Leaky intestine and impaired microbiome in an amyotrophic lateral sclerosis mouse model

Shaoping Wu, Jianxun Yi, Yong-Guo Zhang, Jingsong Zhou, Jun Sun, Shaoping Wu, Jianxun Yi, Yong-Guo Zhang, Jingsong Zhou, Jun Sun

Abstract

Emerging evidence has demonstrated that intestinal homeostasis and the microbiome play essential roles in neurological diseases, such as Parkinson's disease. Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by a progressive loss of motor neurons and muscle atrophy. Currently, there is no effective treatment. Most patients die within 3-5 years due to respiratory paralysis. Although the death of motor neurons is a hallmark of ALS, other organs may also contribute to the disease progression. We examined the gut of an ALS mouse model, G93A, which expresses mutant superoxide dismutase (SOD1(G93A)), and discovered a damaged tight junction structure and increased permeability with a significant reduction in the expression levels of tight junction protein ZO-1 and the adherens junction protein E-cadherin. Furthermore, our data demonstrated increased numbers of abnormal Paneth cells in the intestine of G93A mice. Paneth cells are specialized intestinal epithelial cells that can sense microbes and secrete antimicrobial peptides, thus playing key roles in host innate immune responses and shaping the gut microbiome. A decreased level of the antimicrobial peptides defensin 5 alpha was indeed found in the ALS intestine. These changes were associated with a shifted profile of the intestinal microbiome, including reduced levels of Butyrivibrio Fibrisolvens, Escherichia coli, and Fermicus, in G93A mice. The relative abundance of bacteria was shifted in G93A mice compared to wild-type mice. Principal coordinate analysis indicated a difference in fecal microbial communities between ALS and wild-type mice. Taken together, our study suggests a potential novel role of the intestinal epithelium and microbiome in the progression of ALS.

Keywords: Antimicrobial peptides; Paneth cells; ZO‐1; autophagy; cell biology; dysbiosis; intestinal permeability; microbiome; signal transduction; tight junction.

© 2015 The Authors. Physiological Reports published by Wiley Periodicals, Inc. on behalf of the American Physiological Society and The Physiological Society.

Figures

Figure 1
Figure 1
Disrupted intestinal junctions in the 2-month-old amyotrophic lateral sclerosis (ALS) mice. (A) Western blots of ZO-1 and E-cadherin in the colon. The relative band intensities of ZO-1 and E-cadherin are presented as the means ± SD (n = 3 per group, *P < 0.05). Western blot bands were quantified using Image Lab 4.01 (Bio-Rad). (B) Immunostaining of ZO-1 and E-cadherin in the colon. The tight junction protein ZO-1 shows discontinuity in the colon of ALS model mice. (C) H&E staining of the colon. Data are from a single experiment and are representative of n = 3 mice per group.
Figure 2
Figure 2
Enhanced IL-17 and intestinal permeability in amyotrophic lateral sclerosis (ALS) mice. (A, B) The inflammatory cytokine IL-17 in mouse serum and the small intestine (n = 3, *P < 0.05). (C) Intestinal permeability is increased in ALS mice (n = 3, *P < 0.05).
Figure 3
Figure 3
Defects in Paneth cells in the small intestine of amyotrophic lateral sclerosis (ALS) mice. (A) Lysozyme staining in the small intestine of ALS mice. (B) Number of Paneth cells/crypt in the small intestine. Ten slides from each mouse were counted. (C) Percentage of normal and abnormal Paneth cells in ALS mice (n = 3/group, *P < 0.05). (D) Defensin 5 alpha mRNA level in 3-month-old mice with symptoms (n = 3, *P < 0.05).
Figure 4
Figure 4
Shift of fecal microbial communities in amyotrophic lateral sclerosis (ALS) mice. (A) Butyrivibrio Fibrisolvens was decreased in G93A amyotrophic lateral sclerosis model mice. Real-time PCR of the bacterial universal 16S rRNA gene and 16S rRNA for Escherichia coli, Bacteroides Fragilis, and Butyrivibrio Fibrisolvens in fecal samples from both G93A and wild-type mice. Primers specific to universal 16S rRNA were used as an endogenous control to normalize loading between samples. The relative amount of 16S rRNA in each sample was estimated using the ΔΔCT method (n = 3, *P < 0.05). (B) Bacterial community of fecal samples from ALS and WT mice using 454 16S rRNA sequencing data (n = 3/group). (C) Principal coordinates analysis (PCoA) of unweighted UniFrac distances of 16S rRNA genes. An analysis was conducted for ALS (red) and WT (blue) mice. The results indicate that fecal microbial communities differ in ALS and WT mice.

References

    1. Alonso A, Logroscino G, Jick SS. Hernan MA. Incidence and lifetime risk of motor neuron disease in the United Kingdom: a population-based study. Eur. J. Neurol. 2009;16:745–751.
    1. Blikslager AT, Moeser AJ, Gookin JL, Jones SL. Odle J. Restoration of barrier function in injured intestinal mucosa. Physiol. Rev. 2007;87:545–564.
    1. Cadwell K, Liu JY, Brown SL, Miyoshi H, Loh J, Lennerz JK, et al. A key role for autophagy and the autophagy gene Atg16 l1 in mouse and human intestinal Paneth cells. Nature. 2008;456:259–263.
    1. Collins SM. Bercik P. The relationship between intestinal microbiota and the central nervous system in normal gastrointestinal function and disease. Gastroenterology. 2009;136:2003–2014.
    1. Crippa V, Boncoraglio A, Galbiati M, Aggarwal T, Rusmini P, Giorgetti E, et al. Differential autophagy power in the spinal cord and muscle of transgenic ALS mice. Front. Cell. Neurosci. 2013;7:234.
    1. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505:559–563.
    1. Deng HX, Shi Y, Furukawa Y, Zhai H, Fu R, Liu E, et al. Conversion to the amyotrophic lateral sclerosis phenotype is associated with intermolecular linked insoluble aggregates of SOD1 in mitochondria. Proc. Natl Acad. Sci. USA. 2006;103:7142–7147.
    1. Farhadi A, Banan A, Fields J. Keshavarzian A. Intestinal barrier: an interface between health and disease. J. Gastroenterol. Hepatol. 2003;18:479–497.
    1. Finegold SM, Dowd SE, Gontcharova V, Liu C, Henley KE, Wolcott RD, et al. Pyrosequencing study of fecal microflora of autistic and control children. Anaerobe. 2010;16:444–453.
    1. Forsyth CB, Shannon KM, Kordower JH, Voigt RM, Shaikh M, Jaglin JA, et al. Increased intestinal permeability correlates with sigmoid mucosa alpha-synuclein staining and endotoxin exposure markers in early Parkinson's disease. PLoS One. 2011;6:e28032.
    1. Fung KY, Cosgrove L, Lockett T, Head R. Topping DL. A review of the potential mechanisms for the lowering of colorectal oncogenesis by butyrate. Br. J. Nutr. 2012;108:820–831.
    1. Gurney ME, Pu H, Chiu AY, Dal Canto MC, Polchow CY, Alexander DD, et al. Motor neuron degeneration in mice that express a human Cu, Zn superoxide dismutase mutation. Science. 1994;264:1772–1775.
    1. Hsiao EY, McBride SW, Hsien S, Sharon G, Hyde ER, McCue T, et al. Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders. Cell. 2013;155:1451–1463.
    1. Ivanova MI, Sievers SA, Guenther EL, Johnson LM, Winkler DD, Galaleldeen A, et al. Aggregation-triggering segments of SOD1 fibril formation support a common pathway for familial and sporadic ALS. Proc. Natl Acad. Sci. USA. 2014;111:197–201.
    1. Joyce PI, Fratta P, Fisher EM. Acevedo-Arozena A. SOD1 and TDP-43 animal models of amyotrophic lateral sclerosis: recent advances in understanding disease toward the development of clinical treatments. Mamm. Genome. 2011;22:420–448.
    1. Kim J, Lee H, Lee JH, Kwon DY, Genovesio A, Fenistein D, et al. Dimerization, oligomerization, and aggregation of human amyotrophic lateral sclerosis Cu/Zn-superoxide dismutase 1 mutant forms in live cells. J. Biol. Chem. 2014;289:15094–15103.
    1. Klionsky DJ, Abeliovich H, Agostinis P, Agrawal DK, Aliev G, Askew DS, et al. Guidelines for the use and interpretation of assays for monitoring autophagy in higher eukaryotes. Autophagy. 2008;4:151–175.
    1. Laukoetter MG, Nava P, Lee WY, Severson EA, Capaldo CT, Babbin BA, et al. JAM-A regulates permeability and inflammation in the intestine in vivo. J. Exp. Med. 2007;204:3067–3076.
    1. Lee H, Ko EH, Lai M, Wei N, Balroop J, Kashem Z, et al. Delineating the relationships among the formation of reactive oxygen species, cell membrane instability and innate autoimmunity in intestinal reperfusion injury. Mol. Immunol. 2014;58(2):151–159.
    1. Li L, Zhang X. Le W. Altered macroautophagy in the spinal cord of SOD1 mutant mice. Autophagy. 2008;4:290–293.
    1. Liao AP, Petrof EO, Kuppireddi S, Zhao Y, Xia Y, Claud EC, et al. Salmonella type III effector AvrA stabilizes cell tight junctions to inhibit inflammation in intestinal epithelial cells. PLoS One. 2008;3:e2369.
    1. Ling J, Liao H, Clark R, Wong MS. Lo DD. Structural constraints for the binding of short peptides to claudin-4 revealed by surface plasmon resonance. J. Biol. Chem. 2008;283:30585–30595.
    1. Luo G, Yi J, Ma C, Xiao Y, Yi F, Yu T, et al. Defective mitochondrial dynamics is an early event in skeletal muscle of an amyotrophic lateral sclerosis mouse model. PLoS One. 2013;8:e82112.
    1. Maes M, Kubera M. Leunis JC. The gut-brain barrier in major depression: intestinal mucosal dysfunction with an increased translocation of LPS from gram negative enterobacteria (leaky gut) plays a role in the inflammatory pathophysiology of depression. Neuro Endocrinol. Lett. 2008;29:117–124.
    1. McCombe PA. Henderson RD. The Role of immune and inflammatory mechanisms in ALS. Curr. Mol. Med. 2011;11:246–254.
    1. Miletta MC, Petkovic V, Eblé A, Ammann RA, Flück CE. Mullis PE. Butyrate increases intracellular calcium levels and enhances growth hormone release from rat anterior pituitary cells via the G-protein-coupled receptors GPR41 and 43. PLoS One. 2014;9:e107388.
    1. Murrow L. Debnath J. Autophagy as a stress–response and quality-control mechanism: implications for cell injury and human disease. Annu. Rev. Pathol. 2013;8:105–137.
    1. Pasinelli P. Brown RH. Molecular biology of amyotrophic lateral sclerosis: insights from genetics. Nat. Rev. Neurosci. 2006;7:710–723.
    1. Pfeiffer RF. Gastrointestinal dysfunction in Parkinson's disease. Lancet Neurol. 2003;2:107–116.
    1. Phan TG, Green JA, Gray EE, Xu Y. Cyster JG. Immune complex relay by subcapsular sinus macrophages and noncognate B cells drives antibody affinity maturation. Nat. Immunol. 2009;10:786–793.
    1. Rajapaksa TE, Stover-Hamer M, Fernandez X, Eckelhoefer HA. Lo DD. Claudin 4-targeted protein incorporated into PLGA nanoparticles can mediate M cell targeted delivery. J. Control. Release. 2010;142:196–205.
    1. Salzman NH, Hung K, Haribhai D, Chu H, Karlsson-Sjoberg J, Amir E, et al. Enteric defensins are essential regulators of intestinal microbial ecology. Nat. Immunol. 2009;11:76–83.
    1. Sasaki S. Autophagy in spinal cord motor neurons in sporadic amyotrophic lateral sclerosis. J. Neuropathol. Exp. Neurol. 2011;70:349–359.
    1. Schulz MD, Atay C, Heringer J, Romrig FK, Schwitalla S, Aydin B, et al. High-fat-diet-mediated dysbiosis promotes intestinal carcinogenesis independently of obesity. Nature. 2014;514:508–512.
    1. Shen L. Turner JR. Role of epithelial cells in initiation and propagation of intestinal inflammation. Eliminating the static: tight junction dynamics exposed. Am. J. Physiol. Gastrointest. Liver Physiol. 2006;290:G577–G582.
    1. Sun J. Chang BE. Exploring gut microbes in human health and disease: pushing the envelope. Genes Dis. 2014;1:132–139.
    1. Takahashi N, Vanlaere I, de Rycke R, Cauwels A, Joosten LA, Lubberts E, et al. IL-17 produced by Paneth cells drives TNF-induced shock. J. Exp. Med. 2008;205:1755–1761.
    1. Turnbaugh PJ, Ley RE, Hamady M, Fraser-Liggett CM, Knight R. Gordon JI. The human microbiome project. Nature. 2007;449:804–810.
    1. Virgin HW. Levine B. Autophagy genes in immunity. Nat. Immunol. 2009;10:461–470.
    1. Wu S, Zhang YG, Lu R, Xia Y, Zhou D, Petrof EO, et al. Intestinal epithelial vitamin D receptor deletion leads to defective autophagy in colitis. Gut. 2014 . doi: .
    1. Xiao Y, Ma C, Wu S, Yi SJ, Lin P, Sun J, et al. Suppressed autophagy flux in skeletal muscle of an amyotrophic lateral sclerosis mouse model upon disease progression. Physiol. Rep. 2015;3:e12271.
    1. Yi J, Ma C, Li Y, Weisleder N, Ríos E, Ma J, et al. Mitochondrial calcium uptake regulates rapid calcium transients in skeletal muscle during excitation–contraction (E–C) coupling. J. Biol. Chem. 2011;286:32436–32443.
    1. Yuk JM, Yoshimori T. Jo EK. Autophagy and bacterial infectious diseases. Exp. Mol. Med. 2012;44:99–108.
    1. Zhang C, Li S, Yang L, Huang P, Li W, Wang S, et al. Structural modulation of gut microbiota in life-long calorie-restricted mice. Nat. Commun. 2013a;4:2163.
    1. Zhang YG, Wu S, Xia Y. Sun J. Salmonella infection upregulates the leaky protein claudin-2 in intestinal epithelial cells. PLoS One. 2013b;8:e58606.
    1. Zhang X, Chen S, Song L, Tang Y, Shen Y, Jia L, et al. MTOR-independent, autophagic enhancer trehalose prolongs motor neuron survival and ameliorates the autophagic flux defect in a mouse model of amyotrophic lateral sclerosis. Autophagy. 2014;10:588–602.
    1. Zhou J, Yi J, Fu R, Liu E, Siddique T, Ríos E, et al. Hyperactive intracellular calcium signaling associated with localized mitochondrial defects in skeletal muscle of an animal model of amyotrophic lateral sclerosis. J. Biol. Chem. 2010;285:705–712.

Source: PubMed

3
Suscribir