Monday, November 28, 2016

AMPK activator Metformin found to alleviate accelerated aging defects in Progeria cells: Hypothesis substantiated linking AMPK with aging and HIV-1

"Hutchinson-Gilford Progeria Syndrome" by The Cell Nucleus and Aging: Tantalizing Clues and Hopeful Promises. Scaffidi P, Gordon L, Misteli T; https://commons.wikimedia.org/wiki/File:HIV-budding-Color.jpg#/media/File:HIV-budding-Color.jpg. "HIV-budding-Color" by Photo Credit: C. Goldsmith. Content Providers: CDC/ C. Goldsmith, P. Feorino, E. L. Palmer, W. R. McManus.


AMPK activator Metformin found to alleviate accelerated aging defects in Progeria cells: Hypothesis substantiated linking AMPK with aging and HIV-1


A recent study published online in the Journal npj Aging and Mechanisms of Disease (part of the Nature Partner Journals series) in November of 2016 provided startling evidence that metformin, a widely-prescribed anti-diabetic drug derived from the plant Galega officinalis that has been shown to increase the lifespan and healthspan of several organisms, decreased the expression of progerin (a toxic protein that leads to accelerated cellular aging defects) and alleviated pathological defects in cells derived Hutchinson–Gilford progeria syndrome (HGPS) patients [1].  Interestingly, metformin also decreased the expression of the gene splicing factor SRSF1, a protein that has been previously shown to promote the use of a cryptic splice site in the LMNA gene, increasing the expression of the toxic protein progerin that leads to the accelerated aging phenotype observed in HGPS [1].  This study provides direct support and substantiates a hypothesis published in 2014, in which I proposed for the first time that AMPK activators including metformin will improve accelerated aging defects in HGPS by decreasing the levels of SRSF1, thus reducing progerin production via modulation of alternative splicing [2]. 

Interestingly, several chemically distinct compounds that have recently been shown to improve accelerated cellular aging defects in HGPS, including rapamycin, methylene blue, sulforaphane, all-trans retinoic acid, MG132, oltipraz, and vitamin D have each been shown to activate AMPK, similar to metformin (see below).  Metformin has also recently been shown to beneficially alter gene splicing in cells taken from patients with the genetic disorder myotonic dystrophy type I (DMI) in an AMPK-dependent manner.  Additionally, metformin beneficially altered gene splicing in diabetic patients who were taking metformin but who did not have DM1 [3].  The confirmation of my 2014 hypothesis via the npj Aging and Mechanisms of Disease study that metformin indeed decreases SRSF1 and improves accelerated aging defects in HGPS provides a powerful indication that AMPK activation represents an “indirect yet common mechanism of action” linking the therapeutic effects of chemically distinct compounds in HGPS.  Furthermore, as explained below, SRSF1 has been shown to prevent the reactivation of latent HIV-1 viral reservoirs and many chemically distinct compounds, including MG132, have been shown to promote reactivation of latent HIV-1 in immune cells (facilitating detection and destruction of the virus), implicating the novel proposition that latent HIV-1 reactivation is critically dependent on AMPK activation, a proposal that I published for the first time in 2015 [4]. 

HGPS is a rare genetic disorder caused by the faulty splicing of a gene called the LMNA gene, producing large amounts of a mutant protein known as progerin [5].  Progerin accumulation at a very early age in HGPS patients leads to distortions in the shape of the nucleus and aberrations in mechanisms that occur in the nucleus, leading to characteristic symptoms of accelerating aging such as thinning of the hair, wrinkling of the skin, and eventual cardiovascular disease [5].  Interestingly, normal humans produce the same toxic protein progerin via use of the same cryptic splice site in the LMNA gene as progeria patients, just at much lower levels that increase with age [6].  Recent evidence has also shown that inhibition of the splicing factor SRSF1 leads to a reduction in progerin at both the mRNA and protein levels (thus altering the LMNA pre-mRNA splicing ratio) and SRSF1 activity promotes the faulty splicing of genes involved in the maintenance of the vascular system in normal humans (e.g. VEGF, tissue factor, endoglin), leading to accelerated endothelial cell senescence [4,7,8].

In the npj Aging and Mechanisms of Disease study, Egesipe et al. initially demonstrated, using mesenchymal stem cells (MSCs) derived from HGPS induced pluripotent stem cells (i.e. HGPS MSCs), a significant dose-dependent decrease in SRSF1 mRNA levels after metformin treatment and up to a 40% decrease in SRSF1 protein levels after treatment with 5 mmol/l of metformin [1]. A significant decrease was also observed in both lamin A and progerin mRNA expression in HGPS MSCs treated with 5 mmol/l metformin, with progerin mRNA expression and protein levels reduced to levels lower than that of lamin A mRNA expression and protein levels, indicating that metformin-induced inhibition of SRSF1 led to an increase in the lamin A/progerin ratio and thus beneficially altered gene splicing [1].          

The results obtained using HGPS MSCs were also replicated in additional in vitro cell models, with 5 mmol/l of metformin decreasing progerin mRNA expression up to 50% in LmnaG609G/G609G mouse primary fibroblasts (HGPS mouse model) and decreasing both lamin A and progerin mRNA expression in primary HGPS fibroblasts [1]. Interestingly, 5 mmol/l of metformin also decreased progerin mRNA expression in wild-type/normal MSCs that had been incubated with a compound that induces progerin expression, indicating that metformin may also prove beneficial in reducing progerin levels in normal humans [1].  

Most importantly, however, treatment of HGPS MSCs with 5 mmol/l of metformin reduced the percentage of abnormal nuclei from 60% pre-treatment to less than 40% after treatment (wild-type MSCs presented less than 20% of abnormal nuclei). The metformin-induced reduction in abnormal nuclei was comparable to the reference treatment tipifarnib (1 μmol/1), a farnesyl-transferase inhibitor [1].  Additionally, as HGPS MSCs are characterized by premature osteogenic differentiation (indicated by increased alkaline phosphatase activity compared to wild-type osteogenic progenitor cells), 5 mmol/l of metformin led to a significant rescue of alkaline phosphatase activity in HGPS osteogenic progenitor cells, comparable to levels found in tipifarnib-treated cells [1].  

Again, this study provides compelling evidence and substantiates my hypothesis published in 2014 that proposed for the first time that AMPK activators including metformin will ameliorate accelerated aging defects in cells derived from HGPS patients by decreasing the levels of SRSF1, thus reducing progerin production via modulation of alternative splicing [2]. However, the results from the npj Aging and Mechanisms of Disease study also provides further support for a novel proposal published for the first time in 2015 in which I proposed that a decrease in the splicing activities of SRSF1 by chemically distinct AMPK activators will also lead to the reactivation of latent HIV-1 viral reservoirs [4].  Known as the “shock and kill” approach, this method is an active area among HIV-1 cure researchers and involves reactivating (i.e. “shock”) a T cell (or another immune cell) that harbors dormant HIV-1, hence reactivating the virus itself and thus inducing destruction of the T cell along with the virus or enhancing recognition and destruction of the virus-infected T cell by the immune system (i.e. “kill”) [4]. 

Interestingly, as a preponderance of evidence has convincingly shown that metformin’s primary mechanism of action is via AMPK activation, AMPK is also critical for the activation of T cells and the mounting of an effective immune response to eliminate viruses, bacteria, and cancer cells [9-11].  Strikingly, the same compounds that have been used to induce a “shock” to initiate the creation of human life/oocyte activation (i.e. ionomycin and A23187) have also been used in combination with other compounds as positive controls to initiate a “shock” to facilitate CD4+ T cell activation and thus reactivate dormant HIV-1  [9,12]. Calcium ionophores including ionomycin have also been used to induce a “shock” to activate cytotoxic CD8+ T cells, a T cell subset that is critical for the destruction of viruses such as HIV-1 and cancer cells. Metformin and AMPK activation has also been shown to promote the formation of long-lived cytotoxic CD8+ memory T cells [10,11,13].

Indeed, studies have shown that efficient reactivation of latent HIV-1 involves a reduction in the splicing of the HIV-1 genome by the splicing factor SRSF1, an upregulation in the activity of the splicing-associated protein p32 (an endogenous inhibitor of SRSF1 that is critical for efficient mitochondrial functionality and oxidative phosphorylation), the production of unspliced HIV-1 mRNA (also known as HIV-1 Gag), and the processing of Gag into the HIV-1 p24 antigen, an antigen that is an endpoint that is frequently measured to determine if efficient reactivation of latent HIV-1 by a candidate compound was successful [14,15,16].  Interestingly, the activity of the splicing factor SRSF1 is also downregulated during activation of T cells not infected with HIV-1 [17].

Because metformin, a well-studied AMPK activator, has been shown to reduce the levels of the splicing factor SRSF1 and thus ameliorate aberrant alternative splicing in HGPS cells and because AMPK activation is critical for T cell activation (and thus latent HIV-1 reactivation) and SRSF1 impedes efficient reactivation of latent HIV-1, it would be expected that compounds that both improve accelerated aging defects in HGPS and reactivate latent HIV-1 would also induce AMPK activation.  Indeed, a recent study has demonstrated that metformin, when combined with the protein kinase C modulator bryostatin, induced reactivation of latent HIV-1 in a monocytic cell line in an AMPK-dependent manner.  Bryostatin was also shown to induce phosphorylation and activation of AMPK in that study, implying that bryostatin is an indirect AMPK activator as well [18]. Furthermore, the calcium ionophores ionomycin and A23187, both of which activate AMPK and induce human oocyte activation, are often combined with phorbol 12-myristate 13-acetate (PMA) and are extremely efficient in promoting T cell activation-induced latent HIV-1 reactivation [9,12,19,20].

The compound MG132, a proteasome inhibitor, has also recently been shown in preliminary studies to reduce the levels of the toxic protein progerin via the induction of autophagy and also to reduce progerin production by decreasing the levels of the splicing factor SRSF1, thus beneficially altering splicing of the LMNA gene in HGPS [21,22].  In a separate study, MG132, either alone or in combination with the vitamin A metabolite all-trans retinoic acid, led to a decrease in progerin levels in HGPS cells via the induction of autophagy [23]. MG132 has also been shown to activate AMPK and significantly induce HIV-1 reactivation in two latent HIV-1 primary human CD4+ T cell models that mimic central and effector memory T cells (two memory T cell subsets that are known reservoirs for latent HIV-1) [24,25].

Interestingly, autophagic induction has been shown to be critical for both the removal of the toxic protein progerin in HGPS cells by compounds including MG132 and all-trans retinoic as well as T cell activation. Indeed, autophagy is essential for and upregulated on T cell activation and AMPK activation significantly increases mitochondrial biogenesis, activates ULK1 to induce autophagy, and promotes activation the master antioxidant transcription factor Nrf2 [26-29]. Because the AMPK activators metformin and MG132 have been shown to inhibit SRSF1 and beneficially alter gene splicing in HGPS cells and because AMPK activation is critical for T cell activation, autophagic induction, mitochondrial biogenesis/functionality, and promotes Nrf2 activation, chemically distinct compounds that have been demonstrated to reduce progerin levels and/or ameliorate accelerated aging defects in HGPS cells would be expected to share a common mechanism of AMPK activation.

Indeed, preclinical studies using the macrolide rapamycin in progeria cells indicated that rapamycin corrected cellular aging defects by inducing the degradation of progerin by activating autophagy [30]. Rapamycin was also recently found to potently activate AMPK in vivo in normal old mice as well as induce autophagy and mitochondrial biogenesis [31]. The induction of ULK1-dependent autophagy by rapamycin was also shown to be significantly decreased when the splicing factor p32 (an endogenous inhibitor of SRSF1) was inhibited, indicating that p32 activity is critical for rapamycin-induced autophagy [32].  Because p32 is critical for rapamycin-induced autophagy by ULK1 and because rapamycin, similar to metformin, activates AMPK and AMPK induces autophagy by phosphorylating and activating ULK1, the beneficial effects of rapamycin in progeria likely involves AMPK-mediated alteration of gene splicing as well as AMPK-mediated induction of autophagy.  Both metformin and rapamycin have also been shown to increase the formation of CD8+ memory T cells and rapamycin has been shown to enhance the immune response to viral infections, indicating that rapamycin-induced AMPK activation represents a central node in ameliorating accelerated aging defects in HGPS cells and improving T cell responses to viral pathogens [10,33-35].

Other compounds that have been shown to reduce the levels of progerin and/or improve accelerated aging defects in HGPS cells via autophagic induction, including all-trans retinoic acid and the Nrf2 activator sulforaphane, have also been shown to activate AMPK [36-39].  As AMPK activates PGC-1a, a key transcription factor that promotes mitochondrial functionality/biogenesis and mitochondrial dysfunction characterizes HGPS cells, methylene blue has been shown to correct mitochondrial functioning in HGPS fibroblasts, increase PGC-1a levels, and ameliorate the characteristic nuclear distortion and blebbing observed in HGPS [40,41]. Expectedly, methylene blue has also been shown in independent studies to induce macroautophagy and activate AMPK in vitro and in vivo [42,43].  

Interestingly, AMPK activation has also been shown to phosphorylate and induce nuclear retention of Nrf2, a master regulator of the antioxidant response, thus enhancing Nrf2 activity [28,29].  Strikingly, a recent study demonstrated that the transcriptional activity of Nrf2 is impaired in HGPS patient cells, leading to an increase in chronic oxidative stress.  The reactivation of Nrf2 in HGPS patient cells by the Nrf2 activator oltipraz reversed nuclear aging defects and also restored the in vivo viability of HGPS patient-derived mesenchymal stem cells (MSCs) that were implanted into animal models [44].  Similar to metformin, all-trans retinoic acid, MG132, rapamycin, and methylene blue, oltipraz and/or its metabolites also induce activation of AMPK, increase expression of genes that encode proteins involved in mitochondrial fuel oxidation, increase mitochondria DNA content and oxygen consumption rate, reduce cellular reactive oxygen species (ROS) production, activate LKB1 (an upstream activator of AMPK), and increase the AMP/ATP ratio (an indication of cellular stress induction) [45-49].  Additionally, similar to MG132, which reactivates latent HIV-1 but inhibits active replication of HIV-1, several studies have shown that oltipraz and/or its metabolites inhibit replication of HIV-1, indicating that oltipraz-induced AMPK activation likely also induces immuno-modulatory effects [25,50-52].

Lastly, a recent study demonstrated that 1α,25-dihydroxyvitamin D3 (1,25D), the most potent metabolite of vitamin D, profoundly improved nuclear morphology, significantly reduced DNA damage, improved cellular proliferation, delayed premature cellular senescence, and dramatically reduced progerin production in HGPS patient cells through the promotion of vitamin D receptor (VDR) signaling [53].  Indeed, 1,25D has been shown to activate AMPK in vivo as well as alter gene splicing in cancer cells [54,55].  1,25D also plays a critical role in immune system regulation, as evidenced by an increase in activated CD4+ T cells in HIV-1 patients administered 1,25D in a placebo-controlled randomized study [56].  VDR signaling plays an integral role in T cell activation, with T cell receptor triggering inducing an upregulation of PLC-γ1 (a protein critical for T cell activation) that is dependent on 1,25D and expression of the VDR [4,57].  Interestingly, as PMA (a positive control extensively used in latent HIV-1 reactivation studies) has been demonstrated to enhance 1,25D-induced promoter binding activity of the VDR, Kitano et al. demonstrated that 1,25D, PMA/TPA, and tumor necrosis factor (TNF) stimulated HIV-1 proviral activation to similar levels in a cell line latently-infected with a monocytotropic strain of HIV-1JR-FL [4,58,59].

In conclusion, the results from the npj Aging and Mechanisms of Disease study demonstrating that metformin decreases the expression of both progerin and the splicing factor SRSF1 and alleviates pathological defects in HGPS patient-derived cells provides direct support and substantiates a hypothesis published in 2014 in which I proposed for the first time that AMPK activators including metformin will ameliorate accelerated aging defects in cells derived from HGPS patients by decreasing the levels of SRSF1, thus reducing progerin production via modulation of alternative splicing [2]. Because AMPK activation is critical for T cell activation, increased SRSF1 activity impedes T cell activation and latent HIV-1 reactivation, and the endogenous SRSF1 inhibitor p32 is upregulated on HIV-1 reactivation, the results from the npj Aging and Mechanisms of Disease study also strongly support a hypothesis published in 2015 in which I proposed for the first time that inhibition of SRSF1 by AMPK activators will promote the induction of latent HIV-1 reactivation, facilitating detection and destruction of the virus [4].  Indeed, p32 has been shown to be essential for ULK-1 mediated autophagic induction by rapamycin, a drug that improves immune system responses to viral infections and ameliorates accelerated aging defects in HGPS.  Additionally, the calcium ionophores ionomycin and A23187, both of which have been shown to activate AMPK and are used in a combinatorial fashion as positive controls to reactive latent HIV-1, also induce human oocyte activation, leading to the birth of healthy children. As AMPK activation is also essential for oocyte meiotic resumption, AMPK activation connects amelioration of accelerated aging defects in HGPS not only with latent HIV-1 reactivation, but also with oocyte activation, a process without which there can be no human life.  Moreover, phosphorylated/activated AMPK (pAMPK) has recently been discovered for the first time in human sperm, localized along the tail and across the entire acrosome in the head of the sperm [60].  Because the acrosome reaction is critical for oocyte penetration and fertilization and because compounds that increase intracellular levels of calcium, including vitamin D and A23187, have been shown to induce the acrosome reaction in human sperm and activate AMPK, AMPK activation is likely also essential for the induction of the acrosome reaction in human sperm, a process that is indispensable for the creation of all human life outside of a clinical setting [61,62].  That the symptoms of accelerated aging associated with HGPS, reactivation of latent HIV-1, oocyte activation, and the acrosome reaction in sperm is connected by common pathway, AMPK activation, is no less than astounding. As evidence continues to support and substantiate this connection, a paradigm shift in assessment of disease pathology and the practice of medicine is inevitable.      

  



References:

  1. Egesipe, Blondel, Cicero, et al. Metformin decreases progerin expression and alleviates pathological defects of Hutchinson–Gilford progeria syndrome cells. npj Aging and Mechanisms of Disease 2, Article number: 16026 (2016); http://www.nature.com/articles/npjamd201626?WT.feed_name=subjects_drug-discovery

  1. Finley J. Alteration of splice site selection in the LMNA gene and inhibition of progerin production via AMPK activation. Med Hypotheses. 2014 Nov;83(5):580-7.

  1. Laustriat D, Gide J, Barrault L, et al. In Vitro and In Vivo Modulation of Alternative Splicing by the Biguanide Metformin. Mol Ther Nucleic Acids. 2015 Nov 3;4:e262.

  1. Finley J. Reactivation of latently infected HIV-1 viral reservoirs and correction of aberrant alternative splicing in the LMNA gene via AMPK activation: Common mechanism of action linking HIV-1 latency and Hutchinson-Gilford progeria syndrome. Med Hypotheses. 2015 Sep;85(3):320-32.

  1. Ullrich NJ, Gordon LB. Hutchinson-Gilford progeria syndrome. Handb Clin Neurol. 2015;132:249-64.

  1. McClintock D, Ratner D, Lokuge M, et al. The mutant form of lamin A that causes Hutchinson-Gilford progeria is a biomarker of cellular aging in human skin. PLoS One. 2007 Dec 5;2(12):e1269.

  1. Lopez-Mejia IC, Vautrot V, De Toledo M, et al. A conserved splicing mechanism of the LMNA gene controls premature aging. Hum Mol Genet. 2011 Dec 1;20(23):4540-55.

  1. Blanco FJ, Bernabéu C. The Splicing Factor SRSF1 as a Marker for Endothelial Senescence. Front Physiol. 2012 Mar 28;3:54.

  1. Finley J. Oocyte activation and latent HIV-1 reactivation: AMPK as a common mechanism of action linking the beginnings of life and the potential eradication of HIV-1. Med Hypotheses. 2016 Aug;93:34-47.

  1. Pearce EL, Walsh MC, Cejas PJ, et al. Enhancing CD8 T-cell memory by modulating fatty acid metabolism. Nature. 2009 Jul 2;460(7251):103-7.

  1. Blagih J, Coulombe F, Vincent EE, et al. The energy sensor AMPK regulates T cell metabolic adaptation and effector responses in vivo. Immunity. 2015 Jan 20;42(1):41-54. 

  1. Gómez-Gonzalo M, Carretero M, Rullas J et al. The hepatitis B virus X protein induces HIV-1 replication and transcription in synergy with T-cell activation signals: functional roles of NF-kappaB/NF-AT and SP1-binding sites in the HIV-1 long terminal repeat promoter. J Biol Chem. 2001 Sep 21;276(38):35435-43.

  1. Rao E, Zhang Y, Zhu G, et al. Deficiency of AMPK in CD8+ T cells suppresses their anti-tumor function by inducing protein phosphatase-mediated cell death. Oncotarget. 2015 Apr 10;6(10):7944-58.

  1. Berro R, Kehn K, de la Fuente C, et al. Acetylated Tat regulates human immunodeficiency virus type 1 splicing through its interaction with the splicing regulator p32. J Virol. 2006 Apr;80(7):3189-204.

  1. Spina CA, Anderson J, Archin NM, et al. An in-depth comparison of latent HIV-1 reactivation in multiple cell model systems and resting CD4+ T cells from aviremic patients. PLoS Pathog. 2013;9(12):e1003834.

  1. Hu M, Crawford SA, Henstridge DC, et al. p32 protein levels are integral to mitochondrial and endoplasmic reticulum morphology, cell metabolism and survival. Biochem J. 2013 Aug 1;453(3):381-91.

  1. Moulton VR, Gillooly AR, Tsokos GC. Ubiquitination regulates expression of the serine/arginine-rich splicing factor 1 (SRSF1) in normal and systemic lupus erythematosus (SLE) T cells. J Biol Chem. 2014 Feb 14;289(7):4126-34.

  1. Mehla R, Bivalkar-Mehla S, Zhang R, et al. Bryostatin modulates latent HIV-1 infection via PKC and AMPK signaling but inhibits acute infection in a receptor independent manner. PLoS One. 2010 Jun 16;5(6):e11160. 

  1. Tamás P, Hawley SA, Clarke RG, et al. Regulation of the energy sensor AMP-activated protein kinase by antigen receptor and Ca2+ in T lymphocytes. J Exp Med. 2006 Jul 10;203(7):1665-70.

  1. Fogarty S, Hawley SA, Green KA, Saner N, Mustard KJ, Hardie DG. Calmodulin-dependent protein kinase kinase-beta activates AMPK without forming a stable complex: synergistic effects of Ca2+ and AMP. Biochem J. 2010 Jan 27;426(1):109-18.

  1. Harhouri: Efficient progerin clearance through autophagy induction and SRSF-1 downregulation in Hutchinson-Gilford Progeria Syndrome. Orphanet Journal of Rare Diseases 2015 10(Suppl 2):O9.

  1. http://www.abstractsonline.com/Plan/ViewAbstract.aspx?sKey=8faca3f9-1d38-414c-8cbf-768476a45b61&cKey=fafcb8e0-bbeb-498b-b8f5-cdf7f7686ec6&mKey=cabdedda-497c-457e-8481-34a866ab3681, last accessed November 28, 2016.

  1. Pellegrini C, Columbaro M, Capanni C, et al. All-trans retinoic acid and rapamycin normalize Hutchinson Gilford progeria fibroblast phenotype. Oncotarget. 2015 Oct 6;6(30):29914-28.

  1. Jiang S, Park DW, Gao Y, et al. Participation of proteasome-ubiquitin protein degradation in autophagy and the activation of AMP-activated protein kinase. Cell Signal. 2015 Feb 26. pii: S0898-6568(15)00070-4.

  1. Miller LK, Kobayashi Y, Chen CC, Russnak TA, Ron Y, Dougherty JP. Proteasome inhibitors act as bifunctional antagonists of human immunodeficiency virus type 1 latency and replication. Retrovirology. 2013 Oct 24;10:120.

  1. Hubbard VM, Valdor R, Patel B, Singh R, Cuervo AM, Macian F. Macroautophagy regulates energy metabolism during effector T cell activation. J Immunol. 2010 Dec 15;185(12):7349-57.

  1. Hardie DG. AMP-activated protein kinase: an energy sensor that regulates all aspects of cell function. Genes Dev. 2011 Sep 15;25(18):1895-908.

  1. Mo C, Wang L, Zhang J, et al. The crosstalk between Nrf2 and AMPK signal pathways is important for the anti-inflammatory effect of berberine in LPS-stimulated macrophages and endotoxin-shocked mice. Antioxid Redox Signal. 2014 Feb 1;20(4):574-88.

  1. Joo MS, Kim WD, Lee KY, Kim JH, Koo JH, Kim SG. AMPK facilitates nuclear accumulation of Nrf2 by phosphorylating at serine 550. Mol Cell Biol. 2016 May 9. pii: MCB.00118-16. [Epub ahead of print].

  1. Cao K, Graziotto JJ, Blair CD, et al. Rapamycin reverses cellular phenotypes and enhances mutant protein clearance in Hutchinson-Gilford progeria syndrome cells. Sci Transl Med. 2011 Jun 29;3(89):89ra58.

  1. Chiao YA, Kolwicz SC, Basisty N, et al. Rapamycin transiently induces mitochondrial remodeling to reprogram energy metabolism in old hearts. Aging (Albany NY). 2016 Feb;8(2):314-27.

  1. Jiao H, Su GQ2, Dong W, et al. Chaperone-like protein p32 regulates ULK1 stability and autophagy. Cell Death Differ. 2015 Apr 29.

  1. Araki K, Turner AP, Shaffer VO. mTOR regulates memory CD8 T-cell differentiation. Nature. 2009 Jul 2;460(7251):108-12.

  1. Mannick JB, Del Giudice G, Lattanzi M, et al. mTOR inhibition improves immune function in the elderly. Sci Transl Med. 2014 Dec 24;6(268):268ra179.

  1. Keating R, Hertz T, Wehenkel M, et al. The kinase mTOR modulates the antibody response to provide cross-protective immunity to lethal infection with influenza virus. Nat Immunol. 2013 Dec;14(12):1266-76.

  1. Pellegrini C, Columbaro M, Capanni C, et al. All-trans retinoic acid and rapamycin normalize Hutchinson Gilford progeria fibroblast phenotype. Oncotarget. 2015 Oct 6;6(30):29914-28.

  1. Gabriel D, Roedl D, Gordon LB, Djabali K. Sulforaphane enhances progerin clearance in Hutchinson-Gilford progeria fibroblasts. Aging Cell. 2015 Feb;14(1):78-91.

  1. Kim YM, Kim JH, Park SW, Kim HJ, Chang KC. Retinoic acid inhibits tissue factor and HMGB1 via modulation of AMPK activity in TNF-α activated endothelial cells and LPS-injected mice. Atherosclerosis. 2015 Aug;241(2):615-23.

  1. Lee JH, Jeong JK, Park SY. Sulforaphane-induced autophagy flux prevents prion protein-mediated neurotoxicity through AMPK pathway. Neuroscience. 2014 Oct 10;278:31-9.

  1. Xiong ZM, Choi JY, Wang K, et al. Methylene blue alleviates nuclear and mitochondrial abnormalities in progeria. Aging Cell. 2015 Dec 14. doi: 10.1111/acel.12434.

  1. Salminen A, Kaarniranta K. AMP-activated protein kinase (AMPK) controls the aging process via an integrated signaling network. Ageing Res Rev. 2012 Apr;11(2):230-41.

  1. Atamna H, Atamna W, Al-Eyd G, Shanower G, Dhahbi JM. Combined activation of the energy and cellular-defense pathways may explain the potent anti-senescence activity of methylene blue. Redox Biol. 2015 Dec;6:426-35. doi: 10.1016/j.redox.2015.09.004.

  1. Xie L, Li W, Winters A, Yuan F, Jin K, Yang S. Methylene blue induces macroautophagy through 5' adenosine monophosphate-activated protein kinase pathway to protect neurons from serum deprivation. Front Cell Neurosci. 2013 May 3;7:56.

  1. Kubben N, Zhang W, Wang L, et al. Repression of the Antioxidant NRF2 Pathway in Premature Aging. Cell. 2016 Jun 2;165(6):1361-74. doi: 10.1016/j.cell.2016.05.017.

  1. Kim TH, Eom JS, Lee CG, Yang YM, Lee YS, Kim SG. An active metabolite of oltipraz (M2) increases mitochondrial fuel oxidation and inhibits lipogenesis in the liver by dually activating AMPK. Br J Pharmacol. 2013 Apr;168(7):1647-61. doi: 10.1111/bph.12057.

  1. Bae EJ, Yang YM, Kim JW, Kim SG. Identification of a novel class of dithiolethiones that prevent hepatic insulin resistance via the adenosine monophosphate-activated protein kinase-p70 ribosomal S6 kinase-1 pathway. Hepatology. 2007 Sep;46(3):730-9.

  1. Shin SM, Kim SG. Inhibition of arachidonic acid and iron-induced mitochondrial dysfunction and apoptosis by oltipraz and novel 1,2-dithiole-3-thione congeners. Mol Pharmacol. 2009 Jan;75(1):242-53. doi: 10.1124/mol.108.051128.

  1. Kwon YN, Shin SM, Cho IJ, Kim SG. Oxidized metabolites of oltipraz exert cytoprotective effects against arachidonic acid through AMP-activated protein kinase-dependent cellular antioxidant effect and mitochondrial protection. Drug Metab Dispos. 2009 Jun;37(6):1187-97.

  1. Hwahng SH, Ki SH, Bae EJ, Kim HE, Kim SG. Role of adenosine monophosphate-activated protein kinase-p70 ribosomal S6 kinase-1 pathway in repression of liver X receptor-alpha-dependent lipogenic gene induction and hepatic steatosis by a novel class of dithiolethiones. Hepatology. 2009 Jun;49(6):1913-25.

  1. Prochaska HJ, Fernandes CL, Pantoja RM, Chavan SJ. Inhibition of human immunodeficiency virus type 1 long terminal repeat-driven transcription by an in vivo metabolite of oltipraz: implications for antiretroviral therapy. Biochem Biophys Res Commun. 1996 Apr 25;221(3):548-53.

  1. Prochaska HJ, Bornmann WG, Baron P, Polsky B. Inhibition of human immunodeficiency virus type 1 replication by 7-methyl-6,8-bis(methylthio)pyrrolo[1,2-a]pyrazine, an in vivo metabolite of oltipraz. Mol Pharmacol. 1995 Jul;48(1):15-20.

  1. Prochaska HJ, Chavan SJ, Baron P, Polsky B. Oltipraz, a novel inhibitor of human immunodeficiency virus type 1 (HIV-1) replication. J Cell Biochem Suppl. 1995;22:117-25.

  1. Kreienkamp R, Croke M, Neumann MA, et al. Vitamin D receptor signaling improves Hutchinson-Gilford progeria syndrome cellular phenotypes. Oncotarget. 2016 Apr 27. doi: 10.18632/oncotarget.9065.

  1. Swami S, Krishnan AV, Williams J, et al. Vitamin D mitigates the adverse effects of obesity on breast cancer in mice. Endocr Relat Cancer. 2016 Apr;23(4):251-64.

  1. Cristobo I, Larriba MJ, de los Ríos V, García F, Muñoz A, Casal JI. Proteomic analysis of 1α,25-dihydroxyvitamin D3 action on human colon cancer cells reveals a link to splicing regulation. J Proteomics. 2011 Dec 21;75(2):384-97.

  1. Bang U, Kolte L, Hitz M, et al. Correlation of increases in 1,25-dihydroxyvitamin D during vitamin D therapy with activation of CD4+ T lymphocytes in HIV-1-infected males. HIV Clin Trials. 2012 May-Jun;13(3):162-70.

  1. von Essen MR, Kongsbak M, Schjerling P, Olgaard K, Odum N, Geisler C. Vitamin D controls T cell antigen receptor signaling and activation of human T cells. Nat Immunol. 2010 Apr;11(4):344-9.

  1. Jiang Y, Fleet JC. Effect of phorbol 12-myristate 13-acetate activated signaling pathways on 1α, 25 dihydroxyvitamin D3 regulated human 25-hydroxyvitamin D3 24-hydroxylase gene expression in differentiated Caco-2 cells. J Cell Biochem. 2012 May;113(5):1599-607.

  1. Kitano K, Rivas CI, Baldwin GC, Vera JC, Golde DW. Tumor necrosis factor-dependent production of human immunodeficiency virus 1 in chronically infected HL-60 cells. Blood. 1993 Nov 1;82(9):2742-8.

  1. Calle-Guisado V, de Llera AH, Martin-Hidalgo D, et al. AMP-activated kinase in human spermatozoa: identification, intracellular localization, and key function in the regulation of sperm motility. Asian J Androl. 2016 Sep 27. doi: 10.4103/1008-682X.185848. [Epub ahead of print].

  1. de Lamirande E, Tsai C, Harakat A, Gagnon C. Involvement of reactive oxygen species in human sperm arcosome reaction induced by A23187, lysophosphatidylcholine, and biological fluid ultrafiltrates. J Androl. 1998 Sep-Oct;19(5):585-94.

  1. Blomberg Jensen M, Bjerrum PJ, Jessen TE, et al. Vitamin D is positively associated with sperm motility and increases intracellular calcium in human spermatozoa. Hum Reprod. 2011 Jun;26(6):1307-17.
















Wednesday, August 31, 2016

AMPK links beneficial responses to the “Fever Effect” in Autism with Progeria, HIV-1 latency, & Oocyte activation: The "Shock and Live" approach

Hey Blogger Fam, check out my latest post when you have a minute. The post basically explains why some children with Autism get better temporarily (i.e. improvement in speech, decrease in repetitive behaviors) when they get a fever.  Also known as the “fever effect”, this is basically another rendition of the “shock and live” approach, in which the induction of mild stress to a cell, in this case heat stress, leads to the activation of the master metabolic regulator AMPK and a beneficial cellular response.  The child’s behavior improves because mild heat stress or a “shock” to neurons of the brain causes those brain cells to do what they were designed to do that much better.  The broccoli sprout compound sulforaphane, which activates AMPK, also activates the heat shock response and has shown efficacious results in patients with autism and also improves accelerated aging defects in skin cells taken from progeria patients.  How is this possible? Is the correct functioning of neurons related to slowing or reversing accelerated aging defects? Indeed it is.  Heat stress, which has been shown in independent studies to activate AMPK, is yet an additional term that equates to “what doesn’t kill you makes you stronger”.  Heat stress, hyperthermia, or the activation of heat shock proteins have been shown to promote T cell functionality, wake up dormant HIV-1 viruses so that the immune system can detect and kill it, promote the maturation of oocytes in preparation for artificial oocyte activation or activation by sperm, promote the induction of the acrosome reaction in sperm (a process necessary for oocyte penetration), promote “activation” and killing of cancer stem cells, promote the develop of embryonic stem cells into cells that will eventually make up the body of the baby, and enhance learning and memory in the brain.  The fancy term to describe the process that enhances learning and memory is called long-term potentiation, which basically involves a strengthening in connections between neurons if they are continuously challenged. Heat stress, an intellectually stimulating environment, exercise, and countless naturally-occurring compounds all enhance long-term potentiation, learning and memory, and activate AMPK. Interestingly, high-frequency stimulation and deep brain stimulation also activate AMPK in neurons and improve neurological symptoms, indicating that mild transient electrical pulses are another form of stress.  Lastly, a stark example of how heat stress and learning and memory in neurons are connected to the creation of human life involves a heat-sensitive channel called TRPV3. Mild heat stress and a compound from oregano (called carvacrol) activates this channel.  This channel is also present in the brain and in oocytes.  If you knock this channel out in the brain, long-term potentiation and learning and memory are impaired.  However, if you activate this channel on oocytes with the oregano compound, the oocyte will become activated as if it had been fertilized by sperm (called parthenogenesis).  Again, heat stress and compounds that induce mild stress promotes a compensatory and beneficial response from a cell. This response is orchestrated by AMPK activation and results in a “shock to live” (e.g. oocyte maturation/activation, stem cell “activation”, learning and memory in the brain, sperm capacitation/acrosome reaction) or a “shock to kill” (e.g. latent HIV-1 reactivation, cancer stem cell “activation” and/or cell death).
      
AMPK links beneficial responses to the “Fever Effect” in Autism with Progeria, HIV-1 latency, & Oocyte activation: The "Shock and Live" approach



In line with recent findings demonstrating that sulforaphane, an isothiocyanate heavily concentrated in broccoli sprouts, significantly ameliorated accelerated aging defects in cells derived from patients diagnosed with Hutchinson-Gilford progeria syndrome (HGPS), a recently completed placebo-controlled, double-blind, randomized trial by researchers from Harvard Medical School, The Johns Hopkins University School of Medicine, and University of Massachusetts Medical School demonstrated that participants diagnosed with moderate to severe Autism Spectrum Disorder (ASD) who received sulforaphane showed substantial and significant improvements in behavior, social interaction, and verbal communication compared to participants assigned to placebo [1,2].  Discontinuation of sulforaphane also led to a reversal of these improvements, mirroring levels obtained before initiation of treatment [2].

Interestingly, parents and clinicians, over the past few decades, have reported noticeable improvements in behavior in children with ASD during or after the onset of febrile illness (i.e. fever).  Also known as the “fever effect”, transient increases in body temperature may positively influence neuronal and synaptic function in the brain of ASD patients, an effect that appears to be at least partially mimicked by sulforaphane. As described below, because sulforaphane and other compounds regulate proteins that are critical for the induction of the heat shock response and because heat shock proteins also play critical roles in the reactivation of latent HIV-1, oocyte maturation, sperm capacitation/acrosome reaction, the promotion of learning and memory in the brain, and differentiation of adult stem cells, the “fever effect” likely represents a “shock” or the induction of cellular stress, leading to the activation of the master metabolic regulator AMPK, resulting in a beneficial cellular stress response. The response to this stressor generates a cell-specific “shock to live” (e.g. oocyte maturation/activation, stem cell differentiation, stimulation of learning and memory, sperm capacitation/acrosome reaction) or a “shock to kill” (e.g. latent HIV-1 reactivation, cancer stem cell differentiation and/or cell death).

ASD, a neuro-developmental disorder that disproportionately affects males, is characterized by repetitive or compulsive behaviors, impairments in social development, and deficiencies in verbal and non-verbal communication that are often observable within the first two years of life.  Causes underlying ASD also appear to be multi-factorial, with interactions between environmental factors and genetics leading to deleterious alterations in neuronal network functionality and immune system regulation [3-5].  As noted above, anecdotes and case reports have indicated that amelioration of symptoms associated with ASD are positively correlated with the onset of febrile illness. A particularly interesting example of the “fever effect” in ASD occurred at New York University’s Bellevue Psychiatric Hospital, in which improvement in concentration and social interactions were noted in children with temperatures between 38.9 to 40.6°C, due to an outbreak of viral upper respiratory tract infections [6-8].  Indeed, a prospective study of 30 children with ASD during and after febrile illness (body temperature greater than or equal to 38.0°C) revealed that compared to afebrile ASD patients, fewer aberrant behaviors for febrile ASD patients were recorded on the Aberrant Behavior Checklist subscales of irritability, hyperactivity, stereotypy (i.e. persistent repetition of an act), and inappropriate speech, indicating a transient enhancement or correction of neuronal functioning in response to fever onset [8].

Because sulforaphane has been shown to transiently induce cellular stress, resulting in the upregulation of the master antioxidant transcription factor Nrf2 and activation of AMPK (which activates Nrf2), febrile illness likely induces a transient induction of cellular stress (i.e. “heat shock”), resulting in a compensatory and beneficial upregulation of cellular factors that enhance or promote neuronal functioning [9,10,11]. Indeed, Singh et al. recently demonstrated in a placebo-controlled, double-blind, randomized trial that patients receiving sulforaphane showed significant behavior improvements as measured by the Aberrant Behavior Checklist and the Social Responsiveness Scale compared to patients receiving placebo [2].  Patients who received sulforaphane also experienced improvements in social interaction, abnormal behavior, and verbal communication on the Clinical Global Impression Improvement Scale [2].

Because sulforaphane induces cellular stress via an increase in stressors including reactive oxygen species (ROS) and because febrile illness represents the induction of cellular stress, the beneficial effects of sulforaphane demonstrated in ASD patients suggests that sulforaphane may also induce cellular stress and a beneficial cellular response by activating or upregulating mediators that promote the heat shock response. Heat shock consists of subjecting a cell to a higher temperature than the ideal body temperature of an organism.  The heat shock response is a cellular response to heat shock that induces the activation of heat shock factor-1 (HSF1), the major regulator of heat shock protein (HSP) transcription, and the upregulation of HSPs that aid in repairing or targeting misfolded proteins for degradation [12].  Interestingly, HSPs also respond to other forms of cellular stress, including ROS and intracellular calcium (Ca2+) increases, both of which also activate AMPK [13-15].  Interestingly, sulforaphane has been shown to induce a significant and rapid HSF1 mediated heat shock response and heat stress has also been shown to increase ROS production, promoting nuclear translocation of Nrf2 and upregulation of Nrf2 target genes [16,17]. Furthermore, as sulforaphane activates AMPK and AMPK activates Nrf2, heat stress has also been shown to activate AMPK, promoting insulin-independent glucose transport in muscle cells and the killing of breast and pancreatic cancer stem cells [18,19].  HSP90 has also been to found to interact with and maintain AMPK activity, providing further evidence that the induction of  heat shock likely leads to activation of AMPK and a beneficial cellular response [53].

Interestingly, as sulforaphane has been shown to activate both AMPK and Nrf2 and significantly ameliorate accelerated aging defects associated with Hutchinson-Gilford progeria syndrome (HGPS) (which is associated with dysfunctional Nrf2 signaling), other AMPK-activating compounds that have demonstrated efficacious results in HGPS may also be expected to induce certain mediators constituting the heat shock response [1].  Indeed, the proteasome inhibitor MG132, the macrolide rapamycin, and vitamin D have each been shown to improve symptoms of accelerated aging in HGPS fibroblasts, activate HSF1 or increase HSP expression, and activate AMPK, indicating that chemically distinct compounds including sulforaphane, MG132, rapamycin, and vitamin D likely induce cellular stress, leading to the activation of AMPK and a beneficial cellular response in diseases as disparate as HGPS and ASD [20-28].

As febrile illness and the induction of the heat shock response have been shown to mitigate irritability, hyperactivity, stereotypy, and inappropriate speech in ASD patients, it would be expected that the application of heat stress or the induction of mediators of the heat shock response would also enhance or improve certain aspects of neuronal functionality.  Indeed, heat stress alone and heat stress preconditioning before diffuse axonal injury led to higher expression levels of HSP70 and a significant improvement in the Morris Water Maze task (a behavioral procedure used to study spatial learning and memory) and long-term potentiation (LTP) in rats compared with diffuse axonal injury alone [29]. LTP is characterized by a “persistent increase in synaptic strength following high-frequency stimulation” and is often studied in pyramidal neurons of the hippocampus, an area of the brain important for learning and memory [30].  Interestingly, neuronal depolarization (i.e. activation) has been shown to activate HSF1, heat shock has been shown to improve synaptic integrity and memory consolidation, and HSF1 agonists (e.g. exercise) have been shown to improve cognition in models of dementia [31]. A recent study by Notenboom et al. also showed that prolonged hyperthermia in rats enhanced hippocampal CA1 long-term potentiation and sprouting of mossy fiber collaterals into the dentate gyrus [32].  Perhaps most proactively, however, is a recent study by Brown et al. demonstrating that high-frequency stimulation-induced LTP was attenuated at CA3-CA1 pyramidal cell synapses in hippocampal slices from Trpv1 and Trpv3 knockout (KO) mice [33].

Interestingly, carvacrol, a monoterpenoid phenol found in the essential oil of Origanum vulgare (oregano), is a potent activator of the transient receptor potential cation channel, subfamily V, member 3 (TRPV3), a nonselective cation channel that functions in vasoregulation and temperature sensation and is activated between the temperatures of 22 and 40 degrees Celsius [34,35].  The TRPV3 channel has also been shown to associate with and form heteromeric channels with the Ca2+-permeable TRPV1 channel, another nonselective cation channel that is also regulated by temperature sensation and is activated by both physical and chemical stimuli, including temperatures greater than 43 degrees Celsius as well as the natural compound capsaicin (an active component of chili peppers from the genus Capsicum) [35-37]. As both carvacrol and capsaicin have been shown to activate AMPK and heat stress has been shown to both enhance hippocampal LTP and activate AMPK, AMPK activation likely represents a common mechanism of action explaining the therapeutic actions of several chemically distinct compounds and methodologies in the promotion of synaptic plasticity and LTP [38,39].  Indeed, a recent study demonstrated that high frequency stimulation of hippocampal neurons in the dentate gyrus activates AMPK and induces early-phase long-term potentiation (E-LTP) in vivo in rats [40]. E-LTP was prevented by pharmacological inhibition of AMPK, highlighting a critical role for AMPK in learning and memory [40]. High frequency deep brain stimulation of the lateral habenula (an area of the brain that plays an important role in emotion, motivation, and reward) has also been shown to activate AMPK, facilitating antidepressant actions in an animal model of tricyclic antidepressant resistance [41].  Resveratrol, a phytoalexin found in grapes and red wine, has also been shown to rapidly increase  protein levels and synaptic accumulation of the AMPA receptor and increase the strength of excitatory synaptic transmission in rat primary neurons via AMPK activation [42]. Because AMPA receptors mediate fast excitatory transmission and are essential for the induction of LTP and synaptic plasticity, AMPK activation likely represents a central node underlying the beneficial effects of heat stress and compounds such as sulforaphane in ameliorating neuro-developmental effects associated ASD.

Interestingly, because knockdown of TRPV3 channels (channels that are activated in response to mild temperature elevations and by compounds including carvacrol) attenuates LTP in hippocampal pyramidal neurons and because TRPV3 channels are also located on oocytes, it would be expected that mild and transient heat stress would enhance or promote oocyte maturation and/or activation.  Indeed, AMPK activation has been consistently shown to play a critical role in the induction of oocyte meiotic resumption and maturation in preparation for oocyte activation and heat stress has also been shown to stimulate oocyte meiotic resumption and maturation in an AMPK-dependent manner [43, 44]. Intriguingly, the proteasome inhibitor MG132 and methylene blue have both been shown to activate AMPK, alleviate accelerated aging defects in fibroblasts derived from Hutchinson-Gilford progeria syndrome (HGPS) patients, and stimulate oocyte meiotic resumption, providing compelling evidence that AMPK activation represents a central node linking the therapeutic effects of chemically distinct compounds that facilitate the creation of human life [20,27,45-48].

Furthermore, Carvacho et al. demonstrated that the TRPV3 channel is differentially expressed in mouse oocytes during maturation and reaches peak density and activity at metaphase II, the stage at which oocytes develop the competency to initiate Ca2+ oscillations in response to fertilization [49].  The authors also showed that strontium chloride (SrCl2), a compound that induces parthenogenetic activation of mammalian oocytes and has resulted in successful term pregnancies and the birth of normal children, promotes Ca2+ oscillations and induces oocyte activation via TRPV3-mediated Sr2+ influx, as TRPV3 deletion in oocytes (TrpV3−/−) failed to respond to Sr2+-induced activation [49,50].  As the TRPV3 channel is Ca2+ permeable and SrCl2-induced oocyte activation is thought to occur via mimicking Ca2+ by sensitizing and potentiating IP3 receptors, the authors also showed that application of the TRPV3 agonist carvacrol in heterozygous oocytes (TrpV3+/−) led to a substantial increase in intracellular Ca2+ levels as well as parthenogenesis in both TrpV3+/− and wild-type oocytes (TrpV3+/+) (as measured by pronuclear formation and cleavage to the 2-cell stage), whereas TRPV3-deficient oocytes failed to respond [49]. Because TRPV3 channels are also critical for hippocampal LTP in the brain and because the TRPV3 channel agonist carvacrol activates AMPK and AMPK activation is essential for the induction of hippocampal LTP, activation of AMPK is likely critical for the creation of all human life (via oocyte activation) and for the processes of learning and memory formation [51].

Interestingly, recent studies have also demonstrated that knockout of HSF1 in oocytes leads to depletion of HSP90alpha, delayed meiotic resumption, and defective asymmetrical division [52]. As HSP90 has been shown to interact with and maintain AMPK activity and AMPK activation is essential for efficient T cell activation, heat stress or the induction of mediators of the heat shock response would be expected to promote the reactivation of latent HIV-1 in CD4+ memory T cells via low-level T cell activation, a method known as the “shock and kill” approach [51,53]. T cell activation-induced latent HIV-1 reactivation will likely facilitate destruction of the virus through immune-system detection or via virus-induced destruction of the host cell [51]. Indeed, recent studies have demonstrated that T cell activation at fever temperatures (39.5°C) activates HSF1 and HSF1 is essential for T cell proliferation in vitro [54,55]. Also, knockdown of AMPK and CaMKK2 (an upstream activator of AMPK) has been shown to significantly inhibit HIV-1 replication [56]. Strikingly, Roesch et al. showed that HIV-1 replication was increased 2 to 7 fold by culturing primary CD4+ T lymphocytes at a fever-like temperature (39.5°C) and that hyperthermia enhanced HIV-1 reactivation in a model of latently-infected cells in a HSP90-dependent manner [57].  Several recent studies have also demonstrated that HSP90 promotes HIV-1 reactivation from latency in CD4+ T cells by enhancing the activity of host cell several transcription factors (NF-κB, NFAT, and STAT5) that are critical for both T cell activation and HIV-1 reactivation and replication [58,59].  Particularly compelling is a recent study by Pan et al. showing that HSF1 participates in HIV-1 transcription and is essential for latent HIV-1 reactivation by binding to the HIV 5'-long terminal repeat (LTR) to reactivate viral transcription [60].  Overexpression of HSF1 improved HIV transcription whereas knockout of  HSF1 inhibited HIV transcription [60].  Interestingly, in this same study, resveratrol, MG132, and hemin (an iron-containing porphyrin) were also shown to reactivate latent HIV-1 in a T cell line [60]. Similar to resveratrol and MG132, hemin has also been shown to activate AMPK, again placing AMPK activation as a centerpiece facilitating the reactivation of latent HIV-1, oocyte meiotic induction and activation, and learning and memory formation induced by LTP [61].

Activation of mediators of the heat shock response may also play critical roles in other physiological processes that are essential for the creation and the beginnings of human life.  As noted above, in addition to heat stress, HSPs also respond to other forms of cellular stress, including ROS and intracellular Ca2+ increases, both of which activate AMPK [13-15].  Additionally, HSP90 has been found to localize in the neck, midpiece, and tail regions of human sperm and HSP90 inhibition significantly decreases intracellular Ca2+ concentrations during capacitation, a process that is essential for oocyte fertilization [62].  Indeed, an increase in the levels of intracellular Ca2+ is critical for the initiation of the acrosome reaction in sperm, a process that facilitates sperm penetration of the oocyte and is thus indispensable for fertilization.  Interestingly, ROS and vitamin D, both of which activate AMPK, have each been shown to induce the acrosome reaction in sperm, indicating that induction of cellular stress is critical for the promotion of the acrosome reaction in sperm, facilitating the creation of human life [15,28,63,64].  Vitamin D has also been shown to improve accelerated cellular aging defects in progeria, indicating that the creation of human life and the amelioration of symptoms of accelerated aging may depend on the induction of a cellular stress response [22].  Heat stress has also been shown to promote the differentiation of human adult stem cells and AMPK activation has also recently been shown to be critical in embryonic development by facilitating the differentiation of mouse embryonic stem cells into endoderm, again indicating that AMPK activation is critical, if not indispensable, for the creation and beginnings of life [65,66,67].

In conclusion, the evidence presented above strongly supports the provocative implication that the induction of cellular stress, mediated by heat, ROS, intracellular Ca2+ increases, an AMP/ATP ratio increase,  etc. leads to a beneficial compensatory cellular response.  The induction of this cellular response and subsequent activation of AMPK by chemically distinct compounds including sulforaphane, MG132, vitamin D, rapamycin, methylene blue, carvacrol, and likely many others indicates that cellular stress-induced AMPK activation may represent a central node in the amelioration of disease and the creation of human life. Indeed, cell and context-specific AMPK activation may induce a “shock to live” (e.g. oocyte maturation/activation, stem cell differentiation, learning and memory in the brain, sperm capacitation/acrosome reaction) or a “shock to kill” (e.g. latent HIV-1 reactivation, cancer stem cell differentiation and/or cell death).

 References:
1.     Gabriel D, Roedl D, Gordon LB, Djabali K. Sulforaphane enhances progerin clearance in Hutchinson-Gilford progeria fibroblasts. Aging Cell. 2015 Feb;14(1):78-91.
2.     Singh K, Connors SL, Macklin EA, et al. Sulforaphane treatment of autism spectrum disorder (ASD). Proc Natl Acad Sci U S A. 2014 Oct 28;111(43):15550-5.
3.     Myers SM, Johnson CP; American Academy of Pediatrics Council on Children With Disabilities. Management of children with autism spectrum disorders. Pediatrics. 2007 Nov;120(5):1162-82.
4.     Levy SE, Mandell DS, Schultz RT. Autism. Lancet. 2009 Nov 7;374(9701):1627-38.
5.     Hsiao EY. Immune dysregulation in autism spectrum disorder. Int Rev Neurobiol. 2013;113:269-302.
6.     Sullivan RC. Why do autistic children . . . ? J Autism Dev Disord.1980;10:231–241.
7.     Cotterill RM. Fever in autistics. Nature. 1985;313:426.
8.     Curran LK, Newschaffer CJ, Lee LC, Crawford SO, Johnston MV, Zimmerman AW. Behaviors associated with fever in children with autism spectrum disorders. Pediatrics. 2007 Dec;120(6):e1386-92.
9.     Lee YJ, Lee SH. Sulforaphane induces antioxidative and antiproliferative responses by generating reactive oxygen species in human bronchial epithelial BEAS-2B cells. J Korean Med Sci. 2011 Nov;26(11):1474-82.
10. Lee JH, Jeong JK, Park SY. Sulforaphane-induced autophagy flux prevents prion protein-mediated neurotoxicity through AMPK pathway. Neuroscience. 2014 Oct 10;278:31-9.
11. Mo C, Wang L, Zhang J, et al. The crosstalk between Nrf2 and AMPK signal pathways is important for the anti-inflammatory effect of berberine in LPS-stimulated macrophages and endotoxin-shocked mice. Antioxid Redox Signal. 2014 Feb 1;20(4):574-88.
12. Richter K, Haslbeck M, Buchner J. The heat shock response: life on the verge of death. Mol Cell. 2010 Oct 22;40(2):253-66.
13. Madamanchi NR, Li S, Patterson C, Runge MS. Reactive oxygen species regulate heat-shock protein 70 via the JAK/STAT pathway. Arterioscler Thromb Vasc Biol. 2001 Mar;21(3):321-6.
14. Wakita H, Tokura Y, Furukawa F, Takigawa M. High calcium induces heat shock proteins 72 and 60 in cultured human keratinocytes: comparative study with heat shock and sunlamp light irradiation. J Dermatol Sci. 1994 Oct;8(2):136-44.
15. Sundararaman A, Amirtham U, Rangarajan A. Calcium-Oxidant Signaling Network Regulates AMP-activated Protein Kinase (AMPK) Activation upon Matrix Deprivation. J Biol Chem. 2016 Jul 8;291(28):14410-29.
16. Gan N, Wu YC, Brunet M, et al. Sulforaphane activates heat shock response and enhances proteasome activity through up-regulation of Hsp27. J Biol Chem. 2010 Nov 12;285(46):35528-36.
17. Chang SW, Lee SI, Bae WJ, et al. Heat stress activates interleukin-8 and the antioxidant system via Nrf2 pathways in human dental pulp cells. J Endod. 2009 Sep;35(9):1222-8.
18. Goto A, Egawa T, Sakon I, et al. Heat stress acutely activates insulin-independent glucose transport and 5'-AMP-activated protein kinase prior to an increase in HSP72 protein in rat skeletal muscle. Physiol Rep. 2015 Nov;3(11). pii: e12601.
19. Lee H, Park HJ, Park CS, et al. Response of breast cancer cells and cancer stem cells to metformin and hyperthermia alone or combined. PLoS One. 2014 Feb 5;9(2):e87979.
20. Pellegrini C, Columbaro M, Capanni C, et al. All-trans retinoic acid and rapamycin normalize Hutchinson Gilford progeria fibroblast phenotype. Oncotarget. 2015 Oct 6;6(30):29914-28.
21. Cao K, Graziotto JJ, Blair CD, et al. Rapamycin reverses cellular phenotypes and enhances mutant protein clearance in Hutchinson-Gilford progeria syndrome cells. Sci Transl Med. 2011 Jun 29;3(89):89ra58.
22. Kreienkamp R, Croke M, Neumann MA, et al. Vitamin D receptor signaling improves Hutchinson-Gilford progeria syndrome cellular phenotypes. Oncotarget. 2016 Apr 27. doi: 10.18632/oncotarget.9065.
23. Pierce A, Podlutskaya N, Halloran JJ, et al. Over-expression of heat shock factor 1 phenocopies the effect of chronic inhibition of TOR by rapamycin and is sufficient to ameliorate Alzheimer's-like deficits in mice modeling the disease. J Neurochem. 2013 Mar;124(6):880-93.
24. Du ZX, Zhang HY, Meng X, et al. Proteasome inhibitor MG132 induces BAG3 expression through activation of heat shock factor 1. J Cell Physiol. 2009 Mar;218(3):631-7.
25. Lee JH, Park S, Cheon S, et al. 1,25-Dihydroxyvitamin D₃ enhances NK susceptibility of human melanoma cells via Hsp60-mediated FAS expression. Eur J Immunol. 2011 Oct;41(10):2937-46.
26. Chiao YA, Kolwicz SC, Basisty N, et al. Rapamycin transiently induces mitochondrial remodeling to reprogram energy metabolism in old hearts. Aging (Albany NY). 2016 Feb;8(2):314-27.
27. Jiang S, Park DW, Gao Y, et al. Participation of proteasome-ubiquitin protein degradation in autophagy and the activation of AMP-activated protein kinase. Cell Signal. 2015 Jun;27(6):1186-97.
28. Swami S, Krishnan AV, Williams J, et al.. Vitamin D mitigates the adverse effects of obesity on breast cancer in mice. Endocr Relat Cancer. 2016 Apr;23(4):251-64.
29. Su Z, Han D, Sun B, et al. Heat stress preconditioning improves cognitive outcome after diffuse axonal injury in rats. J Neurotrauma. 2009 Oct;26(10):1695-706.
30. Paradiso, Michael A.; Bear, Mark F.; Connors, Barry W. (2007). Neuroscience: Exploring the Brain. Hagerstown, MD: Lippincott Williams & Wilkins.
31. Hooper PL, Durham HD, Török Z, Hooper PL, Crul T, Vígh L. The central role of heat shock factor 1 in synaptic fidelity and memory consolidation. 2016 Jun 9. [Epub ahead of print].
32. Notenboom RG, Ramakers GM, Kamal A, Spruijt BM, de Graan PN. Long-lasting modulation of synaptic plasticity in rat hippocampus after early-life complex febrile seizures. Eur J Neurosci. 2010 Sep;32(5):749-58.
33. Brown TE, Chirila AM, Schrank BR, Kauer JA. Loss of interneuron LTD and attenuated pyramidal cell LTP in Trpv1 and Trpv3 KO mice. Hippocampus. 2013 Aug;23(8):662-71.
34. Xu H, Ramsey IS, Kotecha SA, et al. TRPV3 is a calcium-permeable temperature-sensitive cation channel. Nature. 2002 Jul 11;418(6894):181-6.
35. Xu H, Delling M, Jun JC, Clapham DE. Oregano, thyme and clove-derived flavors and skin sensitizers activate specific TRP channels. Nat Neurosci. 2006 May;9(5):628-35.
36. Cheng W, Yang F, Liu S, et al. Heteromeric heat-sensitive transient receptor potential channels exhibit distinct temperature and chemical response. J Biol Chem. 2012 Mar 2;287(10):7279-88.
37. Tominaga M, Tominaga T. Structure and function of TRPV1. Pflugers Arch. 2005 Oct;451(1):143-50.
38.  Kim E, Choi Y, Jang J, Park T. Carvacrol Protects against Hepatic Steatosis in Mice Fed a High-Fat Diet by Enhancing SIRT1-AMPK Signaling. Evid Based Complement Alternat Med. 2013;2013:290104.
39. Lee GR, Jang SH, Kim CJ, et al. Capsaicin suppresses the migration of cholangiocarcinoma cells by down-regulating matrix metalloproteinase-9 expression via the AMPK-NF-κB signaling pathway. Clin Exp Metastasis. 2014 Dec;31(8):897-907.
40. Yu DF, Shen ZC, Wu PF, et al. HFS-Triggered AMPK Activation Phosphorylates GSK3β and Induces E-LTP in Rat Hippocampus In Vivo. CNS Neurosci Ther. 2016 Jun;22(6):525-31.
41. Kim Y, Morath B, Hu C, et al. Antidepressant actions of lateral habenula deep brain stimulation differentially correlate with CaMKII/GSK3/AMPK signaling locally and in the infralimbic cortex. Behav Brain Res. 2016 Jun 1;306:170-7.
42. Wang G, Amato S, Gilbert J, Man HY. Resveratrol up-regulates AMPA receptor expression via AMP-activated protein kinase-mediated protein translation. Neuropharmacology. 2015 Aug;95:144-53.
43. Chen J, Hudson E, Chi MM, et al. AMPK regulation of mouse oocyte meiotic resumption in vitro. Dev Biol. 2006 Mar 15;291(2):227-38.
44. LaRosa C, Downs SM. Meiotic induction by heat stress in mouse oocytes: involvement of AMP-activated protein kinase and MAPK family members. Biol Reprod. 2007 Mar;76(3):476-86.
45. Shin SY, Kim TH, Wu H, Choi YH, Kim SG. SIRT1 activation by methylene blue, a repurposed drug, leads to AMPK-mediated inhibition of steatosis and steatohepatitis. Eur J Pharmacol. 2014 Mar 15;727:115-24.
46. Xiong ZM, Choi JY, Wang K, et al. Methylene blue alleviates nuclear and mitochondrial abnormalities in progeria. Aging Cell. 2016 Apr;15(2):279-90.
47. Huo LJ, Fan HY, Zhong ZS, Chen DY, Schatten H, Sun QY. Ubiquitin-proteasome pathway modulates mouse oocyte meiotic maturation and fertilization via regulation of MAPK cascade and cyclin B1 degradation. Mech Dev. 2004 Oct;121(10):1275-87.
48. Downs SM, Humpherson PG, Leese HJ. Meiotic induction in cumulus cell-enclosed mouse oocytes: involvement of the pentose phosphate pathway. Biol Reprod. 1998 Apr;58(4):1084-94.
49. Carvacho I, Lee HC, Fissore RA, Clapham DE. TRPV3 channels mediate strontium-induced mouse-egg activation. Cell Rep. 2013 Dec 12;5(5):1375-86.
50. Kyono K, Kumagai S, Nishinaka C, et al. Birth and follow-up of babies born following ICSI using SrCl2 oocyte activation. Reprod Biomed Online. 2008 Jul;17(1):53-8.
51. Finley J. Oocyte activation and latent HIV-1 reactivation: AMPK as a common mechanism of action linking the beginnings of life and the potential eradication of HIV-1. Med Hypotheses. 2016 Aug;93:34-47.
52. Metchat A, Akerfelt M, Bierkamp C, et al. Mammalian heat shock factor 1 is essential for oocyte meiosis and directly regulates Hsp90alpha expression. J Biol Chem. 2009 Apr 3;284(14):9521-8.
53. Zhang L, Yi Y, Guo Q, et al. Hsp90 interacts with AMPK and mediates acetyl-CoA carboxylase phosphorylation. Cell Signal. 2012 Apr;24(4):859-65.
54. Gandhapudi SK, Murapa P, Threlkeld ZD, et al. Heat shock transcription factor 1 is activated as a consequence of lymphocyte activation and regulates a major proteostasis network in T cells critical for cell division during stress. J Immunol. 2013 Oct 15;191(8):4068-79.
55. Murapa P, Gandhapudi S, Skaggs HS, Sarge KD, Woodward JG. Physiological fever temperature induces a protective stress response in T lymphocytes mediated by heat shock factor-1 (HSF1). J Immunol. 2007 Dec 15;179(12):8305-12.
56. Zhou H, Xu M, Huang Q, et al. Genome-scale RNAi screen for host factors required for HIV replication. Cell Host Microbe. 2008 Nov 13;4(5):495-504.
57. Roesch F, Meziane O, Kula A, et al. Hyperthermia stimulates HIV-1 replication. PLoS Pathog. 2012;8(7):e1002792.
58. Anderson I, Low JS, Weston S, et al. Heat shock protein 90 controls HIV-1 reactivation from latency. Proc Natl Acad Sci U S A. 2014 Apr 15;111(15):E1528-37.
59. Joshi P, Maidji E, Stoddart CA. Inhibition of Heat Shock Protein 90 Prevents HIV Rebound. J Biol Chem. 2016 May 6;291(19):10332-46.
60. Pan XY, Zhao W, Zeng XY, et al. Heat Shock Factor 1 Mediates Latent HIV Reactivation. Sci Rep. 2016 May 18;6:26294.
61. Ju TJ, Kwon WY, Kim YW, et al. Hemin improves insulin sensitivity in skeletal muscle in high fat-fed mice. J Pharmacol Sci. 2014;126(2):115-25.
62. Li K, Xue Y, Chen A, et al. Heat shock protein 90 has roles in intracellular calcium homeostasis, protein tyrosine phosphorylation regulation, and progesterone-responsive sperm function in human sperm. PLoS One. 2014 Dec 26;9(12):e115841.
63. de Lamirande E, Tsai C, Harakat A, Gagnon C. Involvement of reactive oxygen species in human sperm arcosome reaction induced by A23187, lysophosphatidylcholine, and biological fluid ultrafiltrates. J Androl. 1998 Sep-Oct;19(5):585-94.
64. Blomberg Jensen M, Bjerrum PJ, Jessen TE, et al. Vitamin D is positively associated with sperm motility and increases intracellular calcium in human spermatozoa. Hum Reprod. 2011 Jun;26(6):1307-17.
65. Choudhery MS, Badowski M, Muise A, Harris DT. Effect of mild heat stress on the proliferative and differentiative ability of human mesenchymal stromal cells. Cytotherapy. 2015 Apr;17(4):359-68.
66. Nørgaard R, Kassem M, Rattan SI. Heat shock-induced enhancement of osteoblastic differentiation of hTERT-immortalized mesenchymal stem cells. Ann N Y Acad Sci. 2006 May;1067:443-7.
67. Young NP, Kamireddy A, Van Nostrand JL, et al. AMPK governs lineage specification through Tfeb-dependent regulation of lysosomes. Genes Dev. 2016 Mar 1;30(5):535-52.

Friday, July 22, 2016

AMPK links cognitive decline reversal in Alzheimer’s disease with Progeria, HIV-1 latency, & Oocyte activation: The "Shock and Live" approach

Hey Blogger Fam, take a look at my most recent post on LinkedIn when you have a minute.  Basically, a recent human study showed that Alzheimer's disease is likely reversible, contradicting what's commonly accepted in the medical profession (i.e. that it's irreversible). The super interesting thing about this study was that the protocol that the patients adhered to is just a contextual redefinition of the “Shock and Live” approach, consisting of changes in diet, exercise, and vitamin and herbal supplementation that synergistically causes the induction of cellular stress (i.e. "shock"), leading to a beneficial response in brain cells (i.e. "live”) mediated by the activation of the master metabolic regulator AMPK.  Indeed, most of the components of the protocol, including a low carb/sugar diet, fasting, caloric restriction, exercise, EPA/DHA (fish oil), melatonin, folic acid, selenium, alpha-lipoic acid, acetyl-l-carnitine, resveratrol, curcumin, CoQ10, probiotics, and prebiotics all induce a cellular stress response, activating AMPK and bringing “life” back into cells of the brain. AMPK activation has been shown to promote neurogenesis (birth of new brain cells), increase the growth of new neurons and synapses, promote the viability of neural stem cells (which produce new brain cells), and promote learning and memory formation.

Two things that really caught my eye were the use of brain stimulation and vitamin D.  Although the patients used learning-based simulations to stimulate the brain, electrical pulses/stimulation (“shock”) have been shown to activate AMPK and produce beneficial effects in many people with neurological disorders (Parkinson’s, depression, tremor, obsessive-compulsive disorder).  The cool connection is that electrical pulses have also been shown to activate oocytes, leading to the birth of healthy children.  Thus, AMPK activation via a “shock” likely explains how invasive procedures like deep-brain stimulation produce efficacious results as well as how all human life is created.  The other REALLY cool thing about the study is vitamin D.  Not only does vitamin D promote the activation of T cells (enhances the immune response), but it also induces a slight “shock” (in the form of an increase in calcium), bringing back to life (i.e. “live”) cells taken from Progeria kids that are rapidly aging and nearing death. This slight “shock” (calcium) is also what activates oocytes, bringing forth new life.  Interestingly, vitamin D has also been shown to induce the acrosome reaction in human sperm, a process that allows sperm to penetrate the egg, and without which no human being could be created via “the old fashion way”:-).  That “shock”, in the form of calcium, also ignites the acrosome reaction in sperm.  Unsurprisingly, vitamin D as well as compounds that increase calcium (“shock”) and have been used to activate both human oocytes and induce the acrosome reaction in human sperm activate AMPK.  And here’s a little extra nugget from something I’m working on.  AMPK activation is critical during a  “shock” to embryonic stem cells, forcing those cells to develop into different cells (liver, pancreas, gut, urethra, etc.) that will eventually form the baby, another rendition of the “Shock and Live” approach.  Let’s see….a “shock” induces oocyte activation, the sperm acrosome reaction, neural stem cell viability, progeria cell growth, and development of a baby from an embryo.  It looks as if the “Shock and Live” approach may be a universal theme in human biology.  If this turns out to be true…………..:-)


AMPK links cognitive decline reversal in Alzheimer’s disease with Progeria, HIV-1 latency, & Oocyte activation: The "Shock and Live" approach  


https://www.linkedin.com/pulse/ampk-links-cognitive-decline-reversal-alzheimers-disease-finley?trk=prof-post 

In a small but highly publicized study conducted by researchers from the University of California, Los Angeles and the Buck Institute for Research on Aging, the use of a personalized therapeutic program to improve or reverse cognitive decline in 10 patients with early Alzheimer’s disease led to a clear improvement in mild and subjective cognitive impairment (MCI and SCI, respectively--Alzheimer’s precursors) for all ten patients, with a dramatic and unprecedented reversal of cognitive decline for two patients with well documented Alzheimer’s disease [32].  The therapeutic approach, known as metabolic enhancement for neurodegeneration (MEND), is a multi-pronged protocol designed to promote metabolic enhancement that consists of dietary modifications, exercise, vitamin and herbal supplementation, stress reduction incorporation, exercise, and hormonal balancing [33].       

After 10 months on the program, patient 1 (a 66-year-old professional male with documented Alzheimer’s disease, a strongly positive amyloid PET scan, and abnormal neuropsychological studies) experienced an increase in hippocampal volume from the 17th to the 75th percentile and an absolute increase in hippocampal volume of 11.7% as measured via volumetric analyses by Neuroquant [32].  Interestingly, interruption of compliance to the protocol led to an episode of memory loss for the patient (failed to remember that he left the car in the driveway idling).  However, re-adherence to the protocol led to marked symptomatic improvement (memory improvement and “work came more easily to him”) in addition to a significant increase in hippocampal volume [32].

Patient 2, a 69-year-old professional male and entrepreneur, also had well-documented Alzheimer’s disease, with an FDG PET scan showing reduced glucose utilization in the parietotemporal cortices and the temporal lobes, a reduction in CVLT (California Verbal Learning Test), auditory delayed memory at the 13th percentile, and a Stroop color test at the 16th percentile. The patient also reported subjective instances of memory loss (difficulty recognizing faces at work) and declines in cognitive capacity (loss the ability to rapidly add columns of numbers in his head) [32]. After 6 months on the MEND program, the patient’s ability to recognize faces at work and to rapidly add columns of numbers in his head returned. Strikingly, quantitative neuropsychological testing after 22 months on the program revealed an increase from the 3rd to the 84th percentile for CVLT-IIB, reverse digit span from the 24th to the 74th percentile, auditory delayed memory from the 13th to the 79th percentile, and CVLT-II from the 54th to the 96th percentile [32].

Interestingly, the authors also noted that the improvement experienced by all 10 patients had been sustained and no patient exhibited the cognitive decline characteristic of Alzheimer’s disease, despite claims that Alzheimer’s disease can not be prevented, delayed, or reversed [32,34].

The details of the MEND protocol (reproduced and adapted below) clearly indicate that a significant number of disparate modalities and compounds that synergistically modulated metabolism, leading to a striking reversal of cognitive decline in patients diagnosed with early Alzheimer’s disease, are in fact inducers of a cellular stress response. The induction of cellular stress leads to the activation of the master metabolic regulator AMPK (see red notations for references).  Synergistic AMPK activation by this protocol is essentially a contextual redefinition of the “Shock and Live” approach that defines the process of oocyte activation, a mechanism that is responsible for the creation of all human life.

(Adapted from: Bredesen DE. Reversal of cognitive decline: a novel therapeutic program. Aging (Albany NY). 2014 Sep;6(9):707-17.

Therapeutic System 1.0
Goal
Approach
Rationale and References
Optimize diet: minimize simple CHO, minimize inflammation.
Patients given choice of several low glycemic, low inflammatory, low grain diets.  Glucose deprivation activates AMPK [1,2].
Minimize inflammation, minimize insulin resistance.
Enhance autophagy, ketogenesis
Fast 12 hr each night, including 3 hr prior to bedtime. Fasting and caloric restriction activates AMPK.  AMPK stimulates brain ketogenesis and authophagy [3-6].
Reduce insulin levels, reduce Aβ.
Reduce stress
Personalized—yoga or meditation or music, etc.  AMPK reduces cortiol levels [7].
Reduction of cortisol, CRF, stress axis.
Optimize sleep
8 hr sleep per night; melatonin 0.5mg po qhs; Trp 500mg po 3x/wk if awakening. Exclude sleep apnea.  Melatonin activates AMPK [8].
[36]
Exercise
30-60′ per day, 4-6 days/wk.  Aerobic exercise activates AMPK [9].
[37, 38]
Brain stimulation
Posit or related.  Brain stimulation via high frequency stimulation (HFS) activates AMPK [10].
[39]
Homocysteine <7
Me-B12, MTHF, P5P; TMG if necessary.  MTHF (Folic acid) activates AMPK [11].
[40]
Serum B12 >500
Me-B12
[41]
CRP <1.0; A/G >1.5
Anti-inflammatory diet; curcumin; DHA/EPA; optimize hygiene. EPA, DHA, and curcumin activate AMPK [12-14].
Critical role of inflammation in AD
Fasting insulin <7; HgbA1c <5.5
Diet as above
Type II diabetes-AD relationship
Hormone balance
Optimize fT3, fT4, E2, T, progesterone, pregnenolone, cortisol. fT3, E2, and T activate AMPK [15-17].
[5, 42]
GI health
Repair if needed; prebiotics and probiotics. Prebiotics and probiotics activate AMPK [18,19].
Avoid inflammation, autoimmunity
Reduction of A-beta
Curcumin, Ashwagandha.  Curcumin activates AMPK.  Ashwagandha likely activates AMPK [12,20].
Cognitive enhancement
Bacopa monniera, MgT.  MgT likely activates AMPK [21].
[46, 47]
25OH-D3 = 50-100ng/ml
Vitamins D3, K2.  Vitamin D3 activates AMPK [22].
[48]
Increase NGF
H. erinaceus or ALCAR.  ALCAR activates AMPK [23].
[49, 50]
Provide synaptic structural components
Citicoline, DHA. DHA activates AMPK [14].
[51].
Optimize antioxidants
Mixed tocopherols and tocotrienols, Se, blueberries, NAC, ascorbate, α-lipoic acid.  Tocotrienols, Se, blueberries, and α-lipoic acid activate AMPK [24-27]. 
[52]
Optimize Zn:fCu ratio
Depends on values obtained
[53]
Ensure nocturnal oxygenation
Exclude or treat sleep apnea
[54]
Optimize mitochondrial function
CoQ or ubiquinol, α-lipoic acid, PQQ, NAC, ALCAR, Se, Zn, resveratrol, ascorbate, thiamine.  CoQ10, α-lipoic acid, ALCAR, Se, and resvertrol activate AMPK.  PQQ likely activates AMPK [23,25-30].
[55]
Increase focus
Pantothenic acid
Acetylcholine synthesis requirement
Increase SirT1 function
Resveratrol. Resveratrol activates AMPK [26,29].
[32]
Exclude heavy metal toxicity
Evaluate Hg, Pb, Cd; chelate if indicated
CNS effects of heavy metals
MCT effects
Coconut oil or Axona.  MCT/Coconut oil likely activates AMPK [31].
[56]

CHO, carbohydrates; Hg, mercury; Pb, lead; Cd, cadmium; MCT, medium chain triglycerides; PQQ, polyquinoline quinone; NAC, N-acetyl cysteine; CoQ, coenzyme Q; ALCAR, acetyl-L-carnitine; DHA, docosahexaenoic acid; MgT, magnesium threonate; fT3, free triiodothyronine; fT4, free thyroxine; E2, estradiol; T, testosterone; Me-B12, methylcobalamin; MTHF, methyltetrahydrofolate; P5P, pyridoxal-5-phosphate; TMG, trimethylglycine; Trp, tryptophan


As initially proposed in my most recent publication, AMPK activation is likely critical, if not indispensable, for oocyte activation via the induction of a cellular stress response (mediated by an increase in intracellular calcium levels, an AMP/ATP ratio increase, or reactive oxygen species (ROS) generation) [35].  AMPK has been shown to be critical for the resumption and maturation of meiosis in oocytes, two processes that are critical for efficient sperm- or artificially-induced oocyte activation [35].  Interestingly, several distinct compounds and modalities that induce a cellular stress response have been used to artificially activate oocytes during in vitro fertilization procedures to produce healthy children, including electrical stimulation and the calcium (Ca2+) ionophores ionomycin and A23187 (increases intracellular Ca2+ levels similar to sperm-induced oocyte activation) [35-37]. 

The “Shock and Live” approach is also analogous to the “Shock and Kill” approach, a method currently being pursued by HIV-1 cure researchers to potentially eradicate HIV-1 [35].  The “shock and kill” approach involves reactivating (i.e. “shock”) a T cell that harbors dormant HIV-1, hence reactivating the virus itself and thus inducing destruction of the T cell along with the virus or enhancing recognition and destruction of the virus-infected T cell by the immune system (i.e. “kill”) [35].  AMPK is also critical for the activation of T cells and the mounting of an effective immune response to eliminate viruses, bacteria, and cancer cells [35,38,39].  Interestingly, the same compounds that have been used to induce a “shock” to initiate the creation of human life (i.e. ionomycin and A23187) have also been used in combination with other compounds to initiate a “shock” to facilitate T cell activation and thus reactivate dormant HIV-1  [35,40]. Ca2+ ionophores including ionomycin have also been used to induce a “shock” to activate cytotoxic CD8+ T cells, a T cell subset that is critical for the destruction of viruses such as HIV-1 and cancer cells. AMPK activation has also been shown to promote the formation of long-lived cytotoxic CD8+ memory T cells [38,39,41].

Unsurprisingly, AMPK activation has been shown to promote neurogenesis, increase the levels of BDNF (a protein essential for the growth and differentiation of new neurons and synapses), promote viability of neural stem cells, and enhance intracellular mechanisms that promote learning and memory formation [42-44].  As AMPK is activated as a result of the induction of cellular stress, many of the components of the MEND protocol likely activate AMPK by inducing a “shock”, thus enhancing and sustaining the viability of higher level brain functioning (i.e. “live”).  Indeed, as evident by the chart above and the associated references, glucose deprivation, fasting, caloric restriction, and exercise all induce varying levels of “shock” (mediated by increased levels of ROS, intracellular Ca2+, or AMP/ATP ratio), leading to the activation of AMPK and AMPK-induced autophagy (“live”).  Several naturally-occurring compounds, including EPA, DHA, melatonin, folic acid, selenium, alpha-lipoic acid, and acetyl-l-carnitine also activate AMPK, likely via the induction of a cellular stress response.  Additionally, as also noted from the chart, brain stimulation via electrical pulses also leads to the activation of AMPK (and long-term potentiation—a process that underlies learning and memory), similar to oocyte activation induced by electrical stimulation.   

Interestingly, resveratrol, a polyphenol produced by several plants that activates AMPK, has been shown to not only promote long-term potentiation but also enhance reactivation of latent HIV-1 while an analog of resveratrol has been shown to promote meiotic resumption in mouse oocytes (a model for human oocytes), indicating that an appropriate “shock” or cellular stress induction characterizes beneficial physiological responses in oocytes, T cells, and brain cells [45-47]. Curcumin (a compound derived from the plant Curcuma longa) and butyrate (a short chain fatty acid produced by certain gut bacteria/probiotics) have also been shown to increase neural stem cell differentiation and enhance latent HIV-1 reactivation [48-51].

A particularly striking example of an AMPK-orchestrated “Shock and Live” approach is exemplified by 1α,25-dihydroxyvitamin D3 (vitamin D).  In addition to activating AMPK in vivo, vitamin D has recently been shown to profoundly improve nuclear morphology, significantly reduce DNA damage, improve cellular proliferation, delay premature cellular senescence, and dramatically reduce progerin production in patients diagnosed with the accelerated aging disorder Hutchinson-Gilford progeria syndrome (HGPS) [52].  As vitamin D has been shown to increase Ca2+ levels in the cell, HGPS cells treated with vitamin D initially experienced a slight decrease in proliferation (i.e. “shock”) followed by an increased improvement in growth rate compared to control cells, effectively delaying premature entry into cellular senescence (i.e. “live”) [52]. 

Vitamin D administered to HIV-1 patients also led to an increase in activated CD4+ T cells in a placebo-controlled randomized study [53].  The phorbol ester PMA/TPA (which has been shown to induce mouse oocyte activation when combined with Ca2+ ionophores) and vitamin D have also been shown to stimulate HIV-1 proviral activation to similar levels in a cell line latently-infected with a monocytotropic strain of HIV-1JR-FL [54,55].

Startlingly, vitamin D has also been shown to induce the acrosome reaction in human sperm, a process that is indispensable for sperm-induced fertilization by facilitating penetration of the oocyte plasma membrane [56].  Interestingly, similar to oocyte activation, an increase in intracellular Ca2+ levels (i.e. “shock”) is also critical for the induction of the acrosome reaction in sperm and the same compounds that have been used to activate human oocytes to produce healthy children (e.g. A23187) (i.e. “live”) have also been used to induce the acrosome reaction in human sperm (i.e. “live”), indicating that the acrosome reaction is also likely dependent on AMPK activation [57].

The induction of cellular stress and the activation of AMPK thus appear to be a central biological theme that is likely responsible for the creation of all human life, the promotion of lifespan and healthspan, and the potential amelioration or eradication of disease.  Just as the “Shock and Live” approach defines the beginnings of life, the activation of T cells, and the improvement or reversal of accelerated aging symptoms in HGPS, AMPK activation, via application of the “Shock and Live” approach, also likely represents a common mechanism of action through which distinct compounds and methodologies are able to ameliorate or reverse neurodegeneration.     

References:

  1. Salt IP, Johnson G, Ashcroft SJ, Hardie DG. AMP-activated protein kinase is activated by low glucose in cell lines derived from pancreatic beta cells, and may regulate insulin release. Biochem J. 1998 Nov 1;335 ( Pt 3):533-9.

  1. Hardie DG. Sensing of energy and nutrients by AMP-activated protein kinase. Am J Clin Nutr. 2011 Apr;93(4):891S-6.

  1. Egan DF, Shackelford DB, Mihaylova MM, et al. Phosphorylation of ULK1 (hATG1) by AMP-activated protein kinase connects energy sensing to mitophagy. Science. 2011 Jan 28;331(6016):456-61.

  1. Blázquez C, Woods A, de Ceballos ML, Carling D, Guzmán M. The AMP-activated protein kinase is involved in the regulation of ketone body production by astrocytes. J Neurochem. 1999 Oct;73(4):1674-82.

  1. Kajita K, Mune T, Ikeda T, et al. Effect of fasting on PPARgamma and AMPK activity in adipocytes. Diabetes Res Clin Pract. 2008 Aug;81(2):144-9.

  1. Alcocer-Gómez E, Garrido-Maraver J, Bullón P, et al. Metformin and caloric restriction induce an AMPK-dependent restoration of mitochondrial dysfunction in fibroblasts from Fibromyalgia patients. Biochim Biophys Acta. 2015 Jul;1852(7):1257-67.

  1. Cho K, Chung JY, Cho SK, et al. Antihyperglycemic mechanism of metformin occurs via the AMPK/LXRα/POMC pathway. Sci Rep. 2015 Jan 30;5:8145.

  1. Chen Y, Qing W, Sun M, Lv L, Guo D, Jiang Y. Melatonin protects hepatocytes against bile acid-induced mitochondrial oxidative stress via the AMPK-SIRT3-SOD2 pathway. Free Radic Res. 2015 Oct;49(10):1275-84.

  1. Gibala MJ, McGee SL, Garnham AP, Howlett KF, Snow RJ, Hargreaves M. Brief intense interval exercise activates AMPK and p38 MAPK signaling and increases the expression of PGC-1alpha in human skeletal muscle. J Appl Physiol (1985). 2009 Mar;106(3):929-34.

  1. Yu DF, Shen ZC, Wu PF, et al. HFS-Triggered AMPK Activation Phosphorylates GSK3β and Induces E-LTP in Rat Hippocampus In Vivo. CNS Neurosci Ther. 2016 Jun;22(6):525-31.

  1. Sid V, Wu N, Sarna LK, Siow YL, House JD, OK. Folic acid supplementation during high-fat diet feeding restores AMPK activation via an AMP-LKB1-dependent mechanism. Am J Physiol Regul Integr Comp Physiol. 2015 Nov 15;309(10):R1215-25.

  1. Pu Y, Zhang H, Wang P, et al. Dietary curcumin ameliorates aging-related cerebrovascular dysfunction through the AMPK/uncoupling protein 2 pathway. Cell Physiol Biochem. 2013;32(5):1167-77.

  1. Wu Y, Zhang C, Dong Y, et al. Activation of the AMP-activated protein kinase by eicosapentaenoic acid (EPA, 20:5 n-3) improves endothelial function in vivo. PLoS One. 2012;7(4):e35508.

  1. Deng X, Dong Q, Bridges D, Raghow R, Park EA, Elam MB. Docosahexaenoic acid inhibits proteolytic processing of sterol regulatory element-binding protein-1c (SREBP-1c) via activation of AMP-activated kinase. Biochim Biophys Acta. 2015 Dec;1851(12):1521-9.

  1. Wang CZ, Wei D, Guan MP, Xue YM. Triiodothyronine regulates distribution of thyroid hormone receptors by activating AMP-activated protein kinase in 3T3-L1 adipocytes and induces uncoupling protein-1 expression. Mol Cell Biochem. 2014 Aug;393(1-2):247-54.

  1. Yang S, Wang J. Estrogen Activates AMP-Activated Protein Kinase in Human Endothelial Cells via ERβ/Ca(2+)/Calmodulin-Dependent Protein Kinase Kinase β Pathway. Cell Biochem Biophys. 2015 Jul;72(3):701-7.

  1. Mitsuhashi K, Senmaru T, Fukuda T, et al. Testosterone stimulates glucose uptake and GLUT4 translocation through LKB1/AMPK signaling in 3T3-L1 adipocytes. Endocrine. 2016 Jan;51(1):174-84.

  1. Pyra KA, Saha DC, Reimer RA. Prebiotic fiber increases hepatic acetyl CoA carboxylase phosphorylation and suppresses glucose-dependent insulinotropic polypeptide secretion more effectively when used with metformin in obese rats. J Nutr. 2012 Feb;142(2):213-20.

  1. Zhang M, Wang C, Wang C, et al. Enhanced AMPK phosphorylation contributes to the beneficial effects of Lactobacillus rhamnosus GG supernatant on chronic-alcohol-induced fatty liver disease. J Nutr Biochem. 2015 Apr;26(4):337-44.

  1. Thirunavukkarasu M, Penumathsa SV, Juhasz B, et al. Niacin-bound chromium enhances myocardial protection from ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol. 2006 Aug;291(2):H820-6.

  1. Sheu MJ, Chou PY, Lin WH, et al. Deep sea water modulates blood pressure and exhibits hypolipidemic effects via the AMPK-ACC pathway: an in vivo study. Mar Drugs. 2013 Jun 17;11(6):2183-202.  

  1. Swami S, Krishnan AV, Williams J, et al. Vitamin D mitigates the adverse effects of obesity on breast cancer in mice. Endocr Relat Cancer. 2016 Apr;23(4):251-64.

  1. Zhang Z, Zhao M, Li Q, Zhao H, Wang J, Li Y. Acetyl-l-carnitine inhibits TNF-alpha-induced insulin resistance via AMPK pathway in rat skeletal muscle cells. FEBS Lett. 2009 Jan 22;583(2):470-4.

  1. Wu SJ, Huang GY, Ng LT. γ-Tocotrienol induced cell cycle arrest and apoptosis via activating the Bax-mediated mitochondrial and AMPK signaling pathways in 3T3-L1 adipocytes. Food Chem Toxicol. 2013 Sep;59:501-13.

  1.  Hwang JT, Kim YM, Surh YJ, et al. Selenium regulates cyclooxygenase-2 and extracellular signal-regulated kinase signaling pathways by activating AMP-activated protein kinase in colon cancer cells. Cancer Res. 2006 Oct 15;66(20):10057-63.

  1. Gan W, Dang Y, Han X, et al. ERK5/HDAC5-mediated, resveratrol-, and pterostilbene-induced expression of MnSOD in human endothelial cells. Mol Nutr Food Res. 2016 Feb;60(2):266-77.

  1. Wang Y, Li X, Guo Y, Chan L, Guan X. alpha-Lipoic acid increases energy expenditure by enhancing adenosine monophosphate-activated protein kinase-peroxisome proliferator-activated receptor-gamma coactivator-1alpha signaling in the skeletal muscle of aged mice. Metabolism. 2010 Jul;59(7):967-76.

  1. Lee SK, Lee JO, Kim JH, et al. Coenzyme Q10 increases the fatty acid oxidation through AMPK-mediated PPARα induction in 3T3-L1 preadipocytes. Cell Signal. 2012 Dec;24(12):2329-36.  

  1. Wang G, Amato S, Gilbert J, Man HY. Resveratrol up-regulates AMPA receptor expression via AMP-activated protein kinase-mediated protein translation. Neuropharmacology. 2015 Aug;95:144-53.

  1. Zhang J, Meruvu S, Bedi YS, et al. Pyrroloquinoline quinone increases the expression and activity of Sirt1 and -3 genes in HepG2 cells. Nutr Res. 2015 Sep;35(9):844-9.

  1. Dong YM, Li Y, Ning H, Wang C, Liu JR, Sun CH. High dietary intake of medium-chain fatty acids during pregnancy in rats prevents later-life obesity in their offspring. J Nutr Biochem. 2011 Aug;22(8):791-7.

  1. Bredesen DE, Amos EC, Canick, et al. Reversal of cognitive decline in Alzheimer's disease. Aging (Albany NY). 2016 Jun;8(6):1250-8.

  1. Bredesen DE. Reversal of cognitive decline: a novel therapeutic program. Aging (Albany NY). 2014 Sep;6(9):707-17.

  1. http://www.nih.gov/news-events/news-releases/independent-panel-finds-insufficient-evidence-support-preventive-measures-alzheimers-disease

  1. Finley J. Oocyte activation and latent HIV-1 reactivation: AMPK as a common mechanism of action linking the beginnings of life and the potential eradication of HIV-1. Med Hypotheses. 2016 Aug;93:34-47.

  1. Yanagida K, Katayose H, Yazawa H, et al. Successful fertilization and pregnancy following ICSI and electrical oocyte activation. Hum Reprod. 1999 May;14(5):1307-11.

  1. Ebner T, Montag M, Oocyte Activation Study Group, et al. Live birth after artificial oocyte activation using a ready-to-use ionophore: a prospective multicentre study. Reprod Biomed Online. 2015 Apr;30(4):359-65.

  1.  Pearce EL, Walsh MC, Cejas PJ, et al. Enhancing CD8 T-cell memory by modulating fatty acid metabolism. Nature. 2009 Jul 2;460(7251):103-7.

  1. Blagih J, Coulombe F, Vincent EE, et al. The energy sensor AMPK regulates T cell metabolic adaptation and effector responses in vivo. Immunity. 2015 Jan 20;42(1):41-54. 

  1. Gómez-Gonzalo M, Carretero M, Rullas J et al. The hepatitis B virus X protein induces HIV-1 replication and transcription in synergy with T-cell activation signals: functional roles of NF-kappaB/NF-AT and SP1-binding sites in the HIV-1 long terminal repeat promoter. J Biol Chem. 2001 Sep 21;276(38):35435-43.

  1. Rao E, Zhang Y, Zhu G, et al. Deficiency of AMPK in CD8+ T cells suppresses their anti-tumor function by inducing protein phosphatase-mediated cell death. Oncotarget. 2015 Apr 10;6(10):7944-58.

  1. Yau SY, Li A, Hoo RL, et al. Physical exercise-induced hippocampal neurogenesis and antidepressant effects are mediated by the adipocyte hormone adiponectin. Proc Natl Acad Sci U S A. 2014 Nov 4;111(44):15810-5.

  1. Chung MM, Chen YL, Pei D, et al. The neuroprotective role of metformin in advanced glycation end product treated human neural stem cells is AMPK-dependent. Biochim Biophys Acta. 2015 May;1852(5):720-31.

  1. Kim DM, Leem YH. Chronic stress-induced memory deficits are reversed by regular exercise via AMPK-mediated BDNF induction. Neuroscience. 2016 Jun 2;324:271-85.

  1. Zhao YN, Li WF, Li F, et al. Resveratrol improves learning and memory in normally aged mice through microRNA-CREB pathway. Biochem Biophys Res Commun. 2013 Jun 14;435(4):597-602.

  1. Pan XY, Zhao W, Zeng XY, et al. Heat Shock Factor 1 Mediates Latent HIV Reactivation. Sci Rep. 2016 May 18;6:26294.

  1. Ya R, Downs SM. Suppression of chemically induced and spontaneous mouse oocyte activation by AMP-activated protein kinase. Biol Reprod. 2013 Mar 21;88(3):70.

  1. Chen F, Wang H, Xiang X, et al. Curcumin increased the differentiation rate of neurons in neural stem cells via wnt signaling in vitro study. J Surg Res. 2014 Dec;192(2):298-304.

  1. Kim HJ, Leeds P, Chuang DM. The HDAC inhibitor, sodium butyrate, stimulates neurogenesis in the ischemic brain. J Neurochem. 2009 Aug;110(4):1226-40.

  1. Imai K, Ochiai K, Okamoto T. Reactivation of latent HIV-1 infection by the periodontopathic bacterium Porphyromonas gingivalis involves histone modification. J Immunol. 2009 Mar 15;182(6):3688-95.

  1. Barquero AA, Dávola ME, Riva DA, Mersich SE, Alché LE. Naturally occurring compounds elicit HIV-1 replication in chronically infected promonocytic cells. Biomed Res Int. 2014;2014:989101.

  1. Kreienkamp R, Croke M, Neumann MA, et al. Vitamin D receptor signaling improves Hutchinson-Gilford progeria syndrome cellular phenotypes. Oncotarget. 2016 Apr 27. doi: 10.18632/oncotarget.9065.

  1. Bang U, Kolte L, Hitz M, et al. Correlation of increases in 1,25-dihydroxyvitamin D during vitamin D therapy with activation of CD4+ T lymphocytes in HIV-1-infected males. HIV Clin Trials. 2012 May-Jun;13(3):162-70.

  1. Uranga JA, Pedersen RA, Arechaga J. Parthenogenetic activation of mouse oocytes using calcium ionophores and protein kinase C stimulators. Int J Dev Biol. 1996 Apr;40(2):515-9.

  1. Kitano K, Rivas CI, Baldwin GC, Vera JC, Golde DW. Tumor necrosis factor-dependent production of human immunodeficiency virus 1 in chronically infected HL-60 cells. Blood. 1993 Nov 1;82(9):2742-8.

  1. Blomberg Jensen M, Dissing S. Non-genomic effects of vitamin D in human spermatozoa. Steroids. 2012 Aug;77(10):903-9.

  1. de Lamirande E, Tsai C, Harakat A, Gagnon C. Involvement of reactive oxygen species in human sperm arcosome reaction induced by A23187, lysophosphatidylcholine, and biological fluid ultrafiltrates. J Androl. 1998 Sep-Oct;19(5):585-94.

  1. Blomberg Jensen M, Dissing S. Non-genomic effects of vitamin D in human spermatozoa. Steroids. 2012 Aug;77(10):903-9.