Monday, December 11, 2017

Metformin shown for the first time to inhibit Zika virus in human umbilical cells: AMPK links virus destruction with Progeria, HIV & Cancer stem cells

By Jim Gathany [Public domain], via Wikimedia Commons; CC-BY-SA-3.0 http://creativecommons.org/licenses/by-sa/3.0/)], via Wikimedia Commons


A study published in the Journal of Virology in December of 2017 by researchers from the University of Southern California showed for the first time that AMPK activators including the anti-diabetic drug metformin caused an approximately 60% to 80% decrease in virus production from human umbilical vein endothelial cells (HUVECs) infected with Zika virus (ZIKV) [1]. ZIKV has been causally linked to microcephaly (head circumference smaller than normal due to abnormal brain development) and has been shown to efficiently infect HUVECs, which directly contact the fetal blood stream [2]. Metformin-induced inhibition of ZIKV replication in HUVECs may thus represent a powerful, safe, and economically viable option to treat and/or prevent conditions associated with ZIKV infection. Interestingly, as further explained below, metformin and AMPK have recently been shown to exert antiviral and anti-parasitic effects against dengue virus (which is transmitted by the same mosquito vector as ZIKV) and different Plasmodium species (the etiological agent of malaria), respectively.

Additionally, metformin activates AMPK and alleviates accelerating aging defects in cells from children with the genetic disorder Hutchinson-Gilford progeria syndrome (HGPS), promotes the differentiation and/or apoptosis of cancer stem cells in an AMPK-dependent manner, and destabilizes the latent HIV-1 reservoir in chronically-infected HIV-1 patients (facilitating virus elimination and potentially contributing to an HIV-1 cure). Also, because AMPK is critical for oocyte maturation and bacteria-derived antibiotics (e.g. ionomycin, A23187) that activate AMPK are used extensively to activate human oocytes to create normal, healthy babies, it is likely that stress-induced AMPK activation (e.g. via reactive oxygen species, intracellular calcium, and/or AMP/ATP ratio increase, etc.) represents a common mechanism linking pathogen elimination with HGPS, caner stem cell elimination, HIV-1 eradication, and the creation of all human life, as I originally proposed in several recent publications (see below) [3-6].   

As noted above, the AMPK activators metformin and AICAR potently inhibited ZIKV replication in HUVECs [1]. Although the authors unexplainably found that compound C (an AMPK inhibitor) also inhibited ZIKV replication, AMPK activation has recently been found to exert significant antiviral effects against Rift Valley Fever virus as well as multiple arbovirus family members including the Flavivirus Kunjin virus, the Togavirus Sindbis virus, and the Rhabdovirus Vesicular stomatitis virus [1,7]. Indeed, several AMPK-activating compounds have also recently demonstrated antiviral effects against ZIKV infection and replication. For example, EGCG (found in green tea) inhibited ZIKV entry in Vero E6 cells, curcumin (derived from the plant Curcuma longa) inhibited ZIKV replication in HeLa cells, NDGA (derived from the plant Larrea tridentate) reduced viral yield in Vero cells infected with a ZIKV strain isolated from a human patient, sophoraflavenone G (isolated from the plant Sophora Flavecens) inhibits ZIKV replication in A549 cells, hemin (an iron-containing porphyrin) significantly inhibited ZIKV replication in primary human monocyte-derived macrophages, quercetin (found in a variety of plants) exerted antiviral activity against ZIKV in both tissue culture and knockout mice, and chloroquine (an anti-malarial compound) inhibited ZIKV infection in vitro and protected fetal mice from ZIKV-induced microcephaly [8-14]. Similar to metformin, each of the aforementioned compounds or the plant extracts from which they are derived activates AMPK in vivo and/or in vitro [15-19].

Interestingly, as both ZIKV and dengue virus (DENV) are transmitted by the same mosquito vector, a study recently published in the journal PLoS Pathogens in April of 2017 demonstrated for the first time that metformin exerted significant antiviral effects in DENV-infected human liver cells that was dependent on activation of the master metabolic regulator AMPK [20]. The authors showed that an increase in HMG-CoA reductase (HMGCR) activity, a target of AMPK, was associated with DENV-infected cells, AMPK activation was reduced in DENV-infected cells at 12 and 24 hours post infection (hpi), and metformin significantly decreased the number of infected cells, viral yield, and viral genome copies, leading the authors to conclude that metformin-induced AMPK activation generates a strong antiviral effect against DENV [20]. Recent efforts funded by the U.S. and British governments, the Bill & Melinda Gates Foundation, and the Google health spin-off Verily have sought to decrease the spread of dengue and Zika viruses through the coordinated release of female and/or male mosquitoes (called Aedes aegypti) that were purposely infected with a bacterium that inhibits the mosquito’s ability to transmit the two viruses to humans [21,22]. Studies have shown that this bacterium, called Wolbachia, enhances the mosquito’s immune response by increasing the levels of reactive oxygen species (ROS), thus enhancing inhibition of DENV replication [23]. Because AMPK is activated by cellular stress (e.g. ROS increase, intracellular calcium [Ca2+] increase, AMP/ATP ratio increase, etc.), has been found in Aedes aegypti (Ae. aegypti), and AMPK activation by stress-inducing compounds (e.g. resveratrol) increased average life span and enhanced the immune response in Ae. aegypti in an AMPK-dependent manner, the recent finding that metformin also inhibits DENV replication in human cells in an AMPK-dependent manner provides compelling evidence that the anti-viral and antimicrobial effects of AMPK activation likely crosses species boundaries [24]. 

Moreover, a study published in the journal Cell Reports in September of 2016 by researchers from the Massachusetts Institute of Technology (MIT) and the University of Lisbon also showed for the first time that metformin and other AMPK activators significantly reduced parasite load in human liver cells of different species of Plasmodium, a protozoan parasite that is the etiological agent of malaria [25]. Importantly, the authors also showed that AMPK activation inhibits growth and replication of different Plasmodium spp. (species) and AMPK activators as well as dietary restriction, which activates AMPK, reduces Plasmodium berghei (malaria-causing species in rodents often used as a model for the study of human malaria) infection in mice [25]. The AMPK-activating compounds salicylate and A769662 also reduced P. berghei and P. falciparum (malaria parasite that infects humans) merozoite formation (infectious parasites generated through replication in erythrocytes) in vitro while salicylate decreased parasitaemia in mice in vivo [26].

Cellular stress-induced AMPK activation has also been shown to exert antiviral effects against HIV-1. AMPK activation and several AMPK-activating compounds, including EGCG, curcumin, tanshinone II A (derived from the plant Salvia miltiorrhiza), byrostatin-1 (isolated from the marine organism Bugula neritina), and resveratrol (found in grapes and in the plant Polygonum cuspidatum) have been shown to exhibit antiviral activity in vitro against HIV-1 [27-31].

An active area among HIV-1 cure researchers, known as the “shock and kill” approach, 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]. Strikingly, 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 [5,32,33]. A recent study 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 [30]. 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 [34-36].

Perhaps most convincingly, at the International AIDS Society’s (IAS) HIV Cure and Cancer Forum held in Paris, France in July of 2017, researchers from the University of Hawaii demonstrated for the first time that metformin decreased the percentage of CD4+ T cells expressing the immune checkpoint receptors PD-1, TIGIT, and TIM-3 in HIV-1 patients, receptors that are positively associated with T cells that harbor latent HIV-1. Metformin also destabilized the latent viral reservoir in chronically-infected HIV-1 patients, indicating that metformin may indeed contribute to HIV-1 eradication by inducing an AMPK-mediated reactivation of latent HIV-1, as I initially proposed in 2015 and 2016 [4,5, 37-41].

Also, stress-induced AMPK activation likely also links latent HIV-1 reactivation with alleviation of accelerated aging defects in cells derived from children with the genetic disorder Hutchinson-Gilford progeria syndrome (HGPS). Studies have shown that efficient reactivation of latent HIV-1 involves a reduction in the splicing of the HIV-1 genome by the gene splicing factor SRSF1 [42-44]. Accelerated cellular aging-like phenotypes in HGPS are primarily linked to aberrant splicing of the LMNA gene, leading to the over production of a toxic protein called progerin [3]. Evidence has also shown that inhibition of the splicing factor SRSF1 leads to a reduction in progerin at both the mRNA and protein levels [4,45].

A recent study published online in the Journal npj Aging and Mechanisms of Disease in November of 2016 provided startling evidence that metformin decreased the expression of progerin and SRSF1 and alleviated pathological defects in cells derived from HGPS patients [46]. Another study published online in the Journal Experimental Dermatology in February of 2017 confirmed that metformin alleviated nuclear defects and premature aging phenotypes and activated AMPK in fibroblasts derived from HGPS patients, substantiating my original hypotheses from 2014 and 2015 proposing that AMPK activators including metformin would improve accelerated aging defects in HGPS cells by inhibiting SRSF1 and activating AMPK [3,4,47]. Temsirolimus, an analog of the macrolide rapamycin (which activates AMPK in vivo), also partially rescued the HGPS cellular phenotype but significantly increased the levels of ROS and superoxide within the first hour of treatment, providing further indication that the induction of cellular stress and subsequent AMPK activation links virus and pathogen elimination with alleviation of accelerated cellular aging defects in HGPS [48,49].

Furthermore, ROS and calcium are well-studied mediators of cellular stress-induced differentiation of embryonic and adult stem cells, AMPK has recently been shown to be essential for mouse embryonic stem cell differentiation, and metformin targets and promotes differentiation and/or apoptosis of cancer stem cells in the deadliest of cancers in an AMPK-dependent manner, including glioblastoma and pancreatic cancer [6]. Such evidence strongly suggests that cellular stress-induced AMPK activation by compounds including metformin links pathogen and virus elimination with HGPS and cancer stem cell differentiation and/or apoptosis, a hypothesis that I first proposed in 2017 [6].

Lastly, AMPK activation also promotes oocyte meiotic induction and maturation (processes that are critical for efficient oocyte activation) and AMPK has recently been found localized across the entire acrosome in human spermatozoa [5,50,51]. The induction of cellular stress (e.g. increases in ROS, intracellular calcium, and/or AMP/ATP ratio increase), which activates AMPK, also promotes oocyte meiotic induction/maturation, oocyte activation, and the acrosome reaction in human sperm, processes critical for the creation of all human life [50,52,53]. Indeed, oocyte activation is indispensable for the creation of all human life and the bacteria-derived calcium ionophore ionomycin, which activates AMPK, is commonly used to promote latent HIV-1 reactivation and is extensively used to activate human oocytes, creating normal healthy children (i.e. the “shock and live” approach) [53-57]. Such evidence indicates and further substantiates the novel and provocative assertion that AMPK activation links Zika virus inhibition and pathogen elimination with the amelioration of accelerated aging defects in HGPS cells, HIV-1 latency and replication, adult and cancer stem cells, and the creation of all human life [1,3-6]. 

https://www.linkedin.com/pulse/metformin-shown-first-time-inhibit-zika-virus-human-umbilical-finley/



References

  1. Cheng F, Ramos da Silva S, Huang IC, Jung JU, Gao SJ. Suppression of Zika virus infection and replication in endothelial cells and astrocytes by PKA inhibitor PKI 14-22. J Virol. 2017 Dec 6. pii: JVI.02019-17. doi: 10.1128/JVI.02019-17. [Epub ahead of print].
  2. Richard AS, Shim BS, Kwon YC, et al. AXL-dependent infection of human fetal endothelial cells distinguishes Zika virus from other pathogenic flaviviruses. Proc Natl Acad Sci U S A. 2017 Feb 21;114(8):2024-2029.
  3. 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.
  4. 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.
  5. 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.
  6. Finley J. Elimination of cancer stem cells and reactivation of latent HIV-1 via AMPK activation: Common mechanism of action linking inhibition of tumorigenesis and the potential eradication of HIV-1. Med Hypotheses. 2017 Jul;104:133-146.
  7. Moser TS, Schieffer D, Cherry S. AMP-activated kinase restricts Rift Valley fever virus infection by inhibiting fatty acid synthesis. PLoS Pathog. 2012;8(4):e1002661.
  8. Carneiro BM, Batista MN, Braga AC, Nogueira ML, Rahal P. The green tea molecule EGCG inhibits Zika virus entry. Virology. 2016 Sep;496:215-8.
  9. Mounce BC, Cesaro T, Carrau L, Vallet T, Vignuzzi M. Curcumin inhibits Zika and chikungunya virus infection by inhibiting cell binding. Antiviral Res. 2017 Jun;142:148-157.
  10. Merino-Ramos T, Jiménez de Oya N, Saiz JC, Martín-Acebes MA. Antiviral activity of nordihydroguaiaretic acid and its derivative tetra-O-methyl nordihydroguaiaretic acid against West Nile virus and Zika virus. Antimicrob Agents Chemother. 2017 May 15. pii: AAC.00376-17.
  11. Sze A, Olagnier D, Hadj SB, et al. Sophoraflavenone G Restricts Dengue and Zika Virus Infection via RNA Polymerase Interference. Viruses. 2017 Oct 3;9(10). pii: E287.
  12. Huang H, Falgout B, Takeda K, Yamada KM, Dhawan S. Nrf2-dependent induction of innate host defense via heme oxygenase-1 inhibits Zika virus replication. Virology. 2017 Mar;503:1-5.
  13. Wong G, He S, Siragam V, et al. Antiviral activity of quercetin-3-β-O-D-glucoside against Zika virus infection. Virol Sin. 2017 Sep 5. doi: 10.1007/s12250-017-4057-9. [Epub ahead of print].
  14. Li C, Zhu X, Ji X, et al. Chloroquine, a FDA-approved Drug, Prevents Zika Virus Infection and its Associated Congenital Microcephaly in Mice. EBioMedicine. 2017 Oct;24:189-194.
  15. Kim I, He YY. Targeting the AMP-Activated Protein Kinase for Cancer Prevention and Therapy. Front Oncol. 2013 Jul 15;3:175.
  16. Zhang H, Li Y, Hu J, et al. Effect of Creosote Bush-Derived NDGA on Expression of Genes Involved in Lipid Metabolism in Liver of High-Fructose Fed Rats: Relevance to NDGA Amelioration of Hypertriglyceridemia and Hepatic Steatosis. PLoS One. 2015 Sep 22;10(9):e0138203.
  17. Yang X, Yang J, Xu C, et al. Antidiabetic effects of flavonoids from Sophora flavescens EtOAc extract in type 2 diabetic KK-ay mice. J Ethnopharmacol. 2015 Aug 2;171:161-70.
  18. Park EJ, Kim YM, Chang KC. Hemin Reduces HMGB1 Release by UVB in an AMPK/HO-1-dependent Pathway in Human Keratinocytes HaCaT Cells. Arch Med Res. 2017 Jul;48(5):423-431.
  19. Spears LD, Tran AV, Qin CY, et al. Chloroquine increases phosphorylation of AMPK and Akt in myotubes. Heliyon. 2016 Mar;2(3):e00083.
  20. Soto-Acosta R, Bautista-Carbajal P, Cervantes-Salazar M, Angel-Ambrocio AH, Del Angel RM. DENV up-regulates the HMG-CoA reductase activity through the impairment of AMPK phosphorylation: A potential antiviral target. PLoS Pathog. 2017 Apr 6;13(4):e1006257.
  21. http://www.reuters.com/article/us-health-dengue-mosquitoes-idUSKCN12Q1PE
  22. https://www.bloomberg.com/news/articles/2016-10-06/alphabet-s-verily-joins-zika-fight-with-sterile-mosquito-lab
  23. Pan X, Zhou G, Wu J, et al. Wolbachia induces reactive oxygen species (ROS)-dependent activation of the Toll pathway to control dengue virus in the mosquito Aedes aegypti. Proc Natl Acad Sci U S A. 2012 Jan 3;109(1):E23-31.
  24. Nunes RD, Ventura-Martins G, Moretti DM, et al. Polyphenol-Rich Diets Exacerbate AMPK-Mediated Autophagy, Decreasing Proliferation of Mosquito Midgut Microbiota, and Extending Vector Lifespan. PLoS Negl Trop Dis. 2016 Oct 12;10(10):e0005034.
  25. Ruivo MT, Vera IM, Sales-Dias J, et al. Host AMPK Is a Modulator of Plasmodium Liver Infection. Cell Rep. 2016 Sep 6;16(10):2539-45.
  26. Mancio-Silva L, Slavic K, Grilo Ruivo MT, et al. Nutrient sensing modulates malaria parasite virulence. Nature. 2017 Jul 13;547(7662):213-216.
  27. Zhang HS, Wu TC, Sang WW, Ruan Z. EGCG inhibits Tat-induced LTR transactivation: role of Nrf2, AKT, AMPK signaling pathway. Life Sci. 2012 May 22;90(19-20):747-54.
  28. Zhang HS, Ruan Z, Sang WW. HDAC1/NFκB pathway is involved in curcumin inhibiting of Tat-mediated long terminal repeat transactivation. J Cell Physiol. 2011 Dec;226(12):3385-91.
  29. Zhang HS, Chen XY, Wu TC, Zhang FJ. Tanshinone II A inhibits tat-induced HIV-1 transactivation through redox-regulated AMPK/Nampt pathway. J Cell Physiol. 2014 Sep;229(9):1193-201.
  30. 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.
  31. Zhang HS, Wu MR. SIRT1 regulates Tat-induced HIV-1 transactivation through activating AMP-activated protein kinase. Virus Res. 2009 Dec;146(1-2):51-7.
  32. 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.
  33. 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.
  34. 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.
  35. 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.
  36. 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.
  37. G.M. Chew, D.C. Chow, S.A. Souza, et al. Impact of adjunctive metformin therapy on T cell exhaustion and viral persistence in a clinical trial of HIV-infected adults on suppressive ART. Journal of Virus Eradication 2017; 3 (Supplement 1): 6–19.
  38. http://viruseradication.com/supplement-details/Abstracts_of_the_IAS_HIV_Cure_and_Cancer_Forum_2017/
  39. http://www.iasociety.org/HIV-Programmes/Towards-an-HIV-Cure/Events/HIV-Cure-Cancer-Forum
  40. Fromentin R, Bakeman W, Lawani MB, et al. CD4+ T Cells Expressing PD-1, TIGIT and LAG-3 Contribute to HIV Persistence during ART. PLoS Pathog. 2016 Jul 14;12(7):e1005761.
  41. Chew GM, Fujita T, Webb GM, et al. TIGIT Marks Exhausted T Cells, Correlates with Disease Progression, and Serves as a Target for Immune Restoration in HIV and SIV Infection. PLoS Pathog. 2016 Jan 7;12(1):e1005349.
  42. 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.
  43. 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.
  44. 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.
  45. 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.
  46. Egesipe AL, Blondel S, Cicero AL, et al. Metformin decreases progerin expression and alleviates pathological defects of Hutchinson-Gilford progeria syndrome cells. NPJ Aging Mech Dis. 2016 Nov 10;2:16026.
  47. Park SK, Shin OS. Metformin Alleviates Ageing Cellular Phenotypes in Hutchinson-Gilford Progeria Syndrome Dermal Fibroblasts. Exp Dermatol. 2017 Oct;26(10):889-895.
  48. Gabriel D, Gordon LB, Djabali K. Temsirolimus Partially Rescues the Hutchinson-Gilford Progeria Cellular Phenotype. PLoS One. 2016 Dec 29;11(12):e0168988.
  49. 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.
  50. LaRosa C, Downs SM. Stress stimulates AMP-activated protein kinase and meiotic resumption in mouse oocytes. Biol Reprod. 2006 Mar;74(3):585-92.
  51. 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].
  52. 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.
  53. Tesarik J, Sousa M, Testart J. Human oocyte activation after intracytoplasmic sperm injection. Hum Reprod. 1994 Mar;9(3):511-8.
  54. Deemeh MR, Tavalaee M, Nasr-Esfahani MH. Health of children born through artificial oocyte activation: a pilot study. Reprod Sci. 2015 Mar;22(3):322-8.
  55. 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.
  56. 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.
  57. Liu WC, Slusarchyk DS, Astle G, Trejo WH, Brown WE, Meyers E. Ionomycin, a new polyether antibiotic. J Antibiot (Tokyo). 1978 Sep;31(9):815-9.