; 2015. [Google Scholar]
- 325. Ritchie SJ, Bates TC, Deary IJ. Is education associated with improvements in general cognitive ability, or in specific skills? Develop Psychol. 2015;51:573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 326. Stern Y. Cognitive reserve in ageing and Alzheimer’s disease. Lancet Neurol. 2012;11:1006–1012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 327. Avalos JL, Bever KM, Wolberger C. Mechanism of sirtuin inhibition by nicotinamide: altering the NAD+ cosubstrate specificity of a Sir2 enzyme. Mol Cell. 2005;17:855–868. [DOI] [PubMed] [Google Scholar]
- 328. Bonkowski MS, Sinclair DA. Slowing ageing by design: the rise of NAD+ and sirtuin-activating compounds. Nat Rev Mol Cell Biol. 2016;17:679–690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 329. Pissios P. Nicotinamide N-methyltransferase: more than a vitamin B3 clearance enzyme. Trends Endocrinol Metab. 2017;28:340–353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 330. Schmeisser K, Mansfeld J, Kuhlow D, et al. Role of sirtuins in lifespan regulation is linked to methylation of nicotinamide. Nat Chem Biol. 2013;9:693–700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 331. Bitterman KJ, Anderson RM, Cohen HY, Latorre-Esteves M, Sinclair DA. Inhibition of silencing and accelerated aging by nicotinamide, a putative negative regulator of yeast sir2 and human SIRT1. J Biol Chem. 2002;277:45099–45107. [DOI] [PubMed] [Google Scholar]
- 332. Garrido A, Djouder N. NAD+ deficits in age-related diseases and cancer. Trends Cancer. 2017;3:593–610. [DOI] [PubMed] [Google Scholar]
- 333. Gaur U, Tu J, Li D, et al. Molecular evolutionary patterns of NAD+/Sirtuin aging signaling pathway across taxa. PLoS ONE. 2017;12:e0182306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 334. Pehar M, Harlan BA, Killoy KM, Vargas MR. Nicotinamide adenine dinucleotide (NAD+) metabolism and neurodegeneration. Antioxid Redox Signal. 2018;28:1652–1668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 335. Watroba M, Dudek I, Skoda M, Stangret A, Rzodkiewicz P, Szukiewicz D. Sirtuins, epigenetics and longevity. Ageing Res Rev. 2017;40:11–19. [DOI] [PubMed] [Google Scholar]
- 336. Bipath P, Levay PF, Viljoen M. Tryptophan depletion in context of the inflammatory and general nutritional status of a low-income South African HIV-infected population. J Health Popul Nutr. 2016;35:5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 337. Hou Y, Lautrup S, Cordonnier S, et al. NAD+ supplementation normalizes key Alzheimer’s features and DNA damage responses in a new AD mouse model with introduced DNA repair deficiency. Proc Natl Acad Sci U S A. 2018:E1876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 338. Saini JS, Corneo B, Miller JD, et al. Nicotinamide ameliorates disease phenotypes in a human iPSC model of age-related macular degeneration. Cell Stem Cell. 2017;20:635.e7–647.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 339. Saint-Geniez M, Rosales MAB. Eyeing the fountain of youth. Cell Stem Cell. 2017;20:583–584. [DOI] [PubMed] [Google Scholar]
- 340. Liebmann JM, Cioffi GA. Nicking glaucoma with nicotinamide. N Engl J Med. 2017;376:2079–2081. [DOI] [PubMed] [Google Scholar]
- 341. Diani-Moore S, Shoots J, Singh R, Zuk JB, Rifkind AB. NAD+ loss, a new player in AhR biology: prevention of thymus atrophy and hepatosteatosis by NAD+ repletion. Sci Rep. 2017;7:2268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 342. Elhassan YS, Philp AA, Lavery GG. Targeting NAD+ in metabolic disease; new insights into an old molecule. J Endocr Soc. 2017;1:816–835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 343. Fletcher RS, Ratajczak J, Doig CL, et al. Nicotinamide riboside kinases display redundancy in mediating nicotinamide mononucleotide and nicotinamide riboside metabolism in skeletal muscle cells. Mol Metab. 2017;6:819–832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 344. Huang Q, Sun M, Li M, et al. Combination of NAD+ and NADPH offers greater neuroprotection in ischemic stroke models by relieving metabolic stress. Mol Neurobiol. 2018;55:6063–6075. [DOI] [PubMed] [Google Scholar]
- 345. Liu J, Yang B, Zhou P, et al. Nicotinamide adenine dinucleotide suppresses epileptogenesis at an early stage. Sci Rep. 2017;7:7321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 346. Perez-Lobos R, Lespay-Rebolledo C, Tapia-Bustos A, et al. Vulnerability to a metabolic challenge following perinatal asphyxia evaluated by organotypic cultures: neonatal nicotinamide treatment. Neurotox Res. 2017;32:426–443. [DOI] [PubMed] [Google Scholar]
- 347. Shi H, Enriquez A, Rapadas M, et al. NAD deficiency, congenital malformations, and niacin supplementation. N Engl J Med. 2017;377:544–552. [DOI] [PubMed] [Google Scholar]
- 348. Vaur P, Brugg B, Mericskay M, et al. Nicotinamide riboside, a form of vitamin B3, protects against excitotoxicity-induced axonal degeneration. FASEB J. 2017;31:5440–5452. [DOI] [PubMed] [Google Scholar]
- 349. Wang X, Zhang Q, Bao R, et al. Deletion of Nampt in projection neurons of adult mice leads to motor dysfunction, neurodegeneration, and death. Cell Rep. 2017;20:2184–2200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 350. Wei C, Kong Y, Li G, et al. NAD replenishment with nicotinamide mononucleotide protects blood-brain barrier integrity and attenuates delayed tPA-induced haemorrhagic transformation after cerebral ischemia. Br J Pharmacol. 2017;174:3823–3836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 351. Westenskow PD. Nicotinamide: a novel treatment for age-related macular degeneration? Stem Cell Investig. 2017;4;86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 352. Braidy N, Grant R, Sachdev PS. Nicotinamide adenine dinucleotide and its related precursors for the treatment of Alzheimer’s disease. Curr Opin Psychiatry. 2018;31:160–166. [DOI] [PubMed] [Google Scholar]
- 353. Cao B, Sun X-Y, Zhang C-B, et al. Association between B vitamins and schizophrenia: a population-based case-control study. Psychiatry Res. 2018;259:501–505. [DOI] [PubMed] [Google Scholar]
- 354. Lanz TV, Williams SK, Stojic A, et al. Tryptophan-2,3-dioxygenase (TDO) deficiency is associated with subclinical neuroprotection in a mouse model of multiple sclerosis. Sci Rep. 2017;7:41271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 355. Merlo LM, Grabler S, DuHadaway JB, et al. Therapeutic antibody targeting of indoleamine-2, 3-dioxygenase (IDO2) inhibits autoimmune arthritis. Clin Immunol. 2017;179:8–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 356. Nejabati HR, Mihanfar A, Pezeshkian M, et al. N1-methylnicotinamide (MNAM) as a guardian of cardiovascular system. J Cell Physiol. 2018;233:6386–6394. [DOI] [PubMed] [Google Scholar]
- 357. Platten M, Litzenburger U, Wick W. The aryl hydrocarbon receptor in tumor immunity. Oncoimmunology. 2012;1:396–397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 358. Terness P, Bauer TM, Rose L, et al. Inhibition of allogeneic T cell proliferation by indoleamine 2, 3-dioxygenase-expressing dendritic cells. J Exp Med. 2002;196:447–457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 359. Shackelford RE, Mayhall K, Maxwell NM, Kandil E, Coppola D. Nicotinamide phosphoribosyltransferase in malignancy: a review. Genes Cancer. 2013;4:447–456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 360. Abram DM, Fernandes LGR, Ramos Filho ACS, Simioni PU. The modulation of enzyme indoleamine 2,3-dioxygenase from dendritic cells for the treatment of type 1 diabetes mellitus. Drug Des Devel Ther. 2017;11:2171–2178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 361. Crujeiras AB, Pissios P, Moreno-Navarrete JM, et al. An epigenetic signature in adipose tissue is linked to nicotinamide N-methyltransferase gene expression. Mol Nutr Food Res. 2018:e1700933. [DOI] [PubMed] [Google Scholar]
- 362. Dong G, Chen W, Wang X, et al. Small molecule inhibitors simultaneously targeting cancer metabolism and epigenetics: discovery of novel nicotinamide phosphoribosyltransferase (NAMPT) and histone deacetylase (HDAC) dual inhibitors. J Med Chem. 2017;60:7965–7983. [DOI] [PubMed] [Google Scholar]
- 363. Giuliante R, Sartini D, Bacchetti T, et al. Potential involvement of nicotinamide N-methyltransferase in the pathogenesis of metabolic syndrome. Metab Syndr Relat Disord. 2015;13:165–170. [DOI] [PubMed] [Google Scholar]
- 364. Kannt A, Pfenninger A, Teichert L, et al. Association of nicotinamide-N-methyltransferase mRNA expression in human adipose tissue and the plasma concentration of its product, 1-methylnicotinamide, with insulin resistance. Diabetologia. 2015;58:799–808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 365. Kraus D, Yang Q, Kong D, et al. Nicotinamide N-methyltransferase knockdown protects against diet-induced obesity. Nature. 2014;508:258–262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 366. Kroon T, Baccega T, Olsen A, Gabrielsson J, Oakes ND. Nicotinic acid timed to feeding reverses tissue lipid accumulation and improves glucose control in obese Zucker rats[S]. J Lipid Res. 2017;58:31–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 367. Lu X, Long H. Nicotinamide N-methyltransferase as a potential marker for cancer. Neoplasma. 2018;65:656–663. [DOI] [PubMed] [Google Scholar]
- 368. Neelakantan H, Vance V, Wetzel MD, et al. Selective and membrane-permeable small molecule inhibitors of nicotinamide N-methyltransferase reverse high fat diet-induced obesity in mice. Biochem Pharmacol. 2018;147:141–152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 369. Routy JP, Routy B, Graziani GM, Mehraj V. The kynurenine pathway is a double-edged sword in immune-privileged sites and in cancer: implications for immunotherapy. Int J Tryptophan Res. 2016;9:67–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 370. Rudolphi B, Zapp B, Kraus NA, Ehebauer F, Kraus BJ, Kraus D. Body weight predicts Nicotinamide N-Methyltransferase activity in mouse fat. Endocr Res. 2018;43:55–63. [DOI] [PubMed] [Google Scholar]
- 371. Strom K, Morales-Alamo D, Ottosson F, et al. N(1)-methylnicotinamide is a signalling molecule produced in skeletal muscle coordinating energy metabolism. Sci Rep. 2018;8:3016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 372. Uddin GM, Youngson NA, Doyle BM, Sinclair DA, Morris MJ. Nicotinamide mononucleotide (NMN) supplementation ameliorates the impact of maternal obesity in mice: comparison with exercise. Sci Rep. 2017;7:15063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 373. Zou XD, Guo SQ, Hu ZW, Li WL. NAMPT protects against 6-hydroxydopamine-induced neurotoxicity in PC12 cells through modulating SIRT1 activity. Mol Med Rep. 2016;13:4058–4064. [DOI] [PubMed] [Google Scholar]
- 374. Griffin SM, Pickard MR, Orme RP, Hawkins CP, Williams AC, Fricker RA. Nicotinamide alone accelerates the conversion of mouse embryonic stem cells into mature neuronal populations. PLoS ONE. 2017;12:e0183358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 375. Green S, Buttrum S, Molloy H, et al. N-methylation of pyridines in Parkinson’s disease. Lancet. 1991;338:120–121. [DOI] [PubMed] [Google Scholar]
- 376. Griffin SM, Pickard MR, Orme RP, Hawkins CP, Fricker RA. Nicotinamide promotes neuronal differentiation of mouse embryonic stem cells in vitro. Neuroreport. 2013;24:1041–1046. [DOI] [PubMed] [Google Scholar]
- 377. Parsons RB, Smith M-L, Williams AC, Waring RH, Ramsden DB. Expression of nicotinamide N-methyltransferase (EC 2.1. 1.1) in the Parkinsonian brain. J Neuropath Exp Neurol. 2002;61:111–124. [DOI] [PubMed] [Google Scholar]
- 378. Williams A, Ramsden D. Nicotinamide: a double edged sword. Parkinsonism Relat Disord. 2005;11:413–420. [DOI] [PubMed] [Google Scholar]
- 379. Lehmann S, Loh SH, Martins LM. Enhancing NAD+ salvage metabolism is neuroprotective in a PINK1 model of Parkinson’s disease. Biol Open. 2017;6:141–147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 380. Barber-Singh J, Seo BB, Nakamaru-Ogiso E, Lau YS, Matsuno-Yagi A, Yagi T. Neuroprotective effect of long-term NDI1 gene expression in a chronic mouse model of Parkinson disorder. Rejuvenation Res. 2009;12:259–267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 381. Lee J-Y, Ahn K, Jang BG, et al. Nicotinamide reduces dopamine in postnatal hypothalamus and causes dopamine-deficient phenotype. Neurosci Lett. 2009;461:163–166. [DOI] [PubMed] [Google Scholar]
- 382. De Lau LM, Breteler MM. Epidemiology of Parkinson’s disease. Lancet Neurol. 2006;5:525–535. [DOI] [PubMed] [Google Scholar]
- 383. Hong S, Zhai B, Pissios P. Nicotinamide N-methyltransferase interacts with enzymes of the methionine cycle and regulates methyl donor metabolism. Biochemistry. 2018;57:5775–5779. [DOI] [PubMed] [Google Scholar]
- 384. Muangpaisan W, Mathews A, Hori H, Seidel D. A systematic review of the worldwide prevalence and incidence of Parkinson’s disease. J Med Assoc Thai. 2011;94:749. [PubMed] [Google Scholar]
- 385. Pringsheim T, Jette N, Frolkis A, Steeves TD. The prevalence of Parkinson’s disease: a systematic review and meta-analysis. Movement Disord. 2014;29:1583–1590. [DOI] [PubMed] [Google Scholar]
- 386. Shin JH, Park CW, Yoon G, Hong SM, Choi KY. NNMT depletion contributes to liver cancer cell survival by enhancing autophagy under nutrient starvation. Oncogenesis. 2018;7:58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 387. Reich D. Who We Are and How We Got Here: Ancient DNA and the New Science of the Human Past. Oxford, UK: Oxford University Press; 2018. [Google Scholar]
- 388. Munn DH, Mellor AL. IDO in the tumor microenvironment: inflammation, counter-regulation, and tolerance. Trends Immunol. 2016;37:193–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 389. Carter AJ, Nguyen AQ. Antagonistic pleiotropy as a widespread mechanism for the maintenance of polymorphic disease alleles. BMC Med Genet. 2011;12:160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 390. Gagnon A. Natural fertility and longevity. Fertil Steril. 2015;103:1109–1116. [DOI] [PubMed] [Google Scholar]
- 391. Page AE, Chaudhary N, Viguier S, et al. Hunter-gatherer social networks and reproductive success. Sci Rep. 2017;7:1153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 392. Shokeir MH. Investigation on Huntington’s disease in the Canadian Prairies. II. Fecundity and fitness. Clin Genet. 1975;7:349–353. [DOI] [PubMed] [Google Scholar]
- 393. Daum H, Peretz T, Laufer N. BRCA mutations and reproduction. Fertil Steril. 2018;109:33–38. [DOI] [PubMed] [Google Scholar]
- 394. Hageman GJ, Stierum RH. Niacin, poly(ADP-ribose) polymerase-1 and genomic stability. Mutat Res. 2001;475:45–56. [DOI] [PubMed] [Google Scholar]
- 395. Kwiatkowski F, Arbre M, Bidet Y, Laquet C, Uhrhammer N, Bignon YJ. BRCA mutations increase fertility in families at hereditary breast/ovarian cancer risk. PLoS ONE. 2015;10:e0127363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 396. Lohani M, Dhasmana A, Haque S, et al. Niacin deficiency modulates genes involved in cancer: are smokers at higher risk? J Cell Biochem. 2019;120:232–242. [DOI] [PubMed] [Google Scholar]
- 397. Motegi A, Masutani M, Yoshioka KI, Bessho T. Aberrations in DNA repair pathways in cancer and therapeutic significances [published online ahead of print March 25, 2019]. Semin Cancer Biol. doi: 10.1016/j.semcancer.2019.02.005. [DOI] [PubMed] [Google Scholar]
- 398. Sharif T, Martell E, Dai C, et al. Regulation of cancer and cancer-related genes via NAD. Antioxid Redox Signal. 2019;30:906–923. [DOI] [PubMed] [Google Scholar]
- 399. Doblhammer G, Oeppen J. Reproduction and longevity among the British peerage: the effect of frailty and health selection. Proc Biol Sci. 2003;270:1541–1547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 400. Jasienska G. Reproduction and lifespan: trade-offs, overall energy budgets, intergenerational costs, and costs neglected by research. Am J Hum Biol. 2009;21:524–532. [DOI] [PubMed] [Google Scholar]
- 401. Kaptijn R, Thomese F, Liefbroer AC, Van Poppel F, Van Bodegom D, Westendorp RG. The trade-off between female fertility and longevity during the epidemiological transition in the Netherlands. PLoS ONE. 2015;10:e0144353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 402. Kirkwood TB, Austad SN. Why do we age? Nature. 2000;408:233. [DOI] [PubMed] [Google Scholar]
- 403. Liu J, Rotkirch A, Lummaa V. Maternal risk of breeding failure remained low throughout the demographic transitions in fertility and age at first reproduction in Finland. PLoS ONE. 2012;7:e34898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 404. Mason JS, Wileman T, Chapman T. Lifespan extension without fertility reduction following dietary addition of the autophagy activator Torin1 in Drosophila melanogaster. PLoS ONE. 2018;13:e0190105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 405. Meij J, van Bodegom D, Ziem JB, et al. Quality-quantity trade-off of human offspring under adverse environmental conditions. J Evol Biol. 2009;22:1014–1023. [DOI] [PubMed] [Google Scholar]
- 406. Nenko I, Hayward AD, Simons MJP, Lummaa V. Early-life environment and differences in costs of reproduction in a preindustrial human population. PLoS ONE. 2018;13:e0207236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 407. Pettay JE, Helle S, Jokela J, Lummaa V. Natural selection on female life-history traits in relation to socio-economic class in pre-industrial human populations. PLoS ONE. 2007;2:e606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 408. Westendorp RG. Are we becoming less disposable? Evolution has programmed us for early survival and reproduction but has left us vulnerable to disease in old age. In our present affluent environment, we are better adapting to these improved conditions. EMBO Rep. 2004;5:2–6.14710174 [Google Scholar]
- 409. Bolund E, Hayward A, Pettay JE, Lummaa V. Effects of the demographic transition on the genetic variances and covariances of human life-history traits. Evolution. 2015;69:747–755. [DOI] [PubMed] [Google Scholar]
- 410. Hales CN, Barker DJP. The thrifty phenotype hypothesis: type 2 diabetes. Br Med Bull. 2001;60:5–20. [DOI] [PubMed] [Google Scholar]
- 411. Lindstrom J. Early development and fitness in birds and mammals. Trends Ecol Evol. 1999;14:343–348. [DOI] [PubMed] [Google Scholar]
- 412. Metcalfe NB, Monaghan P. Compensation for a bad start: grow now, pay later. Trends Ecol Evol. 2001;16:254–260. [DOI] [PubMed] [Google Scholar]
- 413. Prentice AM, Rayco-Solon P, Moore SE. Insights from the developing world: thrifty genotypes and thrifty phenotypes. Proc Nutr Soc. 2005;64:153–161. [DOI] [PubMed] [Google Scholar]
- 414. Rickard IJ, Holopainen J, Helama S, Helle S, Russell AF, Lummaa V. Food availability at birth limited reproductive success in historical humans. Ecology. 2010;91:3515–3525. [DOI] [PubMed] [Google Scholar]
- 415. Jablonka E, Lamb MJ. Evolution in Four Dimensions: Genetic, Epigenetic, Behavioral, and Symbolic Variation in the History of Life. Cambridge, MA: The MIT Press; 2005. [Google Scholar]
- 416. West-Eberhard MJ. Developmental Plasticity and Evolution. New York, NY: Oxford University Press; 2003. [Google Scholar]
- 417. Odling-Smee FJ, Laland KN, Feldman MW. Niche Construction: The Neglected Process in Evolution (MPB-37). Princeton, NJ: Princeton University Press; 2013. [Google Scholar]
- 418. West GB, Brown JH, Enquist BJ. The fourth dimension of life: fractal geometry and allometric scaling of organisms. Science. 1999;284:1677–1679. [DOI] [PubMed] [Google Scholar]
- 419. West GB, Woodruff WH, Brown JH. Allometric scaling of metabolic rate from molecules and mitochondria to cells and mammals. Proc Natl Acad Sci U S A. 2002;99:2473–2478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 420. Marean CW. Pinnacle Point Cave 13B (Western Cape Province, South Africa) in context: the Cape Floral kingdom, shellfish, and modern human origins. J Hum Evol. 2010;59:425–443. [DOI] [PubMed] [Google Scholar]
- 421. Smith EI, Jacobs Z, Johnsen R, et al. Humans thrived in South Africa through the Toba eruption about 74,000 years ago. Nature. 2018;555:511. [DOI] [PubMed] [Google Scholar]
- 422. Tulchinsky TH. Micronutrient deficiency conditions: global health issues. Public Health Reviews. 2010;32:243–255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 423. Valente FLS. Towards the full realization of the human right to adequate food and nutrition. Development. 2014;57:155–170. [Google Scholar]
- 424. Ehrlich PR, Ehrlich AH. Population, Resources, Environment: Issues in Human Ecology. New York, NY: Freeman; 1972. [Google Scholar]
- 425. Rees M. On the Future: Prospects for Humanity. Princeton, NJ: Princeton University Press; 2018. [Google Scholar]
- 426. Mann ME, Toles T. The Madhouse Effect: How Climate Change Denial Is Threatening Our Planet, Destroying Our Politics, and Driving Us Crazy. New York, NY: Columbia University Press; 2016. [Google Scholar]
- 427. Doyle RM. Darwin’s Pharmacy: Sex, Plants, and the Evolution of the Noosphere. Washington, DC: University of Washington Press; 2011. [Google Scholar]
- 428. Wilson EO. Consilience: The Unity of Knowledge. New York, NY: Vintage Books; 1999. [Google Scholar]
- 429. Jones M. Feast: Why Humans Share Food. Oxford, UK: Oxford University Press; 2008. [Google Scholar]
- 430. Connelly MJ. Fatal Misconception: The Struggle to Control World Population. Cambridge, MA: Belknap Press of Harvard University Press; 2008. [Google Scholar]
- 431. Kupchan CA. No One’s World: The West, the Rising Rest, and the Coming Global Turn. Oxford, UK: Oxford University Press; 2012. [Google Scholar]
- 432. Maçães B. The Dawn of Eurasia: On the Trail of the New World Order. London, England: Penguin Books; 2018. [Google Scholar]
- 433. Levenstein H. Paradox of Plenty: A Social History of Eating in Modern America. Berkeley, CA: University of California Press; 2003. [Google Scholar]
- 434. Shapiro P, Harari YN. Clean Meat: How Growing Meat Without Animals Will Revolutionize Dinner and the World. New York, NY: Gallery Books; 2018. [Google Scholar]
- 435. Starmans C, Sheskin M, Bloom P. Why people prefer unequal societies. Nat Human Behav. 2017;1:0082. [Google Scholar]
- 436. Willett W, Rockstrom J, Loken B, et al. Food in the Anthropocene: the EAT-Lancet Commission on healthy diets from sustainable food systems. Lancet. 2019;393:447–492. [DOI] [PubMed] [Google Scholar]
- 437. Fox-Genovese E. The Origins of Physiocracy: Economic Revolution and Social Order in Eighteenth-Century France. Ithaca, NY: Cornell University Press; 1976. [Google Scholar]
- 438. Desmarais AA, Wiebe N, Wittman H. Food Sovereignty: Reconnecting Food, Nature and Community. Oxford, UK: Pambazuka Press; 2011. [Google Scholar]
- 439. Temkin O. Galenism: Rise and Decline of a Medical Philosophy. Ithaca, NY: Cornell University Press; 1973. [Google Scholar]
- 440. Cannon WB. The Wisdom of the Body. New York, NY: Norton; 1963. [Google Scholar]
- 441. García H, Miralles F. Ikigai: The Japanese Secret to a Long and Happy Life: New York, NY: Random House; 2017. [Google Scholar]
- 442. Holt-Gimenez E. Can We Feed the World Without Destroying It? Hoboken, NJ: Wiley; 2019. [Google Scholar]
- 443. Provenza F. Nourishment: What Animals Can Teach Us About Rediscovering Our Nutritional Wisdom. White River Junction, VT: Chelsea Green Publishing; 2018. [Google Scholar]
- 444. Romagnolo DF, Selmin OI. Mediterranean diet and prevention of chronic diseases. Nutr Today. 2017;52:208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 445. Smith WD. The Hippocratic Tradition. Ithaca, NY: Cornell University Press; 1979. [Google Scholar]
- 446. Hirshberg B. Traditional Nutrition: From Weston A. Price to the Blue Zones; Healthy Diets From Around the Globe. Crafers, SA, Australia: Eudaimonia Press; 2015. [Google Scholar]
- 447. Tudge C. Six Steps Back to the Land: Why We Need Small Mixed Farms and Millions More Farmers. Totnes, UK: Green Books; 2016. [Google Scholar]
- 448. Buchan W, Forsyth JS. Dr. Buchan’s Domestic Medicine: Or, A Treatise on the Prevention and Cure of Diseases by Regimen and Simple Medicines . . . To which Is Annexed a Complete Family Dispensatory, for the Use of Private Practitioners. Philadelphia, PA: Claxton, Remsen & Haffelfinger; 1871. [Google Scholar]
- 449. Pimbert M. Food Sovereignty, Agroecology and Biocultural Diversity: Constructing and Contesting Knowledge. Abingdon, UK: Taylor & Francis; 2017. [Google Scholar]
- 450. Carpenter R. Discontinuity in Greek Civilization. New York, NY: Norton; 1968. [Google Scholar]
- 451. Weiss H. Quantifying collapse: the late third millennium Khabur Plains. In: Weiss H, ed. Seven Generations Since the Fall of Akkad. Wiesbaden, Germany: Harrassowitz; 2012:1–24. [Google Scholar]
- 452. Mason P, Lang T. Sustainable Diets: How Ecological Nutrition Can Transform Consumption and the Food System. Abingdon, UK: Taylor & Francis; 2017. [Google Scholar]
- 453. Springmann M, Mason-D’Croz D, Robinson S, et al. Health-motivated taxes on red and processed meat: a modelling study on optimal tax levels and associated health impacts. PLoS ONE. 2018;13:e0204139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 454. Hanlon J, Barrientos A, Hulme D. Just Give Money to the Poor: The Development Revolution From the Global South. Sterling, VA: Kumarian Press; 2010. [Google Scholar]
- 455. Sheahen A. Basic Income Guarantee: Your Right to Economic Security. New York, NY: Palgrave Macmillan; 2012. [Google Scholar]
- 456. Steensland B. The Failed Welfare Revolution: America’s Struggle Over Guaranteed Income Policy. Princeton, NJ: Princeton University Press; 2011. [Google Scholar]









