Wednesday, August 30, 2017

Melatonin and TNPIX Modulates ROS IN Acute and Chronic Oxidative Stress

This first paragraph implies that cleaning up oxidative stress inhibits or reverses malignancy, diabetes, neurodegenerative diseases and aging.

It further shows that TNPIX is elevated in oxidative stress.
No stress low TNPIX.
Challenging stress low levels.
Overwhelming stress high levels.
TNPIX modulates ROS signaling AND has both pro oxidant and antioxidant effects dependent on level. (Interestingly melatonin has that same duality and may imply it shares TNPIX ROS modulation function.)
The article implicitly states that TNPIX declines with age.
It is firmly established that Melatonin declines with age.

High levels are associated with growth arrest and tumor suppression.
In diabetes formation TNPIX is elevated and beta cells decline (arrested.)
Challenging levels signal that "more antioxidant capacity" is needed.

Oxidative stress accelerates aging and reduces stem cells number and quality leading to stem cell exhaustion.
Does treating oxidative stress with hydrogen rich water, sulforaphane, melatonin, wheat germ spermidine reduce both aging and cancer?

Sepsis (and cardiac or respiratory failure ) is a major acute oxidative stressor.  Antioxidant rescue of sepsis model animals that targets mitochondria increase survival and organ function survival acutely.

Does increasing net total antioxidant capacity protect against age related chronic oxidative stress including diabetes, neurodegenerative disease and malignancy?

TXNIP Maintains the Hematopoietic Cell Pool by Switching the Function of p53 under Oxidative Stress

Oxidative stress occurs mainly due to excessive accumulation of cellular reactive oxygen species (ROS) or deficiency of antioxidant defense system. Oxidative stress often leads to pathologic diseases such as diabetes, neurodegenerative diseases, and cancer (Hole et al., 2011Sinha et al., 2013). There is growing evidence that balanced regulation of ROS is critical for hematopoiesis. Hematopoietic cells are vulnerable to oxidative stress, and malignancy of hematopoietic tissues is observed in the presence of chronic oxidative stress (Ghaffari, 2008). Homeostatic regulation of redox status in hematopoietic tissues is important for normal hematopoiesis.
Thioredoxin-interacting protein (TXNIP) is a 397 amino acid, 50 kDa protein that belongs to the arrestin family, and Txnip−/− mice show a high incidence of hepatocellular carcinoma (HCC) (Jeong et al., 2009Kwon et al., 2011Lee et al., 2005Song et al., 2003). TXNIP expression is reduced in many types of tumors, and TXNIP overexpression inhibits tumor growth by blocking cell-cycle progression (Han et al., 2003). The numbers of natural killer (NK) cells in the bone marrow (BM) of Txnip−/− mice are reduced, and the long-term reconstituting HSC population shows an exhausted phenotype and is reduced in frequency (Jeong et al., 2009Lee et al., 2005).
The tumor suppressor p53 plays a key role in restricting the expansion of abnormal cells through either growth arrest or apoptosis in response to genotoxic stresses (Olovnikov et al., 2009Sablina et al., 2005). The p53 pathway is regulated by mouse double minute 2 (MDM2), an E3 ubiquitin ligase that targets the p53 protein for proteasomal degradation (Sasaki et al., 2011). p53 engages powerful prosurvival pathways by inducing the expression of antiapoptotic or antioxidant genes (Bensaad and Vousden, 2007Jänicke et al., 2008). In addition, p53 is a critical regulator of HSC quiescence through its target genes (Liu et al., 2009). Previous reports imply that the protective or antiaging effects of TXNIP are important in maintaining hematopoietic cell pool (Jeong et al., 2009Kim et al., 2007).
In this study, we demonstrate that Txnip−/− hematopoietic cells had defects in the regulation of ROS levels and were more sensitive than wild-type cells to oxidative stress. We also demonstrated that TXNIP exerted its antioxidant effects in hematopoietic cells by stabilizing p53 under oxidative stress. Our findings suggest that TXNIP plays a critical role in the antioxidant defense mechanisms of hematopoietic cells by activating the p53 pathway during oxidative stress.
To investigate the effects of TXNIP deficiency on hematopoiesis, we analyzed the frequency of hematopoietic stem cells (HSCs) and hematopoietic progenitors from young (12 weeks) and old (22–23 months) Txnip+/+ (wild-type [WT]) and Txnip−/− (KO) mice. Consistent with previous reports (Geiger and Van Zant, 2002Sudo et al., 2000), old WT mice showed much higher frequencies of HSCs and hematopoietic progenitors, but old KO mice showed relatively decreased frequencies (Figure 1A). Next, we performed a competitive repopulation assay and a serial bone marrow transplantation (BMT) experiment (Figure 1B). We transplanted lineagec-Kit+Sca-1+ (LKS) cells or WBM (whole bone marrow) cells from young (12 weeks) and old (22–23 months) mice (CD45.2+) with competitor WBM cells (CD45.1+) into congenic recipients (CD45.1+). Donor-derived cell populations of old WT LKS or WBM cell-transplanted recipients showed little change, but those of old KO LKS or WBM cell-transplanted recipients were markedly decreased (Figure 1C and Figure S1A available online). Also, WBM cells of recipients from young and old KO mice showed a greater reduction in donor-derived cell populations of HSCs and progenitors than those from WT mice (Figures 1D, S1B, and S1C), mostly due to the reduced frequency of HSCs and progenitors in donor WBM or LKS as shown in Figure 1A. Next, we performed a serial BMT experiment. Donor-derived CD45.2+ cells were dramatically decreased in the KO-derived recipient cells (Figures 1E and 1F).
The exhaustion of primitive HSCs is believed to result from increased ROS accumulation following serial transplants, which are a critical determinant of HSC pool maintenance (Abbas et al., 2010Ito et al., 2006). We found dramatically increased ROS levels in old KO BM cells compared with those from WT littermates (Figure 2A). To assess the effects of the intrinsic increase in ROS on the maintenance of KO BM cells, we transplanted WBM cells (CD45.2+) into lethally irradiated WT congenic (CD45.1+) recipients. After 9 months, we confirmed higher levels of ROS in the KO-derived BM cells (Figure 2B). Our observations suggested that TXNIP plays a critical role in hematopoietic cell antioxidant defense through mechanisms other than its known prooxidant function as an inhibitor of thioredoxin (Trx) (Lee et al., 2005Patwari et al., 2006Schulze et al., 2002).
To examine the specificity of antioxidant defense by TXNIP in hematopoietic cells, we analyzed the levels of ROS in WT and KO mouse embryonic fibroblast (MEF) and lung fibroblast cells under oxidative stress. Interestingly, WT MEF and lung fibroblast cells showed the prooxidant function of TXNIP following oxidative stress (Figures S2A–S2D), indicating that TXNIP regulates ROS levels in a cell-type-specific manner. Next, to validate the antioxidant function of TXNIP in hematopoietic cells under oxidative stress, we intraperitoneally (i.p.) injected paraquat (PA), a strong oxidative stress inducer, into young mice. Consistent with our observations of old KO mice, young KO mice showed higher ROS levels and increased cell death in BM cells following PA challenge (Figures 2C and 2D). KO HSCs showed lower ROS levels than nonprimitive cells but also hypersensitivity under oxidative stress. KO HSCs entered into the cell cycle at an early time (0–12 hr) but showed decreased proliferating rates and frequencies at a late time (48–96 hr) following PA challenge (data not shown) (Macip et al., 2003). Taken together, the above data indicate that TXNIP shows cell-type-specific antioxidant function and plays an important role in the maintenance of both HSCs and nonprimitive hematopoietic cells by regulating ROS and cell death following oxidative stress.


Joseph Thomas (Tony) L

Saturday, August 26, 2017

Prevent Osteoarthritis (and Diabetes) by Increasing Net Antioxidant Capacity

Aging is a risk factor for osteoarthritis.
Diabetes is a risk factor for osteoarthritis.
Diabetes accelerates aging.
Why?

Net total antioxidant capacity decreases with both aging and diabetes.
Nrf2 increases net total antioxidant capacity.

In this study bolded  text showing DM vs DM- OA varied according to Nrf2 status and therefore net total antioxidant capacity varied!

How?
See preceding posts.

The nuclear factor-erythroid 2-related factor/heme oxygenase-1 axis is critical for the inflammatory features of type 2 diabetes-associated osteoarthritis

  1. J&eacuter&eacutemie Sellam2
+ Author Affiliations
  1. * Corresponding author; email: francis.berenbaum@sat.aphp.fr
  1. Author contributions: CV-G, AC, M-CL, XH, SK-R, RS, FB and JS were responsible for the study design, manuscript preparation, and data interpretation. AS organized and collected the human tissue samples and participated in designing the experiments with human tissue and in data interpretation. CV-G, AC and AP performed the experiments. SK-R was responsible for generating the Nrf-2-/- mice and was involved in data interpretation. All authors reviewed and approved the final manuscript.
Epidemiological findings support the hypothesis that type 2 diabetes mellitus (T2DM) is a risk factor for osteoarthritis (OA). Moreover, OA cartilage from patients with T2DM exhibits a greater response to inflammatory stress, but the molecular mechanism is unclear. To investigate whether the antioxidant defense system participates in this response, we examined here the expression of nuclear factor-erythroid 2-related factor (Nrf-2), a master antioxidant transcription factor, and of heme oxygenase-1 (HO-1), one of its main target genes, in OA cartilage from T2DM and non-T2DM patients, as well as in murine chondrocytes exposed to high glucose (HG). Ex vivo experiments indicated that Nrf-2 and HO-1 expression is reduced in T2DM vs. non-T2DM OA cartilage (0.57-fold [Nrf-2] and 0.34-fold [HO-1]), and prostaglandin E2 (PGE2) release was increased in samples with low HO-1 expression. HG-exposed, IL-1β-stimulated chondrocytes had lower Nrf-2 levels in vitro, particularly in the nuclear fraction, than chondrocytes exposed to normal glucose (NG). Accordingly, HO-1 levels were also decreased (0.49-fold) in these cells. The HO-1 inducer cobalt protoporphyrin-IX more efficiently attenuated PGE2 and IL-6 release in HG+IL-1β-treated cells than in NG+IL-1β-treated cells. A greater reduction in HO-1 expression and increase in PGE2/IL-6 production were observed in HG+IL-1β-stimulated chondrocytes from Nrf-2-/- mice than in chondrocytes from wild type mice. We conclude that the Nrf-2/HO-1 axis is a critical pathway in the hyperglucidic-mediated dysregulation of chondrocytes. Impairments in this antioxidant system may explain the greater inflammatory responsiveness of OA cartilage from T2DM patients and may inform treatments of such patients.

Saturday, August 19, 2017

Aging and Age Reversal Explained by Combination of Two Theories of Aging

A combo of two theories explain aging.
1.  Oxidative damage.
2.  Impaired proteostasis.

How can "aging" be measured?
1.  Methylation of genes accumulate signifying genes are epigenically turned off.
2. Percentage or accumulation of senescent cells signifying survival without quality control mechanisms such as proteostasis.  A subset of these senescent growth arrested cells become cancer or contribute to neurodegenerative changes.

What proof is known?
1.  Cell, tissue and organ function is inversely related to methylation counts or "biological age."
2.  Cell penetrating protein that kills senescent cells allows stem cells to replace the non functioning cell with a fully functioning and non toxic cell.

Senescent cells arise when pro oxidant forces exceed antioxidant and proteostasis forces.
Proteostasis is impaired by clock gene reduction of spermidine synthesis and reduction of dietary Spermidine supplements.
Antioxidant forces are reduced by methylation of ARE antioxidant response element gene due to pro oxidation forces.

Therefore, accumulation of senescent cells and abnormal protein aggregates in cells indicate inadequate autophagy, proteostasis.

What is the solution to biological AND chronological aging?
Hydrogen rich water, sulforaphane, melatonin and wheat germ/spermidine that activate Nrf2 that stimulates ARE and increases protective antioxidant capacity and restores proteostasis/autophagy.  
Cell, tissue, organ and organism quality control is restored and aging slowed down until agents that promote senescent cells to die via apoptosis can restore a young phenotype is available.

Happily (n)ever after: Aging in the context of oxidative stress, proteostasis loss and cellular senescence

Aging is a complex phenomenon and its impact is becoming more relevant due to the rising life expectancy and because aging itself is the basis for the development of age-related diseases such as cancer, neurodegenerative diseases and type 2 diabetes. Recent years of scientific research have brought up different theories that attempt to explain the aging process. So far, there is no single theory that fully explains all facets of aging. The damage accumulation theory is one of the most accepted theories due to the large body of evidence found over the years. Damage accumulation is thought to be driven, among others, by oxidative stress. This condition results in an excess attack of oxidants on biomolecules, which lead to damage accumulation over time and contribute to the functional involution of cells, tissues and organisms. If oxidative stress persists, cellular senescence is a likely outcome and an important hallmark of aging. Therefore, it becomes crucial to understand how senescent cells function and how they contribute to the aging process. This review will cover cellular senescence features related to the protein pool such as morphological and molecular hallmarks, how oxidative stress promotes protein modifications, how senescent cells cope with them by proteostasis mechanisms, including antioxidant enzymes and proteolytic systems. We will also highlight the nutritional status of senescent cells and aged organisms (including human clinical studies) by exploring trace elements and micronutrients and on their importance to develop strategies that might increase both, life and health span and postpone aging onset.


Monday, August 14, 2017

Speculation About Hypometylation in Autism and Aging

This article shows two changes from normal in autism.

1.  Reduced antioxidant capacity.  Sulforaphane an Nrf2 agonist increases antioxidant capacity and improved behavior in autistic children.
2.  Hypomethylation of gene promoter sites which also occurs in aging and is associated with methylation of histone gene promoter sites.  I suspect that gene specific methylation of histone production dysregulates normal epigenetic control of acetylation deacetylatoon of genes.  Without adequate histones production, methylated CpG gene promoter sites cannot lay down histone to fully block gene transcription, therefore control of gene expression is haphazard and incomplete.  Melatonin through its HDAC INHIBITIONS and ANTIOXIDANT functions might demethylate the histone gene promoter site and restore histone gene function and thereby return command and control for non impaired epigenetic control of cellular functions.  It should be stated that methylation is a dimmer not a light switch because of redundancy of gene copies to express certain products. HIIE and fasting produced beta hydroxybutyrate is also an HDAC INHIBITOR.

CONJECTURE:. HRW, sulforaphane, spermidine, melatonin and lifestyle changes could ameliorate autism through epigenetic change and return to normal command and control of gene expression.

It also follows that hypomethylation secondary to aging related methylation or acetylation of histone CpG gene promoter site would be more and better regulated! This is the reason that heterochromic parabiosis results in a plasma transfer of a youthful factor to the older mouse and an aging factor to the younger mouse with conjoined circulation.  The aging clock genes express a ratio of protein products that stimulate and inhibit histone expression in the elderly cell that mimics histone gene promoter site methylation in the young "autistic" cell.

The hypomethylation of aging increases exponentially and is counterintuitive to epigenetic aging measures of biological aging based on number of methylated genes in peripheral blood.  I interpret this to mean that epigenetic biological aging is directly proportional to methylated gene CpG promoter sites to mortality predictions with a 96% correlation before histone gene promoter methylation that causes exponential increases in its opposite, hypomethylation. 

In effect autistic children lose control of gene expression through methylation of histone gene promoter site.
Aging adults in final descent toward mortality, show clock gene related histone associated down regulation of histone production.
An old gamete  cell or stem cell escapes by turning back the clock genes or increasing the ratio of histone producing/histone inhibiting gene products as highlighted  by Rando in interpreting parabiosis experiments. This reverses replicative aging only.  DNA damage is wear and tear aging and is the homeostasis of gene repair, cell energetics and proteostasis forces working under the histone ratio drag of replicative aging.

Metabolic Imbalance Associated with Methylation Dysregulation and Oxidative Damage in Children with Autism

Oxidative stress and abnormal DNA methylation have been implicated in the pathophysiology of autism. We investigated the dynamics of an integrated metabolic pathway essential for cellular antioxidant and methylation capacity in 68 children with autism, 54 age-matched control children and 40 unaffected siblings. The metabolic profile of unaffected siblings differed significantly from case siblings but not from controls. Oxidative protein/DNA damage and DNA hypomethylation (epigenetic alteration) were found in autistic children but not paired siblings or controls. These data indicate that the deficit in antioxidant and methylation capacity is specific for autism and may promote cellular damage and altered epigenetic gene expression. Further, these results suggest a plausible mechanism by which pro-oxidant environmental stressors may modulate genetic predisposition to autism.


Monday, August 7, 2017

Strategy to Augment Cancer Treatment and Prevention

New strategy to augment cancer prevention and treatment suggested by research showing that cancer is a metabolic disease and is reduced by restoring optimum mitochondrial metabolism.

This article notes that cancer turns off  the enzyme that ultimately converts glucose into fuel for mitochondria.
By depriving the mitochondria of fuel, ROS is reduced.
ROS radical oxygen species kills cancer cells primed for apoptosis.
Blocking or reducing that enzyme directly or indirectly by fasting/ nutrient deprivation results in increased levels of ROS which drives cancer cell death that can be augmented with cancer treatments.

The Warburg Effect is glucose burning without mitochondrial help.  This allows glucose imaging molecules to show cancer like metabolism on PET scans as highly metabolic cells that light up.

It also suggests that increasing mitochondrial metabolism would increase ROS and cancer cell death.  For example the Ketogenic diet.  
If, however, the ketogenic diet is "running hot" with meaningless noise or H2O2 the UPR unfolding protein response dissipates the ROS  produced.

How to run hot with signaling ROS that drives cancer cell death apoptosis without activating the unfolded protein response UFP meant to protect the mitochondria and ER endoplasmic reticulum from harming the non resilient cell?

Clean up noise, ROS, that are unimportant to signaling. (Hydrogen Rich Water)
Block the UPS with mitochondrial/ER specific antioxidant function. (Wheat germ/Spermidine)(sulforaphane)
Add ketones as fuel to produce " important ROS signaling" that kill cancer cells. (Fasting) (modified fasting diet per Valter Longo) (medium chain triglyceride or coconut oil supplements that are metabolized into ketones by the liver- bulletproof coffee)

Breaking Mitochondrial Fasting for Cancer Treatment: Old Wine in New Bottles

Many malignant cells exhibit the Warburg effect, first described by Otto Warburg in 1924 (1). Since then, this phenomenon has been well documented and characterized by augmented aerobic glucose uptake, glycolytic shift, decreased utilization of pyruvate by mitochondria, and increased lactate production, all of which are mainly controlled by hypoxia-induced factor 1 alpha (HIF-1α) and Myc overexpression in neoplastic cells (1). HIF-1α and Myc attenuate mitochondrial function by activating pyruvate dehydrogenase kinases (PDKs), which phosphorylate and inactivate the pyruvate dehydrogenase complex (PDC). Hence, inhibiting the PDKs can shuttle more pyruvate into mitochondrial oxidative phosphorylation and away from lactate synthesis, resulting in oxidative stress, triggering apoptosis, and augmentating host immuno-surveillance, ultimately leading to diminished tumor proliferation (2). Additionally, the Warburg effect leads to increased lactate in cellular and extracellular compartments....

Saturday, August 5, 2017

Preventing Age and Pathological Thrombosis Using Hydrogen Rich Water

Conjecture:. H2S inhibits pathologic platelet activation, adhesion and clots.

Molecular hydrogen has a similar effect as an antioxidant modifier.
H2S is the endogenous gasotransmitter or hydrogen donor.
H2 is the active moiety.
This is analogous to beta hydroxybutyrate the endogenous HDAC inhibitor and starvation gene set gene promoter and Na Butyrate the active moiety that shares the same function in the cell nucleus.

Would molecular hydrogen like H2S prevent clots without increasing bleeding?  Molecular hydrogen reduces oxidative stress without abrogating redox signaling.  It might be a perfect agent for preoperative and postoperative prophylaxis of DVT or adjunctive therapy in DVT.

Calcium sensing receptor initiating cystathionine-gamma-lyase/hydrogen sulfide pathway to inhibit platelet activation in hyperhomocysteinemia rat

H2S is involved in protection of ECs and the mediation of anti-thrombotic in HHcy.
Hyperhomocysteinemia (HHcy, high homocysteine) induces the injury of endothelial cells (ECs). Hydrogen sulfide (H2S) protects ECs and inhibits the activation of platelets. Calcium-sensing receptor (CaSR) regulates the production of endogenous H2S. However, whether CaSR inhibits the injury of ECs and the activation of platelets by regulating the endogenous cystathionine-gamma-lyase (CSE, a major enzyme that produces H2S)/H2S pathway in hyperhomocysteinemia has not been previously investigated. Here, we tested the ultrastructure alterations of ECs and platelets, the changes in the concentration of serum homocysteine and the parameters of blood of hyperhomocysteinemia rats were measured. The aggregation rate and expression of P-selectin of platelets were assessed. Additionally, the expression levels of CaSR and CSE in the aorta of rats were examined by western blotting. The mitochondrial membrane potential and the production of reactive oxygen species (ROS) were measured; the expression of phospho-calmodulin kinases II (p-CaMK II) and Von Willebrand Factor (vWF) of cultured ECs from rat thoracic aortas were measured. We found that the aggregation rate and the expression of P-selectin of platelets increased, and the expression of CaSR and CSE decreased in HHcy rats. In the ECs of HHcy group, the ROS production increased and the mitochondrial membrane potential decreased markedly, the expression of CSE and the p-CaMK II increased after treatment with CaSR agonist while decreased upon administration of U73122 (PLC-specific inhibitor) and 2-APB (IP3 Receptor inhibitor). CaSR agonist or NaHS significantly reversed the ECs injured and platelet aggregation caused by hyperhomocysteinemia. Our results demonstrate that CaSR regulates the endogenous CSE/H2S pathway to inhibit the activation of platelets which concerts the protection of ECs in hyperhomocysteinemia.

Tuesday, August 1, 2017

Tuning Cellular Power Production with Safety; Direct, Indirect Antioxidants and Proteostasis

Oxidative stress is harmful and increases with age and disease.
Anti oxidant capacity declines with age AND parallels the amount of Nrf2 activation.
In the abstract below showing that antioxidants disappoint in cardiovascular studies they do not acknowledge the importance of ROS as metabolic signaling.  One must increase antioxidant CAPACITY and activity without impacting signaling.

How?

Hydrogen rich water removes noise level oxidants only like a filter in a flow of signal rich electrons.
Secondly, sulforaphane increases antioxidant enzymes for PRN use with activation.

Finally, being able to translate signaling from genes requires proteostasis, making, refolding and removing proteins which is a function of wheat germ spermidine that declines with age in parallel to declining antioxidant CAPACITY.

One can measure direct antioxidant capacity and antioxidant stress in peripheral blood.
But can one measure available potential antioxidant capacity?
This would require a challenge or stress test.

These studies have already been performed in part.
E.g..  The product of order and power or speed parameters of HIIE sprints is increased by hydrogen rich water which preserves safe oxidation independent of baseline VO2 max.  The VO2 is directly proportional to the mass of mitochondria.  It had been shown that HYDROGEN RICH WATER makes each mitochondria more efficient and maintains a higher proton gradient which ultimately translates into POWER.

Homeostasis or balance of oxidative signaling or flux.

Abstract

Metabolism of oxygen by cells generates potentially deleterious reactive oxygen species (ROS). Under normal conditions the rate and magnitude of oxidant formation is balanced by the rate of oxidant elimination. However, an imbalance between prooxidants and antioxidants results in oxidative stress, which is the pathogenic outcome of oxidant overproduction that overwhelms the cellular antioxidant capacity. The kidney and vasculature are rich sources of NADPH oxidase–derived ROS, which under pathological conditions play an important role in renal dysfunction and vascular damage. Strong experimental evidence indicates that increased oxidative stress and associated oxidative damage are mediators of renovascular injury in cardiovascular pathologies. Increased production of superoxide anion and hydrogen peroxide, reduced nitric oxide synthesis, and decreased bioavailability of antioxidants have been demonstrated in experimental and human hypertension. These findings have evoked considerable interest because of the possibilities that therapies targeted against free radicals by decreasing ROS generation or by increasing nitric oxide availability and antioxidants may be useful in minimizing vascular injury and renal dysfunction and thereby prevent or regress hypertensive end-organ damage. This article highlights current developments in the field of ROS and hypertension, focusing specifically on the role of oxidative stress in hypertension-associated vascular damage. In addition, recent clinical trials investigating cardiovascular benefits of antioxidants are discussed, and some explanations for the rather disappointing results from these studies are addressed. Finally, important avenues for future research in the field of ROS, oxidative stress, and redox signaling in hypertension are considered.