"Life Without Bread"

SeekinTruth said:
Yeah, good luck Psyche. Hope you're able to take care of yourself and not get to stressed out.

We will miss her and her counsels. Was she living in the chateau? Sorry, maybe this question is too personal. I wish her good luck and happiness.
 
Laura said:
The problem with nuts mainly seems to be the proteins which are similar to some body proteins and in many people trigger an auto-immune or allergic reaction. Almonds are the worst offenders. But everybody is different so after you are sure your gut is healed (can take up to two years on the diet), then you can experiment with nuts.

There's one MORE thing I neglected to mention. Nuts tend to be high in phytic acid, which in turn can chelate important minerals and create nutritional deficiencies. Soaking, dehydrating, and roasting can greatly reduce phytic acid (as can limiting your intake of nuts!).

That "funny feeling" some of us get after eating nuts (for me it is only if I OD on them) could be any combination of response to proteins, mineral imbalance, and excess Ω-6s. Apparently quite a few people trying Paleo have overdone the nuts. A regular small handful of nuts probably isn't going to hurt, but it may not provide the expected nutritional benefit (from minerals) that you thought you were getting.
 
Megan said:
That "funny feeling" some of us get after eating nuts (for me it is only if I OD on them) could be any combination of response to proteins, mineral imbalance, and excess Ω-6s. Apparently quite a few people trying Paleo have overdone the nuts. A regular small handful of nuts probably isn't going to hurt, but it may not provide the expected nutritional benefit (from minerals) that you thought you were getting.

Oh, I'm not eating them for any benefits except pure pleasure!

But I do think I'll reduce them for a bit.

The info about Vitamin B3 and its relation to nicotine is rather fascinating. I've got some downstairs so I think I'll try taking it for awhile.
 
a few months ago I had a pistachios trip (on the paleo diet).
I ate up to 400 g of them per day, for weeks, usually at evening, simply because I couldn't stop to eat them !
they are so darn delicious and on top they don't have much carbohydrates ! :mad:

to be honest I knew that I don't really tolerate them but couldn't withstand the temptation.
In short I was nut addicted ! :lol:

It took me quite some time,will and strength to stop eating them, but I did.
I'm a nutaholic ! so I stand away from them because I know that when I eat some again I'll probably can't control myself again.
 
dugdeep said:
Hi Foxx. I'm sorry, but I don't have a quick reference off the top of my head. I've been studying this stuff for 6+ years so a lot of it has just integrated into my general knowledge. The Wikipedia entry on inulin is pretty good (just skimmed it) and if you haven't read The Fiber Menace, that has some good info. I'm also pretty sure both Stephan Guyenet (_www.wholehealthsource.com) and Chris Masterjohn (_http://blog.cholesterol-and-health.com/) have written about bacterial fermentation and its beneficial byproducts (butyric acid, vitamin K, etc.) in the past. Sorry I can't be more help. If I had more time I'd do some research and track down some good references, but I'm swamped preparing for a class I'm teaching tomorrow all day today.

Thanks dugdeep, this is also helpful! I'll look around online on the topic and see what I can find :)
 
Laura said:
The info about Vitamin B3 and its relation to nicotine is rather fascinating. I've got some downstairs so I think I'll try taking it for awhile.

If you're going to take niacin (as opposed to niacinamide) watch out for the flush. The first time you take it, even 100mg can give a doosy of a flush.

Added: :shock: :O :scared:
 
Laura said:
A little addenda:

Greatly restrict or eliminate sugar and starch (preferably eliminating gluten completely); keep your protein intake adequate (roughly the RDA: 44-56 g per day or 0.8 g of protein/ kg of ideal body weight), amounting to a total of approximately 6 to 7 ounces of organic eggs, grass-fed, or wild-caught meat or seafood per day; eat as many fibrous, "aboveground," nonstarchy vegetables and greens as you like; and eat as much fat (from fattier cuts of meat or fish, nuts, seeds, avocados, coconut, butter or ghee, olives, olive oil, and other sources) as you need to satisfy your appetite.

That is actually a very small amount of protein. It amounts to about 1.5 to 2 ounces per DAY! That really is famine! You would have to eat a lot of veggies to feel satisfied. Or fat. And I'm not sure that I could eat that much fat without feeling sick.

It really is. As for me I would need to eat many other things to feel satisfied.

Eventually it has been already discussed, but somehow this question comes up again. So far ketogenic/VLC diets are considered for loosing weight, but what to do if you don't like to loose any weight? Eventually could skinny people eat more carbs, since carbs and fat in combination could raise the body weight a bit, accodring to Gedgaudas if I remember it correctly and couldn't do any harm? Since trying the leptin reset after Kruse I lost 2kgs from 64 to 62 again.

So I eventually try out again more complex carbs in combination with fat and go for about 40grs just to test things.


Pashalis said:
a few months ago I had a pistachios trip (on the paleo diet).
I ate up to 400 g of them per day, for weeks, usually at evening, simply because I couldn't stop to eat them !
they are so darn delicious and on top they don't have much carbohydrates ! :mad:

These were also many, many carbs.
 
Gawan said:
Pashalis said:
a few months ago I had a pistachios trip (on the paleo diet).
I ate up to 400 g of them per day, for weeks, usually at evening, simply because I couldn't stop to eat them !
they are so darn delicious and on top they don't have much carbohydrates ! :mad:

These were also many, many carbs.

actually 100g of pistachios without shell have about 12g carbohydrates.
the 400g are meant with schell (wich you can't eat and are quite heavy).
so the actual number must have been about 300g wich would make about 40g carbohydrates.
that is one of the reason I even more liked to eat them because they are so low in carbs.
I think they are one of the lowest nuts in terms of carbs.
 
LQB said:
Laura said:
The info about Vitamin B3 and its relation to nicotine is rather fascinating. I've got some downstairs so I think I'll try taking it for awhile.

If you're going to take niacin (as opposed to niacinamide) watch out for the flush. The first time you take it, even 100mg can give a doosy of a flush.

Added: :shock: :O :scared:

Yep, this happened to me last week and I was literally neon pink for about a half hour - not very comfortable!
 
Laura said:
A little addenda:

Greatly restrict or eliminate sugar and starch (preferably eliminating gluten completely); keep your protein intake adequate (roughly the RDA: 44-56 g per day or 0.8 g of protein/ kg of ideal body weight), amounting to a total of approximately 6 to 7 ounces of organic eggs, grass-fed, or wild-caught meat or seafood per day; eat as many fibrous, "aboveground," nonstarchy vegetables and greens as you like; and eat as much fat (from fattier cuts of meat or fish, nuts, seeds, avocados, coconut, butter or ghee, olives, olive oil, and other sources) as you need to satisfy your appetite.

That is actually a very small amount of protein. It amounts to about 1.5 to 2 ounces per DAY! That really is famine! You would have to eat a lot of veggies to feel satisfied. Or fat. And I'm not sure that I could eat that much fat without feeling sick.

It seems OK to me. I imagine only 2 ounces of meat and/or other protein-containing foods per day would lead to starvation (self-consumption), but 2 ounces of protein seems reasonable. I don't know where the 44g low figure comes from, but 56g/d is the RDA for adult males. But some people eat way more than that.

I briefly reviewed the materials above, and I still really like the way Nora brought together paleo & life extension ideas. I think she is on firm ground with it, although it is an evolving area of study and the understanding may shift over time. I am going to resume experimenting with protein restriction. I have mostly avoided it because it can potentially limit weight loss, but I think I am going to quit worrying about that for now. Over the last year I have tried less protein and more protein and it's hard to pin down but my body seems to do better on less, not more.
 
Found some more information on nicotinamide and nicotine ...
This article was published in QJM (QJM (2005) 98 (3): 215-226). It can be found here

Parkinson's disease: the first common neurological disease due to auto-intoxication?
A.C. Williams, L.S. Cartwright 2 and D.B. Ramsden

Parkinson's disease may be a disease of autointoxication. N-methylated pyridines (e.g. MPP+) are well-established dopaminergic toxins, and the xenobiotic enzyme nicotinamide N-methyltransferase (NNMT) can convert pyridines such as 4-phenylpyridine into MPP+, using S-adenosyl methionine (SAM) as the methyl donor. NNMT has recently been shown to be present in the human brain, a necessity for neurotoxicity, because charged compounds cannot cross the blood-brain barrier. Moreover, it is present in increased concentration in parkinsonian brain. This increase may be part genetic predisposition, and part induction, by excessive exposure to its substrates (particularly nicotinamide) or stress. Elevated enzymic activity would increase MPP+-like compounds such as N-methyl nicotinamide at the same time as decreasing intraneuronal nicotinamide, a neuroprotectant at several levels, creating multiple hits, because Complex 1 would be poisoned and be starved of its major substrate NADH. Developing xenobiotic enzyme inhibitors of NNMT for individuals, or dietary modification for the whole population, could be an important change in thinking on primary and secondary prevention.

Introduction

In 1887, a medical student in Strasbourg (W. His) gave a dog a quantity of the exogenous compound pyridine, and found it was predominantly excreted as its N-methyl derivative.1 Subsequent studies of xenobiotic metabolism and pharmacogenetics have yielded many examples of detoxification of foreign chemicals. Among such reactions, methyl conjugation is a well established pathway in the metabolism of endogenous agents, including hormones, neurotransmitters, vitamins and drugs. The xenobiotic metabolizing system must be an important defence mechanism, particularly as most xenobiotics are too small to be recognized by the immune system.

However, like the immune system, while normally protective, this system can also cause rather than prevent toxicity, by converting protoxins to toxins. In 1952, Peters coined the term ‘lethal synthesis from enzymic error', in his Croonian lecture to the Royal Society. He was referring to the neurotoxicity of fluoroacetic acid, a protoxin that crosses the blood brain barrier easily, and after conversion to fluorocitric acid impedes mitochondrial energy production due to blockade of the Krebs cycle,2 but many examples of this can be found from the conversion of prodrugs to drugs to toxic metabolites.

Here we suggest that the basis for idiopathic Parkinson's disease (PD) is a lethal synthesis from, with a failure to preserve, vitamin B3. There is evidence that genetic and environmental factors are important in this common disease,3–6 but until now, nobody has convincingly linked a specific gene with specified diet-derived substrates, let alone a vitamin. We propose a class of ecogenetic diseases of auto intoxication, and predict that chronic poisoning by transformed chemicals may be a common mechanism for conditions linked with ageing. Furthermore, we suggest that this hypothesis gives immediate ideas on primary prevention by dietary manipulation, reducing availability of the methyl donor, or the development of xenobiotic enzyme inhibitors (XEIs), a new class of therapeutic agents.

Nicotinamide N-methyltransferase (NNMT)

The enzyme that His was studying in 1887 was NNMT. This enzyme N-methylates pyridines, in particular nicotinamide to N-methyl nicotinamide, its major metabolite. (Figures 1 and 2). The remainder goes on to form NAD(H) and NADP(H), and therefore is vital to a large number of metabolic reactions. NADP(H) is generally associated with different roles from those of NAD(H),7 and is believed to serve primarily in oxidative defence or reductive metabolism.8 In the brain, a major oxidative defence mechanism consists of peroxide removal using GSH peroxidase. It has been demonstrated that GSH detoxification pathways may ultimately depend on the availability of NADPH reducing equivalents to replenish GSH as a cofactor for GSSG reductase. NAD(H), on the other hand, is believed to serve primarily in the energetic production of ATP. NAD+ depletion is considered a critical factor in precipitating cell death during oxidative stress due to compromised energetics. Of particular interest to PD, NADH is integral to: (i) the normal function of complex 1 of the mitochondrial chain (known to be defective in MPTP Parkinsonism9 and the idiopathic condition10,,11); (ii) the formation of tetrahydrobiopterin (a co-factor for tyrosine hydroxylase and therefore important in the production of dopamine and known to be deficient in PD12) and reduced glutathione (also known to be deficient in early stages of PD13,,14). Nicotinamide provides cytoprotection through pathways that involve poly (ADP-ribose) polymerase (PARP), Akt, mitochondrial membrane potential and cysteine protease activity. Nicotinamide also maintains cellular integrity and prevents cellular removal by maintaining DNA integrity and membrane phosphatidylserine (PS) asymmetry. It affects DNA degradation through a series of cellular pathways that involve PARP, Akt, forkhead transcription factor, mitochondrial membrane polarization, cytochrome c, and inhibition of caspase-1, caspase-3 and caspase-8. In addition, cellular membrane asymmetry, which prevents PS externalization and protects against microglial and cytokine activation with subsequent phagocytic destruction of cells, is maintained principally through activation of caspase-1 and caspase-8.15–26 Many of these pathways have been invoked for MPTP poisoning and PD.27–31 Nicotinamide protects against the toxic effects of L-DOPA in cell culture, as illustrated in Figure 3, and MPTP toxicity.32–3939 Experimental nicotinamide deficiency, produced by the antimetabolite 6-aminonicotinamide, causes dopamine deficiency, loss of cells in the pars compacta, and parkinsonism with a good response to dopamine agonists. Toxicity is based on the lethal synthesis of analogues of NADH and NADPH.40

Toxicity of N-methyl compounds

A single case45 followed by a small epidemic of parkinsonism46 in young heroin injectors was found to be caused by MPTP, a N-methylated pyridine. The similarity with idiopathic PD is striking at the clinical, pathological and biochemical levels and has led to excellent animal models. Most of the biochemical and physiological abnormalities found in the last 20 years from the deficit of complex 1 and the involvement of the subthalamic nucleus in PD were first discovered in the MPTP model, and it has been extensively used for drug and surgical developments. It was soon found that compounds of similar structure that did not have the N-methyl moiety were not toxic to any degree.47,,48 MPTP is a protoxin converted by MAO-B to MPP+.49 MPP+ and similarly charged compounds cannot cross the blood brain barrier. If this conversion occurs within the brain, MPP+ may be taken up into dopaminergic cells and concentrated via the dopamine active transport system and further concentrated in mitochondria where it poisons complex 1.50,,51 Inhibition of MAO-B prevents MPTP toxicity, which is also relatively less toxic by the oral route because MAO-B in liver converts it to MPP+, which then cannot cross the blood-brain barrier. Interestingly, inhibition of MAO-B does not affect the natural history of Parkinson's disease. However, an alternative and direct pathway converts pyridines, including nicotinamide and 4-phenyl pyridine, directly to MPP+ and similar compounds This route is catalysed by NNMT (Figure 1).52 If this happened in vivo, MAO-B inhibition would not obviate the generation of toxins. Several N-methylated compounds and/or their N-methylating enzymes (probably NNMT) have been proposed as being involved in Parkinson's disease.53–64 However these findings have not been generally accepted, in part because it was unclear whether the relevant enzyme occurred in the CNS of mammals.65

Nicotinamide metabolism in parkinsonian patients

An initial study in Man66–69 followed the Wilhelm His protocol in dogs, and demonstrated that patients with Parkinson's disease excreted high quantities of N-methyl nicotinamide, compared with controls or patients with other neurodegenerative disease.70 However, at the time, the brain was not thought to contain NNMT, and therefore there were problems with the hypothesis, because increased methylation peripherally would reduce available nicotinamide but the charged N-methyl compounds would not cross the blood-brain barrier.71 However, NNMT is found in the brain, and indeed is within neurones including dopaminergic neurones of the substantia nigra (Figure 4).72 It has a regional distribution, presumably reflecting nicotinamide requirements.73 . Intra-neuronal nicotinamide levels may be a link between degeneration when too low, and carcinogenesis or developmental problems when too high.Low nicotinamide levels are toxic, but so are high levels,74,,75 so tight control may be necessary via PARP depletion,76 such as might be achieved by high dietary intake of nicotinamide and low methylator status from genetic pre-disposition plus an inhibitor of NNMT such as nicotine. N-methylnicotinamide may have acquired a normally beneficial role as it inhibits the export of choline77 (also a charged N-methylated compound) from the brain, and therefore boosts acetylcholine levels. This may help developing cognition, and even delay the effects of cholinergic degeneration although it would not be pharmacologically helpful in patients who already had PD. On the other hand, N-methyl nicotinamide has MPP+-like toxicity,78–80 and choline as a methyl donor would facilitate further methylation. NNMT levels are high in the spinal cord and in some parts of the cortex, suggesting that such regions prefer lower nicotinamide and higher N-methyl nicotinamide, and vice versa for regions such as substantia nigra with low levels of the enzyme. In Parkinson's disease brain, levels of this enzyme are high (Figure 5).73,81,,82 They were not raised in the substantia nigra, but as a neuronal protein, the destruction from the disease process makes this hardly surprising; levels might have been high before the destruction took place. In any case, N-methyl compounds could diffuse from other parts of the brain and get taken up by dopaminergic cells, as happens with MPTP poisoning.

S-adenosyl methionine (SAM)

SAM is the methyl donor for this and most other important methylation reactions, including that of DNA and RNA. One study showed decreased CSF levels in untreated Parkinson's disease,83 and this was lowered further by treatment.84 The former is presumably due to increased consumption perhaps from N-methylation and the latter from methylation of dopamine by catechol-O-methyl transferase (COMT). Forcing dopamine catabolism down the methylation path by blocking decarboxylation and MAO-B, particularly in patients with the high methylator COMT polymorphism, would consume SAM, reducing the amount available for other methylation reactions. This may explain some of the beneficial effects of Levodopa, which have always been complex and on several timescales not necessarily all explained by dopamine replacement. Nor are side-effects such as dyskinesia fully explained, and it is possible that a de-methylation/re-methylation injury is to blame, which is the reason given when infants who are vitamin-B12-deficient and are treated with this indirect methyl donor develop dyskinesia. Blocking COMT using inhibitors now widely available may therefore have unforeseen consequences, by boosting SAM, which can cross the blood-brain barrier. This is particularly true for Tolcapone, which unlike Entacapone inhibits COMT centrally, and therefore would increase SAM directly in the brain.85,,86 This central effect might explain its better immediate symptomatic response, but may cause longer-term toxicity. Arguing strongly for the methylation hypothesis, increasing SAM in striatum in animal models causes MPP+-like toxicity.87 Trials comparing disease progression with SAM-reducing, SAM-neutral and SAM-increasing strategies would be of interest. Agents that reduce its synthesis from methionine or increase its consumption or catabolism toward cysteine might need to be developed.

Known environmental risk factors

Would this fit with the other known risk factors for Parkinson's disease? Age is the greatest risk factor, and slow poisoning is therefore an attractive mechanism. There may be some brakes on the age increase, or it would be exponential, e.g. NNMT activity may decline with age.70 The strongest environmental factors are nicotine and caffeine, which are both protective.88–95 No convincing mechanism for this has been described, but both are N-methylated compounds, which seems an extraordinary coincidence (Figure 6). As Paracelsus first predicted, causes and cures may be closely related structurally. Nicotine is already N-methylated but has another potential site, and is a substrate (and therefore may act as a inhibitor) of NNMT. Caffeine is already N-methylated at three sites, but given that demethylation is its normal catabolic event, it may get re-methylated by NNMT and also then act as a substrate, or it may be an inhibitor in its own right.

***
Figure 6.

Structural formulae of nicotine and caffeine.

Studies on dietary nicotinamide intake in Parkinson's are confusing.96,,97 It has been proposed that high nicotinamide in diet is protective, but this has been disputed and is confounded by the fact that coffee has a high nicotinamide component. Parkinson's disease is rare in alcoholics who are at risk of nicotinamide deficiency. Pellagra is commoner in women, probably because oestrogen suppresses tryptophan metabolism, which is an important source of NADH in liver. Also, their dietary intake of nicotinamide is lower than in men.98 Parkinson's disease, on the other hand, is commoner in men.97 Parkinson's disease is also rare in regions such as Africa where pellagra still occurs, even though Black Americans get PD at the same rate as White Americans within a few generations.99 The epidemiological data on nicotinamide dietary dose is therefore conflicting, and could fit with a ratio of pyridine intake that is skewed toward protoxic rather than protectant compounds. At an individual level, any safe intake may well differ between high and low methylators, and there may be a window with toxicity at the extremes. Thus the epidemiology may not be explicable until we can either phenotype or genotype patients at the same time as making a complex assessment of dietary intake of most of the relevant compounds. Clearly, high methylators may need less protoxin to cause damage. They also may prove to have different smoking or coffee-drinking habits, as the increased methylation may be either unpleasant and lead to avoidance, or need a higher dose to get the pleasant and addictive effects. Indeed, the interaction between nicotine and NNMT should be investigated separately to understand its role in addiction.

***

Genetic risk factors

There has been immense recent interest in the genetics of the rare Mendelian versions of Parkinson's disease. These discoveries have highlighted the role of proteins such as α-synuclein, Parkin, UCHL1, DJ1 and PINK 1.100–107 The mechanism in these families is not known, but the lead hypothesis is that they interfere with the ubiquitin-proteasome pathway, or with other components of cellular protective mechanisms, including Complex 1 and free radical defence mechanism pathways that had already been implicated in Parkinson's disease. Ubiquitination is important in the removal of already damaged proteins, and is ATP-dependent, so could be impaired secondary to energy failure or as a primary event in these families. What has not been explained is what threatens the cell in the first place. Even in these families, a MPP+ like compound could be the trigger, although needing a lower dose of toxin than in idiopathic Parkinson's disease, perhaps explaining their earlier onset of clinical disease. Certainly α-synuclein knockout mice are resistant to MPP+,108,,109 and inhibition of Complex 1 makes cells sensitive to MPP+,110,,111 as are models of PARKIN mutations;112 one would predict the same with DJ1 and PINK 1 equivalents.113 Toxic exposure can cause upregulation of α-synuclein.114 Cause and effect can easily get confused here, and potential feedback loops from a network of biochemical changes may well be present, emphasizing the need to move proximally for the greatest chance of successful therapeutic intervention. Parkinson's disease may have a myriad of rare causes and common risk and protective factors, but they may all be affecting a single pathway. The methylation hypothesis is inclusive, in that N-methylation may be at the head of a toxico-biochemical pathway that later includes these proteins and mechanisms of cell death.

Conclusion

Hypersusceptibility to chemicals may be a cause of degenerative disease such as Parkinson's disease, and may be happening at the xenobiotic enzyme level.115–117 Both high exposure to protoxic pyridines relative to protectant compounds, and genetic predisposition in the form of high N-methylating capacity, are necessary to cause disease. Consequently, most patients will not have a family history. We suggest that a toxic brew of a variety of N-methyl compounds is a likely scenario, defining at least at first the selective death of dopaminergic neurones. Details of the mix would vary between different individuals, depending on the degree and nature of the exposure. This may explain phenotypic variation, including toxicity outside traditional MPP+ territory, as some of the toxins may not be so selective.118 A logical strategy would be either to reduce environmental exposure to protoxin or to increase exposure to protectants, either in the whole population or in individuals at risk. However, that may not be simple, as a glance at Figure 2 demonstrates. The case of nicotinamide-NNMT is an example of a two-edged sword, because cells may benefit from more of the protectant, nicotinamide, but be damaged by increased levels of toxic N-methylnicotinamide.

High exposure to its substrates, particularly nicotinamide to which the Western population has become increasingly exposed may induce the enzyme, as will stress. This could be the first of several increasingly toxic vicious circles that result from the intra-neuronal deficiency of the vitamin or its downstream direct and indirect products, such as NADH and ATP combined with toxicity from several N-methylated compounds. If true, we are dealing with upstream events and switching them off would not just affect one part of a late cascade of biochemical events or be purely symptomatic, or be only aimed at the dopaminergic damage. Increased irreversible catabolism of nicotinamide may lead to the cellular deficiency of the vitamin and of products, which would also affect non-dopaminergic cells as seen in pellagra. Here dementia and depression are the predominant features, but autonomic, pontine, anosmic and sleep disturbances are seen, and remarkably, also parkinsonism in 10–20% of cases.119 One nicotinamide anti-metabolite, 3-acetyl pyridine, causes atypical parkinsonism, and has been suggested as a model of olivopontocerebellar atrophy.120 In view of the fact that dementia and some of these other atypical features of Parkinson's disease occur in these circumstances, one wonders whether the clinical pathological spectrum of parkinsonism, and not just classic Parkinson's disease, is a hybrid of pyridinium ion poisoning and pellagra.

Altering nicotinamide in the diet as a potential protective manoeuvre may work, but the dose may need to be individualized, and N-methyl compounds would still be produced, including N-methylated nicotinamide. Western societies who suffer more from PD may now have too much nicotinamide in their diet overall, and this may need to be addressed at the population level. Strategies to reduce methylation by removing the co-factor SAM using existing drugs or the development of SAM-depleting agents would be a novel avenue to explore, but would affect other necessary methylation reactions. Inhibition of the enzyme NNMT may be more incisive, as it would boost intracellular nicotinamide and reduce or stop the production of any N-methyl compounds. If the enzyme has been induced over a lifetime by excessive exposure to its substrates or other factors, a reduction in such stimuli may not reverse its overactivity quickly or completely. Competitive inhibitors could be developed, because they are often structurally related to the known substrates. Two lead compounds that may be protective are nicotine and caffeine. Neither may be practical, because they have their own toxicity. Nevertheless, they, along with the structures of nicotinamide or 4-phenyl pyridine may provide clues to the development of a safe pyridine, whether natural or synthetic, leading to attempts at primary and secondary prevention based on a strong hypothesis supported by both biochemical and epidemiological evidence. Symptomatic therapies have obvious limitations, but so does targeting downstream biochemical events, as has been learnt, often painfully, from cancer and inflammatory diseases. XEIs alone or dietary or SAM manipulation may prove to be a revolutionary new class of preventive therapeutic agents, not just for Parkinson's disease. One example of such a possibility is overactive xenobiotic sulphur methylation pathway, which has been implicated in motor neurone disease.121,,122 Given that metal123 or pesticide exposure124,,125 may be a risk factor for MND, and S-methylation of some compounds increases their toxicity, a very similar ecogenetic scenario can be postulated. MPTP was discovered by a freak event in drug users; MND may never have had the equivalent lucky break. One can make similar arguments for some other diseases, including cancer and processes linked with increasing age. Indeed, more varied diets may inadvertently reduce xenobiotic load, perhaps explaining the recent increase in longevity not wholly due to improvements in healthcare. Dietary restriction has long been known to increase life-span and reduce age related disease in experimental animals, and is attributed to lower caloric intake,126 but might also be related to lower daily xenobiotic exposure.
 
anart said:
LQB said:
Laura said:
The info about Vitamin B3 and its relation to nicotine is rather fascinating. I've got some downstairs so I think I'll try taking it for awhile.

If you're going to take niacin (as opposed to niacinamide) watch out for the flush. The first time you take it, even 100mg can give a doosy of a flush.

Added: :shock: :O :scared:

Yep, this happened to me last week and I was literally neon pink for about a half hour - not very comfortable!

The first time I did about 200 mg and the flush went all the way down to my knees - not to mention the neon pink and itching. Fred Klenner MD suggests that you resist itching by pressing down on the skin or using an ice cube.

While running over the doctoryourself website, I ran into one of Fred Klenner's papers on using high-dose Vit B complex to treat/cure MS and other neuropathies. In it there is this:

We categorically make this statement: Any victim of Multiple Sclerosis who will dramatically flush with the use
of nicotinic acid, and who has not yet progressed to the stage of myelin degeneration, as witnessed by sustained ankle
clonus elicited in the orthodox manner, can be cured with the adequate employment of Thiamin Hydrochloride and
other factors of the Vitamin B Complex in conjunction with essential proteins, lipids, carbohydrates and injectable
crude liver. If sustained ankle clonus is not bilateral, then it is not a deterrent. We have had patients who did
demonstrate bilateral sustained ankle clonus, and who were in wheelchairs, and who returned to normal activities after
5 to 8 years of treatment. These patients, fortunately, had not received ACTH. One patient was given a single course
of Medrol 4 mg. QID. This had little effect on her pathology, and apparently no blocking action, on our treatment.
The general use of ACTH in Multiple Sclerosis will extend the recovery period by a time directly proportional to the
amount of the drug employed. It is hoped that this paper will bring an end to this senseless practice of medicine, since
ACTH never works the third time.

The paper is a great read with case histories, and so on.

Response of Peripheral and Central Nerve Pathology to Mega-Doses of the Vitamin B-Complex and Other
Metabolites
by Frederich R. Klenner, BS, MS, MD
Journal of Applied Nutrition, 1973
The protocol of how to effectively treat Multiple Sclerosis. (In two parts, as originally published in 1973.)
_http://www.townsendletter.com/Klenner/KlennerProtocol_forMS.pdf

I posted this primarily for Gimpy since it is such a good read on Klenner's work with MS/neuropathy. Sorry for straying :offtopic:
 
Found another paper on nicotinamide, which also looks among other things at the link to sirtuins.


The Vitamin Nicotinamide: Translating Nutrition into Clinical Care
Kenneth Maiese,1,2,3,4,5,* Zhao Zhong Chong,1 Jinling Hou,1 and Yan Chen Shang1

Abstract

Nicotinamide, the amide form of vitamin B3 (niacin), is changed to its mononucleotide compound with the enzyme nicotinic acide/nicotinamide adenylyl-transferase, and participates in the cellular energy metabolism that directly impacts normal physiology. However, nicotinamide also influences oxidative stress and modulates multiple pathways tied to both cellular survival and death. During disorders that include immune system dysfunction, diabetes, and aging-related diseases, nicotinamide is a robust cytoprotectant that blocks cellular inflammatory cell activation, early apoptotic phosphatidylserine exposure, and late nuclear DNA degradation. Nicotinamide relies upon unique cellular pathways that involve forkhead transcription factors, sirtuins, protein kinase B (Akt), Bad, caspases, and poly (ADP-ribose) polymerase that may offer a fine line with determining cellular longevity, cell survival, and unwanted cancer progression. If one is cognizant of the these considerations, it becomes evident that nicotinamide holds great potential for multiple disease entities, but the development of new therapeutic strategies rests heavily upon the elucidation of the novel cellular pathways that nicotinamide closely governs.

1. Introduction

Nicotinamide (Figure 1) is the amide form of vitamin B3 (niacin) and is obtained through synthesis in the body or as a dietary source and supplement [1]. Nicotinic acid is the other form of the water-soluble vitamin B3 (Figure 1). Although also present from animal sources, the principal form of niacin in dietary plant sources is nicotinic acid that is rapidly absorbed through the gastrointestinal epithelium [2]. Nicotinamide is subsequently generated through the conversion of nicotinic acid in the liver or through the hydrolysis of NAD+. Once nicotinamide is obtained in the body, it functions as the precursor for the coenzyme β-nicotinamide adenine dinucleotide (NAD+) [3,4] and also is essential for the synthesis of nicotinamide adenine dinucleotide phosphate (NADP+) [5]. Initially, nicotinamide is changed to its mononucleotide form (NMN) with the enzyme nicotinic acid/nicotinamide adenylyltransferase yielding the dinucleotides NAAD+ and NAD+. NAAD+ also yields NAD+ through NAD+ synthase [6] or NAD+ can be synthesized through nicotinamide riboside kinase that phosphorylates nicotinamide riboside to NMN [7,8]. These cellular pathways are essential for energy metabolism and may directly impact normal physiology, as well as disease progression [9–12].

In deficiency states, lack of nicotinamide can lead to fatigue, loss of appetite, pigmented rashes of the skin, and oral ulcerations. More severe states of deficiency lead to pellagra that is characterized by cutaneous rashes, oral ulcerations, gastrointestinal difficulties, and cognitive loss. Pellagra can occur during low nicotinamide conditions or due to the inability to absorb nicotinamide. For example, inability to absorb tryptophan that causes Hartnup’s disease, isoniazid treatment, or carcinoid syndrome also can be associated with pellagra. Excessive alcohol consumption that is associated with poor dietary intake also can lead to severe nicotinamide loss and insufficient gastrointestinal absorption.

2. Nicotinamide, Oxidative Stress, and Cellular Survival

Ultimate cellular survival can be determined by a number of factors, but the process of apoptosis can represent one of the critical pathways for a number of disease entities. Apoptosis can contribute to disorders such as diabetes [13–16], tissue ischemia [17–20], bone fatigue [21], Alzheimer's disease [22–32], neurodegenerative disorders [33–36], plasticity associated with ischemic preconditioning [37], aging-related diseases [38–40], and toxic conditions during development [41,42]. The pathology with these disorders can be linked to mitochondrial dysfunction [43–46], especially during metabolic disorders [47] and Alzheimer’s disease [48], that ultimately can lead to cell death in a variety of cells such as neurons, endothelial cells (ECs), cardiomyocytes, and smooth muscle cells [32,49–53].

At the cellular level, apoptosis consists of both the early exposure of membrane phosphatidylserine (PS) residues and the subsequent destruction of genomic DNA [54,55]. Externalization of membrane PS residues can occur first during cellular apoptosis [56,57]. Apoptotic membrane PS exposure occurs in neurons, vascular cells, and inflammatory microglia during reduced oxygen exposure [58–62], β-amyloid (Aβ) exposure [26,63], nitric oxide exposure [64–68], and during the administration of agents that induce the production of reactive oxygen species (ROS), such as 6-hydroxydopamine [69]. Membrane PS externalization also occurs on platelets and has been associated with clot formation in the vascular system [70]. Furthermore, membrane PS exposure can become a signal for the phagocytosis of cells [59,71,72]. The loss of membrane phospholipid asymmetry leads to the exposure of membrane PS residues on the cell surface and assists microglia to target cells for phagocytosis [4,52,73–75]. In conjunction with PS externalization, increased expression of the phosphatidylserine receptor (PSR) on microglia occurs to facilitate activation of these cells [76,77] since blockade of PSR function prevents the activation of microglia [74,78].

Usually following membrane PS exposure [79], the cleavage of genomic DNA into fragments occurs [35,61,80] as a later event during apoptotic injury [52,80–82]. There are a number of enzymes that degrade DNA. These include the acidic, cation independent endonuclease (DNase II), cyclophilins, and the 97 kDa magnesium - dependent endonuclease [83,84]. In addition, three separate endonuclease activities have been found in neurons that include a constitutive acidic cation-independent endonuclease, a constitutive calcium/magnesium-dependent endonuclease, and an inducible magnesium dependent endonuclease [85,86].

One of the inciting factors that can lead to apoptotic cell injury is oxidative stress. Oxidative stress plays a critical role in the pathology of numerous processes and disorders throughout the body that can include metabolic disorders [47,87–95], ocular disease [96], environmental influences such as with tobacco exposure [97,98], cognitive impairment [99–102], ischemic injury [103,–105], epilepsy [106,107], nutrition [108], cardiopulmonary and hepatic disease [109–111], degenerative disorders and psychiatric disorders [112–114], infertility [115–117], excitotoxicity [118–120], and drug toxicity [121–123]. Early work with oxidative stress examined the rate of oxygen consumption in organisms and proposed that increased exposure to oxygen through a high metabolic rate could lead to a shortened life span [124]. Other work demonstrated that increased metabolic rates could be detrimental to animals in an elevated oxygen environment [125].

Recent studies have expanded these observations to show that ROS and mitochondrial DNA mutations have become associated with multiple processes to include cellular injury, aging mechanisms, and accumulated toxicity for an organism [126]. It is the release of ROS that leads to oxidative stress. ROS include superoxide free radicals, hydrogen peroxide, singlet oxygen, nitric oxide (NO), and peroxynitrite [34,83,127] that if expressed at increased concentrations can lead to cellular injury and demise through oxidative stress [59,128,129]. Most ROS occur at low levels and are scavenged by endogenous antioxidant systems that include superoxide dismutase (SOD), glutathione peroxidase, catalase, and small molecule substances such as vitamins C, D, E, and K [76,107,114,130,131]. Yet, one vitamin in particular, namely nicotinamide may be considered to stand-alone among antioxidants since nicotinamide influences multiple pathways tied to both cellular survival and cellular death.

In several scenarios, nicotinamide is a robust cytoprotectant that addresses both early membrane PS externalization and later genomic DNA degradation [3,4,34,76,132] during oxidative stress in a way that is different from other vitamin entities. Administration of nicotinamide during anoxia, oxygen-glucose deprivation, and free radical exposure can prevent exposure of membrane PS residues to block inflammatory cell activation [3,133–135] and inhibit later genomic DNA destruction [134–136] (Figure 2). In addition, nicotinamide prevents membrane PS exposure in vascular cells [4,134] that can reduce risk for cardiovascular disorders, since membrane PS residue externalization in vascular cells can lead to increased propensity for hypercoagulation [137] and cellular inflammation [138,139].

In several instances, nicotinamide also may reverse a previously sustained insult [4,132–135,140]. Post-treatment strategies with nicotinamide that can follow apoptotic injury in “real-time” show that cellular injury can be reversed. Nicotinamide can reverse an initial progression of membrane PS inversion and prevent PS exposure over a twenty-four hour period [4,132,135,141]. These results suggest that apoptosis prior to reaching genomic DNA degradation is dynamic and reversible in nature [4,132,135,141]. Yet, in not all cases may nicotinamide be effective to prevent subsequent DNA degradation [76]. During periods of acidosis-induced cellular toxicity [142], mitochondrial failure can ensue [143]. In addition, ROS can result in the disturbance of intracellular pH that leads to endonuclease activity and DNA injury during apoptosis [85,86,144]. In events that involve decreased pH, nicotinamide cannot prevent cellular injury during intracellular acidification [135]. For example, exposure to ROS leads to a biphasic response for pHi. Treatment with nicotinamide (12.5 mM) alone does not alter neuronal pHi. In addition, pretreatment with nicotinamide (12.5 mM), a neuroprotective concentration, 1 hour prior to ROS exposure does not significantly prevent the rapid acidification in neuronal cultures (pH 6.98 ± 0.06) during ROS exposure [135].

Nicotinamide is considered to have protean endocrine effects [145,146], the ability to scavenge ROS, and offers cellular protection for both neuronal [140,147,148] and vascular cells [3,4,34,76]. In neuronal cell populations, nicotinamide protects against free radical injury [135], anoxia [132], excitotoxicity [149], homocysteine toxicity [150], ethanol-induced neuronal injury [151], and oxygen-glucose deprivation [140,152]. Nicotinamide prevents oxidant-induced apoptotic neuronal injury usually in a specific concentration range. Administration of nicotinamide in a range of 5.0–25.0 mmol/L significantly protect neurons during oxidative stress injuries (Figure 3). This concentration range is similar to other injury paradigms in both animal models [136] and in cell culture models [4,134,135]. In cortical neurons, nicotinamide antagonizes cell injury during ROS generating toxins such as tertiary butylhydroperoxide [153]. Nicotinamide also can protect both rod and cone photoreceptor cells against N-methyl-N-nitrosourea toxicity [136,154] as well as against glycation end products in all layers of the retina [155]. In animal studies, nicotinamide improves cognitive function, cell survival, and reduces edema following cortical trauma [156–161], limits axonal degeneration [162], reduces cerebral ischemia [163–165] sometimes more effectively in models that were absent of comorbidities [166], prevents spinal cord injury [167,168], and lessens disability in models of Parkinson’s disease in specific concentrations [169–171].

In addition to the observed neuroprotection with nicotinamide [140,147,148], the agent is also involved in the maintenance of vascular integrity [3,4,76]. For example, nicotinamide can protect the function of the blood brain barrier [156,157], influence arteriolar dilatation and blood flow [172], increase skin vascular permeability [173], potentially lead to decreased atherosclerotic plaque through inhibition of poly(ADP-ribose) polymerase [174], and promote platelet production through megakaryocyte maturation [175]. Nicotinamide also can maintain EC viability during ROS exposure [132–135,176]. Nicotinamide is believed to be responsible for the preservation of cerebral [177] and endocardial [178,179] ECs during models of oxidative stress [178,179]. However, recent reports suggest that pathways of nicotinamide also may have unclear vascular effects and may either prevent or contribute to atherosclerotic plaques over a three to six month progression [180]. It is possible that these events may occur during acidosis-induced cellular toxicity. During periods such as ischemia and oxidative stress, acidosis-induced cellular toxicity may ensue [142] and lead to subsequent mitochondrial failure [143]. Free radicals [86,144,181] can result in the disturbance of intracellular pH. In addition, modulation of intracellular pH is physiologically relevant for endonuclease activities during apoptosis [85,86,144]. As previously noted, nicotinamide cannot prevent cellular injury during intracellular acidification paradigms [135].

3. Nicotinamide and Inflammatory Cell Modulation

Closely tied to cellular survival and the ultimate disposal of non-functional cells is the activation of inflammatory cells [117,182]. As an example, when one considers disorders such as dementia [183] and inflammatory microglial cells of the brain, these cells can result in the phagocytic removal of both neurons and vascular cells [49,52,71]. During periods of inflammatory cell activation, microglia rely upon cytoprotective pathways [50,72] to proliferate and remove cells that are no longer functional [75,184]. Microglia can be beneficial in many ways to function as immune surveillance for toxic products [185], such as β-amyloid [186], block foreign microorganisms from entering the central nervous system and to allow for the repair of tissues composed of neuronal and vascular cells [50,187]. However, microglia have another side that may be detrimental to an organism. They can generate ROS [188,189], may worsen events with oxidative stress injury [190], and activate cytokines that in some circumstances may initially lead to cell proliferation [191], but later can lead to the demise of cells [192–194].

A number of cytoprotective agents rely upon the modulation of the immune system to control cellular survival. In particular, erythropoietin (EPO) is a prime example of a cytoprotective agent that is strongly associated with immune system pathways. Although EPO is approved by the Food and Drug Administration for the treatment of anemia and can have unwanted effects [127,195–197], it has recently been shown to significantly affect cell survival throughout the body [127,139,198,199], especially in regards to cellular proliferation [200–203]. EPO can reduce cytokine gene expression in endothelial cells exposed to tumor necrosis factor [204], prevent ulcer progression in cases of scleroderma [205], modulate inflammation during experimental autoimmune encephalomyelitis [206], reduce inflammation in murine arthritis models [207], and block primary microglial activation and proliferation during oxidative stress [25,78] to prevent phagocytosis of injured cells through pathways that involve cellular membrane PS exposure, protein kinase B (Akt) [49], and the regulation of caspases [78,208,209]. EPO can directly inhibit several pro-inflammatory cytokines, such as IL-6, tumor necrosis factor (TNF)-α, and monocyte chemoattractant protein 1 [199,210], and reduce leukocyte inflammation [211]. EPO also may foster the preservation of microglial cells for neuronal and vascular restructuring by preventing apoptotic injury in microglia [72,212].

EPO, although concentration dependent [78,138,208,213], can reduce cell injury during multiple events such as hyperoxia [214,215], hypoxia [78,138,216–220], parasitic infections [221–223], ROS exposure [64,208,224], ischemic/reperfusion insults [225–229], endotoxin shock [230,231], pulmonary disease [232–234], epileptic activity [235–237], elevated glucose exposure [238–240], excitotoxicity [224,241,242], mitochondrial failure [64,216,243], amyloid toxicity [25,244,245], cardiac and vascular injury [246–252], trauma [253–256], retinal disease [257], and renal failure [258–260].

Similar to agents such as EPO, nicotinamide can regulate cellular inflammation. Nicotinamide blocks pro-inflammatory cytokines, such as interleukin-1β, interleukin-6, interleukin-8, tissue factor, and TNF-α [261–264] as well as transforming growth factor (TGF) β2, IL-1β, TNF-α, and macrophage chemotactic protein-1 in hepatic cells [265]. Nicotinamide affects major histocompatibility complexes [266], inhibits intracellular adhesion molecule expression [267], and modulates TNF in vascular cells [266] that may account for the ability of nicotinamide to reduce demyelination in models of multiple sclerosis [268]. Nicotinamide also may control inflammatory mechanisms that lead to arthritis, such as the inhibition of collagen II expression [269] as well as contact hypersensitivity reactions [270]. Yet, the role of nicotinamide during inflammation is not entirely clear, since some investigations that examined the ability of oral nicotinamide administration to reduce cytokine production following endotoxin challenge in healthy volunteers did not demonstrate a significant effect upon serum cytokine levels [271].

4. Nicotinamide, Metabolic Disease, and Energy Management

Nicotinamide may have an important role during cellular energy management and metabolic disorders such as diabetes mellitus (DM). DM affects both young and older individuals [15,16]. Almost 20 million individuals in the United States and more than 165 million individuals worldwide suffer from DM with increasing incidence [272]. By the year 2030, it is predicted that more than 360 million individuals will be afflicted with DM and its debilitating conditions. Type 2 DM represents at least 80 percent of all diabetics and is dramatically increasing in incidence as a result of changes in human behavior and increased body mass index [15,91]. Type 1 insulin-dependent DM is present in 5–10 percent of all diabetics and affects three million individuals in the United States alone, but is increasing at a rate of 4%, especially in adolescent minority groups [15,91]. Additional concerns are evident with the knowledge that a significant portion of the population has undiagnosed diabetes and impaired glucose tolerance, illustrating the need for improved early diagnosis [273].

Patients with DM can develop significant neurodegenerative [34,39,91], affective disorders [274], cognitive loss [275], and cardiovascular disease [91,276]. Interestingly, the development of insulin resistance and the complications of DM can be the result of cellular oxidative stress [15,91]. Hyperglycemia can lead to increased production of ROS in endothelial cells, liver cells, and pancreatic β-cells [15,16,91] and lead to apoptotic injury [55,239]. Recent clinical correlates support these experimental studies to show that elevated levels of ceruloplasmin are suggestive of increased ROS [15,16,91]. Furthermore, acute glucose swings in addition to chronic hyperglycemia can trigger oxidative stress mechanisms, illustrating the importance for therapeutic interventions during acute and sustained hyperglycemic episodes [15,91].

In regards to nicotinamide and its role during metabolic disorders, nicotinamide appears to have a close relationship with metabolic pathways that may lead to clinical cognitive effects [277]. Treatment with nicotinamide can maintain approximately normal fasting blood glucose with streptozotocin-induced DM in animal models [278,279]. Nicotinamide also can reduce peripheral nerve injury during elevated glucose [280], lead to the remission of type 1 DM in mice with acetyl-l-carnitine [281], and can inhibit oxidative stress pathways that lead to apoptosis [10,133,134,151,282]. Nicotinamide also affects levels of O-N-acetylglucosamin(O-GlcNAc)ylated proteins [283] and can significantly improve glucose utilization, prevent excessive lactate production and improve electrophysiologic capacity in ischemic animal models [284]. Oral nicotinamide administration (1,200 mg/m2/day) protects β-cell function and prevents clinical disease in islet-cell antibody-positive first-degree relatives of type-1 DM [285]. In addition, nicotinamide administration (25 mg/kg) in patients with recent onset type-1 DM combined with intensive insulin therapy for up to two years after diagnosis significantly reduced HbA1c levels [286]. Potentially relevant to diabetic patients with renal failure, nicotinamide also has been shown to reduce intestinal absorption of phosphate and prevent the development of hyperphosphatemia and progressive renal dysfunction [287]. However, it is important to note that prolonged exposure to nicotinamide in some studies may lead to impaired β-cell function and reduction in cell growth [288,289]. Furthermore, nicotinamide also may inhibit P450 and hepatic metabolism [290] and play a role in the progression of other disorders such as Parkinson's disease [171].

Nicotinamide through NAD+ has a critical physiological role in cellular metabolism and can be directly utilized by cells to synthesize NAD+ [4,34,76]. Nicotinamide also participates in energy metabolism through the tricarboxylic acid cycle by utilizing NAD+ in the mitochondrial respiratory electron transport chain for the production of ATP, DNA synthesis, and DNA repair [291–293]. Furthermore, nicotinamide can significantly increase NAD+ levels in vulnerable regions of the ischemic brain, suggesting that nicotinamide may offer cytoprotection of injured tissue through the maintenance of NAD+ levels [177]. During axonal degeneration, nicotinamide also may promote neuroprotection through NAD+-dependent mechanisms [162].

The preservation of cellular energy reserves is dependent upon the maintenance of mitochondrial integrity during DM [294]. ROS exposure can result in the opening of the mitochondrial membrane permeability transition pore [52,135,208,295], reduce mitochondrial NAD+ stores, and result in apoptotic cell injury [83]. Free fatty acids can lead to ROS release and contribute to mitochondrial DNA damage and impaired pancreatic β-cell function [296]. In patients with type 2 DM, skeletal muscle mitochondria have been described to be smaller than those in control subjects [297]. In addition, a decrease in the levels of mitochondrial proteins and mitochondrial DNA in adipocytes has been correlated with the development of type 2 DM [298]. Insulin resistance in the elderly also has been associated with elevation in fat accumulation and reduction in mitochondrial oxidative and phosphorylation activity [299]. An association also exists with insulin resistance and the impairment of intramyocellular fatty acid metabolism in young insulin-resistance offspring of parents with type 2 DM [300].

Nicotinamide appears to function directly at the level of mitochondrial membrane pore formation [4,134,141] to prevent the release of cytochrome c [133] (Figure 4). Pretreatment of cells with either nicotinamide alone or in combination with the mitochondrial permeability transition pore inhibitor cyclosporin A prior to an injury paradigm can equally prevent mitochondrial membrane depolarization [301,302]. Nicotinamide can prevent the chemical induction of mitochondrial membrane depolarization during exposure to either tert-butylhydroperoxide or atractyloside [140]. There are additional pathways that nicotinamide may use to maintain cellular metabolic homeostasis through the maintenance of mitochondrial membrane potential [134,135]. Nicotinamide can phosphorylate Bad [133] to prevent mitochondrial membrane depolarization and subsequent cytochrome c release. In addition, nicotinamide may inhibit the assembly of the mitochondrial permeability transition pore complex similar to the action of cyclosporin A [303] as well as stabilize cellular energy metabolism since the maintenance of mitochondrial membrane potential is an ATP facilitated process [304].

5. Novel Intracellular Signaling for Nicotinamide
5.1. Forkhead transcription factors

Forkhead transcription factors of the “O” class (FoxOs) have recently been shown to mediate some of the biological effects of nicotinamide [305,306]. FoxO proteins either inhibit or activate target gene expression by binding bind to DNA through the forkhead domain that relies upon fourteen protein-DNA contacts [305,307–310]. According to X-ray crystallography [311] or nuclear magnetic resonance imaging [312], the forkhead domain is described as a "winged helix" as a result of a butterfly-like appearance. Members of this family that include FoxO1, FoxO3, FoxO4, and FoxO6 are found throughout the body and are expressed in tissues of the reproductive system of males and females, skeletal muscle, the cardiovascular system, lung, liver, pancreas, spleen, thymus, and the nervous system [117,309,310,313]. In addition, FoxOs govern a number of processes that involve cellular proliferation, degeneration, longevity, and neoplastic growth that may have associations with several novel pathways including wingless [26,55,77,184,314–320]. Other members of the forkhead family also rely upon wingless signaling that involve regulated as well as unchecked cellular growth [39,77,116,321,322].

Control of FoxO3a is a viable therapeutic target for agents such as metabotropic glutamate receptors [323], neurotrophins [324], cancer [117,309,325], and cytokines such as EPO [248] to increase cell survival. Recent work illustrates that FoxO3a may control early activation and subsequent apoptotic injury in microglia during Aβ exposure through caspase 3 [63]. Since Aβ exposure can facilitate the cellular trafficking of FoxO3a from the cytoplasm to the cell nucleus to potentially lead to apoptosis [63], one program in particular that may be vital for apoptotic injury appears to involve the activation of caspase 3. Aβ exposure leads to a rapid and significant increases in caspase 3 activity with six hours following Aβ administration, but that this induction of caspase 3 activity by Aβ requires FoxO3a, since loss of FoxO3a through gene silencing prevents the induction of caspase 3 activity by Aβ.

Nicotinamide has been shown to inhibit FoxO protein activity [140] and may be protective through two separate mechanisms of post-translational modification of FoxO3a [39,116,117,310,326] (Figure 5). Nicotinamide not only can maintain phosphorylation of FoxO3a and inhibit its activity to potentially block caspase 3 activity [140], but also can preserve the integrity of the FoxO3a protein to block FoxO3a proteolysis that can yield pro-apoptotic amino-terminal fragments [140]. During oxidative stress, an initial inhibitory phosphorylation of FoxO3a at the regulatory phosphorylation sites (Thr32 and Ser253) occurs [140,196]. However, loss of phosphorylated FoxO3a expression appears to subsequently result over twelve hours, possibly by caspase degradation, which potentially can enhance the vulnerability of neurons to apoptotic injury [140]. The loss of both FoxO3a phosphorylation and the integrity of this transcription factor may then lead to apoptosis. FoxO3a proteolysis occurs during cell injury yielding an amino-terminal (Nt) fragment that can become biologically active and lead to cellular injury [327]. Nicotinamide, through the phosphorylation of FoxO3a at regulatory sites that possess high affinity for protein kinase B (Akt) can prevent apoptotic cell injury [140]. In addition, modulation of caspase 3 activity by nicotinamide appears to be tied to a unique regulatory mechanism that blocks the proteolytic degradation of phosphorylated FoxO3a by caspase 3. Since FoxO3a has been shown to be a substrate for caspase 3-like proteases at the consensus sequence DELD304A [327], blockade of caspase 3 activity prevents the destruction of phosphorylated FoxO3a during oxidative stress [140], suggesting that nicotinamide maintains a regulatory loop through the modulation of caspase 3 and the preservation of phosphorylated FoxO3a integrity.

FoxO proteins also have been associated with cell longevity and aging as shown by early studies linking DAF-16 in Caenorhabditis elegans to increased longevity as well as the association with sirtuins [117,306,328–330]. Yet, the relationship among nicotinamide, FoxO transcription factors, and proteins that increased cellular lifespan is not entirely clear. For example, the sirtuin Sirt1 is a NAD+-dependent deacetylase and the mammalian ortholog of the silent information regulator 2 (Sir2) protein associated with increased lifespan in yeast. Some studies suggest that stimulation of Sirt1 during starvation is dependent upon FoxO3a activity as well as p53 [331]. In addition, during exercise, an up-regulation of FoxO3a and Sirt1 activity is observed in the heart [332], suggesting that physical activity may be beneficial for the cardiovascular system through FoxO proteins. Yet, other work has shown that Sirt1 may repress the activity of FoxO1, FoxO3a, and FoxO4, suggesting that cellular longevity may benefit from reduction in FoxO protein generated apoptosis [333].

In regards to nicotinamide, cellular protection and longevity, it appears that a reduction in nicotinamide levels during nicotinamidase expression supports increased cellular survival and longevity [334,335] (Figure 5). Nicotinamide can block cellular Sir2 by intercepting an ADP-ribosyl-enzyme-acetyl peptide intermediate with the regeneration of NAD+ (transglycosidation) [336]. Nicotinamidase expression which reduces nicotinamide concentrations prevents both apoptotic late DNA degradation and early PS exposure that may serve to modulate inflammatory cell activation. In addition, inhibition of sirtuin (Sirt1) activity either by pharmacological methods or siRNA gene silencing is detrimental to cell survival during oxidative stress and blocks nicotinamidase protection, further supporting that Sirt1 activity may be necessary for nicotinamidase protection during oxidative stress. Furthermore, nicotinamide offers gene regulation [337] and cellular protection in millimole concentrations against oxidative stress. Physiological concentrations of nicotinamide noncompetitively inhibit Sir2, suggesting that nicotinamide is a physiologically relevant regulator of Sir2 enzymes [338]. As a result, in relation to cell longevity, it is the lower concentrations of nicotinamide that can function as an inhibitor of sirtuins that are necessary for the promotion of increased lifespan and cellular survival [132–134,140,334,335,339], at least in yeast and metazoans [76,340,341]. Interestingly, it has been postulated that sirtuins also may prevent nicotinamide from assisting with DNA repair by altering the accessibility of DNA damaged sites for repair enzymes [342].

5.2. Protein kinase B (Akt), Bad, caspases, and mitogen-activated protein kinases

Post-translational modulation of FoxO proteins occurs through phosphorylation, acetylation, and ubiquitylation [306,326]. The serine-threonine kinase protein kinase B (Akt) is a principal pathway of phosphorylation of FoxOs that can block activity of these proteins [305,343]. Activation of Akt is usually “pro-survival” and cytoprotective, such as during cell proliferation [344], hyperglycemia [345], ischemia/stress [346,347], hypoxia [216], β-amyloid (Aβ) toxicity [25], cardiomyopathy [348], cellular aging [349], neurodegeneration [350,351], and oxidative stress [49,52,74]. Akt can prevent cellular apoptosis through the phosphorylation of FoxO proteins [117,309] and maintain FoxO transcription factors in the cytoplasm by association with 14-3-3 proteins and prevent the transcription of pro-apoptotic target genes [199,248].

Cytoprotection through Akt also can involve control of apoptotic inflammatory cell activation [52,74,208], maintenance of mitochondrial membrane potential (ΔΨm), and prevention of cytochrome c release [64,78,208]. Akt can prevent early apoptotic membrane PS exposure on injured cells and block the activation of microglia during oxidative stress [52,71,74]. Nicotinamide uses mechanisms that involve Akt to regulate microglial activation and proliferation [133,140] by blocking membrane PS exposure on cells and possibly preventing the shedding of membrane PS residues that is known to occur during apoptosis [352] (Figure 5).

Akt also regulates pathways that involve Bad, a pro-apoptotic Bcl-2 family member that becomes active through phosphorylation on its serine residues [83]. Phosphorylation of Bad by Akt leads to the binding of Bad with the cytosolic protein 14-3-3 to release Bcl-xL and allows Akt to block apoptosis [353]. Bcl-2 and Bcl-xL block Bax translocation to the mitochondria, maintain mitochondrial membrane potential, and prevent the release of cytochrome c from the mitochondria [208,354]. Nicotinamide can promote the phosphorylation of Bad during oxidative stress [133]. This phosphorylation of Bad by nicotinamide can be blocked by lack of Akt activity, suggesting that nicotinamide phosphorylates Bad through an Akt mediated pathway [343]. In addition, Akt may promote cell survival through the inhibition of apoptotic p53 transcriptional activity [355] that may be regulated by nicotinamide. Nicotinamide also has been shown to either directly limit the expression of p53 [153] or prevent an NAD-dependent p53 deacetylation induced by Sir2α [356].

Since Akt can prevent caspase activity [78,208,216], it is conceivable to assume that nicotinamide also may regulate caspase activity. Caspases are a family of cysteine proteases that are synthesized as inactive zymogens which are proteolytically cleaved into subunits during apoptosis [76,357,358]. The apoptotic-associated caspases include initiator caspases, such as caspase 2, 8, 9, and 10, that activate downstream effector caspases, resulting in an amplification of cascade activity. The initiator caspases consist of long N-terminal prodomains that contain caspase recruitment domains (CARDs) in caspase 2 and caspase 9 or death effector domains (DEDs) in caspase 8 and caspase 10. The effector caspases consist of caspase 3, 6, and 7 that function to directly cleave crucial cellular protein substrates to result in cell destruction [55,83,142,357]. Caspase 8 is as an upstream initiator of executioner caspases, such as caspase 3, and also leads to the mitochondrial release of cytochrome c [359,360]. Following caspase 8 and caspase 9 activation, caspase 3 directly leads to genomic DNA degradation [52,74,78].

Caspases 1 and 3 mediate genomic DNA cleavage and cellular membrane PS exposure [64,208, 361]. These caspases [4,134,135,141], in addition to caspase 8 and 9, are also tied to the direct activation and proliferation of microglia [52,74,78]. Caspase 1 is believed to be principally responsible for the externalization of membrane PS residues in several cell systems that can subsequently activate microglial phagocytosis [80,362]. Furthermore, caspase 9 is activated through a process that involves the cytochrome c -apoptotic protease-activating factor-1 (Apaf-1) complex [79,363]. In regards to membrane PS exposure, nicotinamide prevents PS externalization primarily through the inhibition of caspase 1 -like activity [133] (Figure 5). Nicotinamide also prevents genomic DNA cleavage as well as early membrane PS exposure through caspase 8 and caspase 9 - like activities [4,133–135,140]. The precise pathways that are necessary for nicotinamide to modulate caspase pathways remain under investigation. Although some "anti-apoptotic" proteins, such as EPO [199,208] modulate both Apaf-1 expression and cytochrome c release, protection through nicotinamide remains independent from Apaf-1 [140]. However, nicotinamide can significantly prevent cell injury by inhibiting caspase 9 - like activity directly [140].

Nicotinamide also relies upon the stress activated family of mitogen-activated protein kinases (MAPK) that includes the p38 kinases (MAPKp38) and the c-Jun N-terminal kinases (MAPKJNK). The family of MAPKs consists of the subgroups that include ERK1 (MAPKERK1/p44), ERK2 (MAPKERK2/p42), the JNKs (MAPKJNK), and p38 MAPKs (MAPKp38). Although significant activation of MAPKp38 and MAPKJNK is present in cells during oxidative stress [4,132,135,141] and c-Jun leads to apoptosis through transcription activation of some pro-apoptotic genes [364], nicotinamide does not appear to alter the activity of either MAPKp38 or MAPKJNK [133]. These results suggest that nicotinamide cytoprotection does not require the MAPKp38 and MAPKJNK pathways [4,132,135,141].
5.3. Poly (ADP-ribose) polymerase (PARP)

Nicotinamide has an intimate relationship with poly (ADP-ribose) polymerase (PARP) that also been recently associated with both vascular and neurodegenerative disorders [76,174,365] (Figure 5). PARP is a nuclear protein that binds to DNA strand breaks and cleaves NAD+ into nicotinamide and ADP-ribose. PARP catalyses the synthesis of poly (ADP-ribose) from its substrate NAD+, which stimulates the process of DNA repair [366]. Nicotinamide concentrations of at least 1 mM have been shown to provide sufficient stores of NAD+ for PARP activation [367]. Nicotinamide can prevent PARP degradation and allow for DNA repair through the direct inhibition of caspase 3 [133–135]. In contrast, elevated concentrations of nicotinamide can lead to PARP degradation and apoptotic injury [368].

However, other work illustrates that a reduction in PARP activity may enhance cell survival, such as during injury paradigms with photoreceptor cells [369], during homocysteine toxicity [370], during cerebral ischemia [371], or during free radical injury [372,373]. Prevention of NAD+ depletion during enhanced PARP activity also has been demonstrated to prevent cellular lysis during oxidative stress [374]. In human blood lymphocytes during oxidative stress, nicotinamide may block necrotic death through pathways that limit excessive PARP activity that can consume essential NAD+ stores [375]. During diabetic neuropathy, nicotinamide reduces PARP activity to partially restore vital NAD+ and ATP [376].

Inhibition of PARP activity by nicotinamide also may be critical for disorders such as Alzheimer's disease. The National Institute on Aging estimates that almost five million people in the United States have Alzheimer’s disease and worldwide more than twenty-four million people suffer from Alzheimer’s disease, pre-senile dementia, and other disorders of cognitive loss. In Alzheimer's disease [99], Aβ is toxic to cells [25,26,121] and is associated with the phosphorylation of the forkhead transcription factors that can be blocked by scavengers of oxidative stress [377]. A prior pilot study has suggested that administration of nicotinamide adenine dinucleotide (NADH) in patients with Alzheimer’s may show improvement in their cognitive function [378]. More recently, dietary niacin intake examined in a series of patients aged 65 and older has been implicated as a protectant against the development or progression of Alzheimer's disease [379]. Interestingly, it has been shown that in patients with Alzheimer's disease, both PARP and poly(ADP-ribose) is present in the frontal and temporal cortex more frequently than in controls, suggesting that increased levels of functional PARP enzyme are present to potentially lead to the depletion of NAD+ stores [380]. In addition, Aβ toxicity may require increased PARP activity [381].

6. Conclusions

Therapeutic innovation relies heavily upon the understanding and emerging knowledge of the cellular pathways that govern disease. Yet, no therapeutic modality can be used with clear focus and caution to gain the greatest clinical efficacy with the least amount of unwanted detrimental effects. As a result, nicotinamide clearly comes under such constraints. As an agent that offers broad cytoprotective effects that may be applicable to multiple disorders of the cardiovascular, nervous, immune, and metabolic pathways, nicotinamide also has complex biological roles. For example, nicotinamide pathways that rely upon FoxO modulation may not consistently lead to controlled enhanced cell survival, but rather with unchecked cellular proliferation that leads to cancer. Furthermore, nicotinamide offers a fine line with concentration administration with lower concentrations of nicotinamide possibly negating sirtuin activity and decreasing lifespan in organisms. In addition, protection against PARP or preservation of its activity by nicotinamide also may sometimes lead to unwanted outcomes. PARP activation can deplete NAD+, lower ATP production, and precipitate cell death. Under other conditions, nicotinamide has been described as an agent that limits cell growth and promotes cell injury. Nicotinamide in the presence of transforming growth factor β-1 can block hepatic cell proliferation and lead to apoptosis with caspase 3 activation [265]. During moderate temperature hyperthermia or carbogen breathing, nicotinamide also can result in enhanced solid tumor radiosensitivity and assist with tumor load reduction [382]. With these considerations for nicotinamide, it is clear that this agent holds great potential for multiple disease entities, but the development of new therapeutic strategies with nicotinamide rests heavily upon the elucidation of the intimate relationship nicotinamide holds with novel pathways that include forkhead transcription factors, sirtuins, Akt, caspases, MAPK, and PARP.

For me this all means that nicotinamide is a double edged sword: beneficial in the right dosage, detrimental if the dose is too high or too low. And currently there seems not to be a clear indication how to determine the right dose for the individual ... So to experiment with high doses of niacin might potentially (in susceptible individuals) be fraught with danger.
 
Another short article about the link of nicotinamide and nicotine in the treatment of pemphigus, which is a skin condition.

Nicotine and Pemphigus
Sergei A. Grando, MD, PhD, DSc; Mark V. Dahl, MD

Arch Dermatol. 2000;136(10):1269-1269. doi:10.1001/archderm.136.10.1269

We read with interest the article by Mehta and Martin1 titled "A Case of Pemphigus Vulgaris Improved by Cigarette Smoking." We have a better explanation why cigarette smoking might be expected to improve skin lesions of patients with pemphigus vulgaris, particularly when nicotine in cigarette smoke contacts mucous membranes.

Human keratinocytes contain an elaborate acetylcholine network. Specifically, human keratinocytes synthesize, store, release, and degrade acetylcholine.2 Keratinocytes contain choline acetyltransferase and acetylcholinesterase,2 and have on their cell membranes both muscarinic and nicotinic receptors for acetylcholine.3 - 4 Both nicotinic and muscarinic acetylcholine receptors regulate cell-to-cell adhesion of human keratinocytes (reviewed in Grando5 ). Interaction of nicotine (and other nicotinic agonists) with nicotinic acetylcholine receptors on keratinocytes opens ion gates in the cell membrane to help increase cell-to-cell adherence, stop acantholysis, and stimulate keratinocytes to move laterally to heal erosions.6

We believe the keratinocyte cholinergic system is altered in pemphigus.7 Keratinocytes undergoing acantholysis in a tissue culture plate can be quickly restored to confluence by adding acetylcholine.8 Therefore, in a patient such as the one described by Mehta and Martin,1 nicotine might (1) compete with the disease-causing pemphigus antibodies preventing them from attaching to keratinocytes or (2) block the intracellular signaling pathways that mediate the acantholytic effects of pemphigus antibodies. Approximately 85% of patients with pemphigus develop antibodies in their serum to one or more types of keratinocyte acetylcholine receptors.9

Since human keratinocytes have nicotinic acetylcholine receptors, and since nicotine can bind to them, and since activation of nicotinic cholinergic receptors increases cell-to-cell adhesion and promotes lateral migration of keratinocytes, one might predict that smoking would, indeed, improve pemphigus. Nicotine could be delivered to keratinocytes of mucous membranes either topically through the smoke or systemically via absorbed nicotine.

One might also predict that nicotinamide, like nicotine, might improve pemphigus, since it too is a nicotinic agonist.10 Additionally, nicotinamide inhibits the enzyme acetylcholinesterase that degrades acetylcholine.11 Indeed, nicotinamide has helped patients with pemphigus too (reviewed in Chaffins et al12 ).

Thus, the observation that a patient's erosions of the buccal mucosa and blisters on his face and body improved after cigarette smoking suggests that nicotine and other cholinergic agents might help certain patients with pemphigus. The results of our studies defining the keratinocyte cholinergic network provide a rationale for their use.
 
LQB said:
anart said:
LQB said:
Laura said:
The info about Vitamin B3 and its relation to nicotine is rather fascinating. I've got some downstairs so I think I'll try taking it for awhile.

If you're going to take niacin (as opposed to niacinamide) watch out for the flush. The first time you take it, even 100mg can give a doosy of a flush.

Added: :shock: :O :scared:

Yep, this happened to me last week and I was literally neon pink for about a half hour - not very comfortable!

The first time I did about 200 mg and the flush went all the way down to my knees - not to mention the neon pink and itching. Fred Klenner MD suggests that you resist itching by pressing down on the skin or using an ice cube.

I forget at the moment where I read it, but I read that the itch factor in the flush comes from histamine being released and that ascorbic acid should bind (I think) with the histamine to stop the itch. I've tried this and flushed a lot (I took a lot--not sure how much) and was hot and cold at the same time, flushed as a beet except for my very extremities, but I was either not itchy at all or not very itchy, FWIW.
 
Back
Top Bottom