Innervation of the gastrointestinal tract: patterns of aging

Robert J Phillips, Terry L Powley, Robert J Phillips, Terry L Powley

Abstract

The gastrointestinal (GI) tract is innervated by intrinsic enteric neurons and by extrinsic projections, including sympathetic and parasympathetic efferents as well as visceral afferents, all of which are compromised by age to different degrees. In the present review, we summarize and illustrate key structural changes in the aging innervation of the gut, and suggest a provisional list of the general patterns of aging of the GI innervation. For example, age-related neuronal losses occur in both the myenteric plexus and submucosal plexus of the intestines. These losses start in adulthood, increase over the rest of the life span, and are specific to cholinergic neurons. Parallel losses of enteric glia also occur. The extent of neuronal and glial loss varies along an oral-to-anal gradient, with the more distal GI tract being more severely affected. Additionally, with aging, dystrophic axonal swellings and markedly dilated varicosities progressively accumulate in the sympathetic, vagal, dorsal root, and enteric nitrergic innervation of the gut. These dramatic and consistent patterns of neuropathy that characterize the aging autonomic nervous system of the GI tract are candidate mechanisms for some of the age-related declines in function evidenced in the elderly.

Figures

Figure 1
Figure 1
Dramatic age-related changes occur in the myenteric plexus. Immunostaining of neurons (left column, HuC/D) and glia (middle column, S-100) in the myenteric plexus. The merged images (right column) illustrate the relationship of neurons (red) and glia (green). At 6 months of age, HuC/D strongly labeled the nuclei of neurons with weaker labeling of the cytoplasm and no labeling of the neuronal processes, and S-100 strongly labeled the soma of the glia with weaker labeling of processes (A). At 26 months of age, there was a reduction in the presence of the HuC/D antigen in the neurons of the small (B) and large (C) intestines. The complete loss of HuC/D staining of some neurons at 26 months is in stark contrast to the strong labeling of other neurons within the same ganglion, and is consistent with the observation that age-related neuron loss is specific to a subpopulation of neurons. The complete deterioration of some neurons within a ganglion is indicated by the presence of granular debris or lipofuscin within unstained silhouettes outlined by the glial scaffolding. Scale bar = 50 μm in C (applies to A-C).
Figure 2
Figure 2
Age-related neuronal loss in the myenteric plexus of the colon was evident as early as 12 months of age and progressed in a linear manner with increasing age (combined data from Phillips and Powley, 2001; Phillips et al., 2003, 2006a). A runs test following linear regression analysis verified that the distribution of the data was not significantly nonlinear (runs test, p = 0.54). A mean ± S.E.M of 248 ± 19 neurons/cm2/month are lost with age (goodness of fit, r2 = 0.71). The mean (± S.E.M) density of neurons, which were corrected for age-related changes in tissue area, was 146 (3.6), 138 (10), 125 (5.6), 116 (6.6), 101 (3.3), 89 (1.7), and 91 (3.2) neurons/mm2 at 3, 6, 12, 18, 21, 24, and 27 months of age, respectively. The analysis was based on a sample size of 12, 6, 12, 6, 5, 16, and 12 rats at 3, 6, 12, 18, 21, 24, and 27 months of age, respectively.
Figure 3
Figure 3
The pattern of neuronal loss with age was similar for both the myenteric and submucosal plexuses (data from Phillips et al., 2006a). Total neuronal estimates for both plexuses are from the same rats with 6 rats/age sampled.
Figure 4
Figure 4
The myenteric plexus can be broken down into different subpopulations based upon the neurochemical phenotype of the neurons, and aging appears to affect the subpopulations differently. All of the neurons in the myenteric plexus were labeled using the putative pan-neuronal marker HuC/D (brown), and the subpopulation of neurons that are nitrergic were labeled using the histochemical reaction NADPHd (dark blue; A-C). There is no loss of nitrergic neurons throughout the gastrointestinal tract with age; however, markedly swollen nitrergic axons are routinely found in the smooth muscle (B). In addition, swollen nitrergic axons and dystrophic varicosities were also found in the myenteric ganglia (C; HuC/D neurons brown; nitrergic neurons dark blue with nuclei in silhouette; varicosities solid dark blue elements, some as large as neuronal somata) of aged rats. The majority of the myenteric neurons (blue, HuC/D) in the small intestine of the rat that are positive for the calcium binding protein calbindin (green) are not part of the nitrergic (red, nNOS) phenotype (D). Calbindin-positive (brown) neurons are a small subpopulation of the total population of neurons (light blue, Cuprolinic Blue) within the myenteric plexus (E). We have identified markedly swollen axons in the myenteric plexus of middle-age (15 months of age, F) and aged (24 months of age, not shown) Fischer 344 rats that are positive for calbindin (large brown varicosities and brown axons; blue neuronal counterstain, Cuprolinc Blue). Scale bars = 50 μm in F (applies to A,E,F), D (applies to C,D); 100 μm in B.
Figure 5
Figure 5
A reduction in ganglionic area occurs with age. Cuprolinic Blue labeled neurons in the submucosal plexus of the colon. Neurons were well defined with stained Nissl substance outlining the unstained nuclei. At 6 months of age, neurons within a ganglion were evenly spaced, providing room for unstained axons, dendrites, and glia (A). In the plexus of 27-month-old rats, ganglia were often observed that appeared to have collapsed in upon themselves (B,C), with little or no evidence of neuropil, i.e., the ganglionic tissue surrounding the neuronal cell bodies (e.g., glial cells, nerve fibers and processes of ganglion neurons). Scale bar = 25 μm in C (applies to A-C).
Figure 6
Figure 6
There was no clear relationship between the age-related changes seen in nitrergic and sympathetic neurites. The innervation of nitrergic neurons (dark blue, NADPHd) by sympathetic axons (red-brown, TH) in the ganglia of the myenteric plexus appears, based on the amount of contacts of the varicosities on sympathetic axons with the cytoplasm and nucleus of the nitrergic neurons, to range from sparse to dense (A). In the whole mounts of the myenteric plexus from aging rats, markedly swollen sympathetic axons were found in ganglia with healthy nitrergic innervation (B) and conversely, markedly swollen nitrergic varicosities (dense, dark blue varicosities) were found in ganglia with intact sympathetic innervation (C); however, ganglia with both were never observed in our samples. Sympathetic (red-brown, TH) and nitrergic (dark blue) axons frequently traveled together in the same connectives (D). Markedly swollen sympathetic axons were found coursing through the connective with other sympathetic axons and nitrergic axons of normal diameter (E). Conversely, markedly swollen nitrergic axons in close association with sympathetic axons of normal diameter were frequently found throughout the myenteric plexus (F). Scale bars = 50 μm in C (applies to A-C); 25 μm in F (applies to D-F).
Figure 7
Figure 7
A distinct axonopathy represented by markedly swollen sympathetic axons was commonly found in the innervation of the submucosal plexus. All of the ganglia (blue neuronal counterstain, Cuprolinc Blue) within the submucosal plexus were innervated by sympathetic (brown, TH) axons (A-F). Cases of well-labeled sympathetic axons that were healthy in appearance (i.e., identical to the innervation seen at younger ages; A) were routinely found in aged tissue (B). In contrast, however, swollen sympathetic axons and terminals also became both more common and more pronounced with age (C-F). Ganglia were often observed to be innervated by sympathetic axons that were both markedly swollen and healthy looking (C,D). Scale bar = 25 μm in F (applies to A-F).
Figure 8
Figure 8
Visceral afferent innervation of the gastrointestinal tract evidenced signs of deterioration with age. A parent neurite with two markedly swollen varicosities along its length (brown, labeled using the anterograde tracer TMR-B) terminates as a putative vagal mechanoreceptor known as an “intraganglionic laminar ending” (IGLE) in the myenteric plexus (A). The insert is a higher power view of the larger of the two swollen varicosities shown in panel A; small lucent vesicles (mitochondria?) can be seen around the circumference of the bulbous varicosity. In young Fischer 344 rats (B), vagal afferents (brown, TMR-B) course into small intestinal mucosal villi as relatively un-branched neurites and then ramify into plates of free nerve endings immediately subjacent to the epithelium (chemoreceptors?). In old rats (C), however, the vagal afferents in the villi contract into smaller plates of dystrophic neurites. In both panels B and C, the gut lumen is at the top, and the epithelial wall separates the endings from the lumen. Visceral afferents (brown, CGRP), which most likely originate from the dorsal root ganglia, densely innervate the ganglia (blue neuronal counterstain, Cuprolinc Blue) of the submucosal plexus (D) and were typically “healthy” in appearance throughout the lifespan of the Fischer 344 rat; however, swollen axons and terminals were uncommonly found in the submucosal plexus of some aged rats (E). Scale bar = 50 μm in A, E (applies to D,E), C (applies to B,C); 18 μm in A (applies to Insert).
Figure 9
Figure 9
A subpopulation of neurons in the myenteric plexus are positive for alpha-synuclein which is a constituent of, and marker for, Lewy bodies commonly found in individuals diagnosed with Parkinson’s disease. In adult Fischer 344 rats, myenteric neurons and neurites positive for alpha-synuclein are “healthy” in appearance (i.e., not dilated, swollen, or dystrophic; A). In aged rats, build-up and accumulation of alpha-synuclein-positive material was found within the myenteric ganglia around alpha-synuclein-positive neurons (B). Additionally, in aged rats, some alpha-synuclein-positive axons were highly disorganized in appearance, with abnormally large and dystrophic swellings along their lengths (C). We have observed similar alpha-synuclein-positive axonopathies in the innervation of the myenteric plexus of aging mice (D). Scale bar = 50 μm in D (applies to A-D).
Figure 10
Figure 10
The growth curve of ad libitum fed, virgin male Fischer 344 rats obtained for anatomical studies from the National Institute on Aging colony maintained by Harlan Laboratory (Indianapolis, IN, USA). Mean body weight peaked at 15 months of age (449.1 g), remained stable until 18 months of age (449.8 g), and then slowly declined (397.1 g at 27 months of age). Age (n) = 3 (25), 4 (11), 5 (24), 6 (24), 12 (6), 15 (16), 16 (10), 18 (6), 24 (33), 25 (31), 26 (13), and 27 (13) months of age.

References

    1. Abalo R, Jose Rivera A, Vera G, Isabel Martin M. Ileal myenteric plexus in aged guinea-pigs: loss of structure and calretinin-immunoreactive neurones. Neurogastroenterol Motil. 2005;17:123–132.
    1. Baker DM, Santer RM. A quantitative study of the effects of age on the noradrenergic innervation of Auerbach’s plexus in the rat. Mech Ageing Dev. 1988a;42:147–158.
    1. Baker DM, Santer RM. Morphometric studies on pre- and paravertebral sympathetic neurons in the rat: changes with age. Mech Ageing Dev. 1988b;42:139–145.
    1. Bassotti G, De Giorgio R, Stanghellini V, Tonini M, Barbara G, Salvioli B, Fiorella S, Corinaldesi R. Constipation: a common problem in patients with neurological abnormalities. Ital J Gastroenterol Hepatol. 1998;30:542–548.
    1. Belai A, Cooper S, Burnstock G. Effect of age on NADPH-diaphorase-containing myenteric neurones of rat ileum and proximal colon. Cell Tissue Res. 1995;279:379–383.
    1. Bergman E, Ulfhake B. Loss of primary sensory neurons in the very old rat: neuron number estimates using the disector method and confocal optical sectioning. J Comp Neurol. 1998;396:211–222.
    1. Berthoud HR, Kressel M, Raybould HE, Neuhuber WL. Vagal sensors in the rat duodenal mucosa: distribution and structure as revealed by in vivo DiI-tracing. Anat Embryol (Berl) 1995;191:203–212.
    1. Berthoud HR, Patterson LM, Neumann F, Neuhuber WL. Distribution and structure of vagal afferent intraganglionic laminar endings (IGLEs) in the rat gastrointestinal tract. Anat Embryol (Berl) 1997;195:183–191.
    1. Black BJ, Jr, McMahan CA, Masoro EJ, Ikeno Y, Katz MS. Senescent terminal weight loss in the male F344 rat. Am J Physiol Regul Integr Comp Physiol. 2003;284:R336–342.
    1. Bloch A, Probst A, Bissig H, Adams H, Tolnay M. Alpha-synuclein pathology of the spinal and peripheral autonomic nervous system in neurologically unimpaired elderly subjects. Neuropathol Appl Neurobiol. 2006;32:284–295.
    1. Braak H, de Vos RA, Bohl J, Del Tredici K. Gastric alpha-synuclein immunoreactive inclusions in Meissner’s and Auerbach’s plexuses in cases staged for Parkinson’s disease-related brain pathology. Neurosci Lett. 2006;396:67–72.
    1. Brehmer A, Blaser B, Seitz G, Schrodl F, Neuhuber W. Pattern of lipofuscin pigmentation in nitrergic and non-nitrergic, neurofilament immunoreactive myenteric neuron types of human small intestine. Histochem Cell Biol. 2004;121:13–20.
    1. Bush TG, Savidge TC, Freeman TC, Cox HJ, Campbell EA, Mucke L, Johnson MH, Sofroniew MV. Fulminant jejuno-ileitis following ablation of enteric glia in adult transgenic mice. Cell. 1998;93:189–201.
    1. Cabarrocas J, Savidge TC, Liblau RS. Role of enteric glial cells in inflammatory bowel disease. Glia. 2003;41:81–93.
    1. Chiocchetti R, Grandis A, Bombardi C, Lucchi ML, Dal Lago DT, Bortolami R, Furness JB. Extrinsic and intrinsic sources of calcitonin gene-related peptide immunoreactivity in the lamb ileum: a morphometric and neurochemical investigation. Cell Tissue Res. 2006;323:183–196.
    1. Corns RA, Hidaka H, Santer RM. Neurocalcin-alpha immunoreactivity in the enteric nervous system of young and aged rats. Cell Calcium. 2002;31:53–58.
    1. Cowen T, Johnson RJ, Soubeyre V, Santer RM. Restricted diet rescues rat enteric motor neurones from age related cell death. Gut. 2000;47:653–660.
    1. de Souza RR, Moratelli HB, Borges N, Liberti EA. Age-induced nerve cell loss in the myenteric plexus of the small intestine in man. Gerontology. 1993;39:183–188.
    1. El-Salhy M, Sandstrom O, Holmlund F. Age-induced changes in the enteric nervous system in the mouse. Mech Ageing Dev. 1999;107:93–103.
    1. Ferri GL, Probert L, Cocchia D, Michetti F, Marangos PJ, Polak JM. Evidence for the presence of S-100 protein in the glial component of the human enteric nervous system. Nature. 1982;297:409–410.
    1. Furness JB. Types of neurons in the enteric nervous system. J Auton Nerv Syst. 2000;81:87–96.
    1. Furness JB, Costa M. The adrenergic innervation of the gastrointestinal tract. Ergeb Physiol. 1974;69:2–51.
    1. Furness JB, Robbins HL, Xiao J, Stebbing MJ, Nurgali K. Projections and chemistry of Dogiel type II neurons in the mouse colon. Cell Tissue Res. 2004;317:1–12.
    1. Gabella G. Fall in the number of myenteric neurons in aging guinea pigs. Gastroenterology. 1989;96:1487–1493.
    1. Gai WP, Blumbergs PC, Geffen LB, Blessing WW. Age-related loss of dorsal vagal neurons in Parkinson’s disease. Neurology. 1992;42:2106–2111.
    1. Ganns D, Schrodl F, Neuhuber W, Brehmer A. Investigation of general and cytoskeletal markers to estimate numbers and proportions of neurons in the human intestine. Histol Histopathol. 2006;21:41–51.
    1. Gershon MD, Rothman TP. Enteric glia. Glia. 1991;4:195–204.
    1. Green T, Dockray GJ. Calcitonin gene-related peptide and substance P in afferents to the upper gastrointestinal tract in the rat. Neurosci Lett. 1987;76:151–156.
    1. Green T, Dockray GJ. Characterization of the peptidergic afferent innervation of the stomach in the rat, mouse and guinea-pig. Neuroscience. 1988;25:181–193.
    1. Grider JR. CGRP as a transmitter in the sensory pathway mediating peristaltic reflex. Am J Physiol. 1994;266:G1139–1145.
    1. Grider JR. Neurotransmitters mediating the intestinal peristaltic reflex in the mouse. J Pharmacol Exp Ther. 2003;307:460–467.
    1. Gulley S, Sharma SK, Mansour M, Sullivan CN, Moran TH, Sayegh AI. Strain differences in myenteric neuron number and CCK(1) receptor mRNA expression may account for differences in CCK induced c-Fos activation. Brain Res. 2005;1058:109–119.
    1. Hall KE. Aging and neural control of the GI tract. II Neural control of the aging gut: can an old dog learn new tricks? Am J Physiol Gastrointest Liver Physiol. 2002;283:G827–832.
    1. Hays NP, Roberts SB. The anorexia of aging in humans. Physiol Behav. 2006;88:257–266.
    1. Hazzard DG. Relevance of the rodent model to human aging studies. Neurobiol Aging. 1991;12:645–649.
    1. Hazzard DG, Bronson RT, McClearn GE, Strong R. Selection of an appropriate animal model to study aging processes with special emphasis on the use of rat strains. J Gerontol. 1992;47:B63–64.
    1. Holst MC, Kelly JB, Powley TL. Vagal preganglionic projections to the enteric nervous system characterized with Phaseolus vulgaris-leucoagglutinin. J Comp Neurol. 1997;381:81–100.
    1. Hoff S, Wegner M, Schemann M, Ruhl A. Immunohistochemical characterization of glial cells in the human enteric nervous system (ens) Gastroenterology. 2005;128(Suppl 2):A-42.
    1. Honig LS, Rosenberg RN. Apoptosis and neurologic disease. Am J Med. 2000;108:317–330.
    1. Jellinger KA. Pathology of Parkinson’s disease. Changes other than the nigrostriatal pathway. Mol Chem Neuropathol. 1991;14:153–197.
    1. Johnson RJ, Schemann M, Santer RM, Cowen T. The effects of age on the overall population and on sub-populations of myenteric neurons in the rat small intestine. J Anat. 1998;192(Pt 4):479–488.
    1. Jost WH. Gastrointestinal motility problems in patients with Parkinson’s disease. Effects of antiparkinsonian treatment and guidelines for management. Drugs Aging. 1997;10:249–258.
    1. Lee Y, Shiotani Y, Hayashi N, Kamada T, Hillyard CJ, Girgis SI, MacIntyre I, Tohyama M. Distribution and origin of calcitonin gene-related peptide in the rat stomach and duodenum: an immunocytochemical analysis. J Neural Transm. 1987;68:1–14.
    1. Li JY, Henning Jensen P, Dahlstrom A. Differential localization of alpha-, beta- and gamma-synucleins in the rat CNS. Neuroscience. 2002;113:463–478.
    1. Meciano Filho J, Carvalho VC, de Souza RR. Nerve cell loss in the myenteric plexus of the human esophagus in relation to age: a preliminary investigation. Gerontology. 1995;41:18–21.
    1. Maeda H, Gleiser CA, Masoro EJ, Murata I, McMahan CA, Yu BP. Nutritional influences on aging of Fischer 344 rats: II. Pathology. J Gerontol. 1985;40:671–688.
    1. Majumdar AP, Jaszewski R, Dubick MA. Effect of aging on the gastrointestinal tract and the pancreas. Proc Soc Exp Biol Med. 1997;215:134–144.
    1. Mann PT, Furness JB, Southwell BR. Choline acetyltransferase immunoreactivity of putative intrinsic primary afferent neurons in the rat ileum. Cell Tissue Res. 1999;297:241–248.
    1. Masoro EJ. Use of rodents as models for the study of “normal aging”: conceptual and practical issues. Neurobiol Aging. 1991;12:639–643.
    1. Mattson MP, Magnus T. Ageing and neuronal vulnerability. Nat Rev Neurosci. 2006;7:278–294.
    1. Mattson MP, Pedersen WA, Duan W, Culmsee C, Camandola S. Cellular and molecular mechanisms underlying perturbed energy metabolism and neuronal degeneration in Alzheimer’s and Parkinson’s diseases. Ann N Y Acad Sci. 1999;893:154–175.
    1. McCulloch CR, Cooke HJ. Human alpha-calcitonin gene-related peptide influences colonic secretion by acting on myenteric neurons. Regul Pept. 1989;24:87–96.
    1. McKinney M, Jacksonville MC. Brain cholinergic vulnerability: relevance to behavior and disease. Biochem Pharmacol. 2005;70:1115–1124.
    1. Miller RA, Nadon NL. Principles of animal use for gerontological research. J Gerontol A Biol Sci Med Sci. 2000;55:B117–123.
    1. Nadon NL. Maintaining aged rodents for biogerontology research. Lab Anim (NY) 2004;33:36–41.
    1. Nakajima K, Tooyama I, Yasuhara O, Aimi Y, Kimura H. Immunohistochemical demonstration of choline acetyltransferase of a peripheral type (pChAT) in the enteric nervous system of rats. J Chem Neuroanat. 2000;18:31–40.
    1. Nasser Y, Ho W, Sharkey KA. Distribution of adrenergic receptors in the enteric nervous system of the guinea pig, mouse, and rat. J Comp Neurol. 2006;495:529–553.
    1. Neuhuber WL, Raab M, Berthoud HR, Worl J. Innervation of the mammalian esophagus. Adv Anat Embryol Cell Biol. 2006;185:1–73.
    1. Norton C. Constipation in older patients: effects on quality of life. Br J Nurs. 2006;15:188–192.
    1. O’Mahony D, O’Leary P, Quigley EM. Aging and intestinal motility: a review of factors that affect intestinal motility in the aged. Drugs Aging. 2002;19:515–527.
    1. Phillips RJ, Powley TL. Tension and stretch receptors in gastrointestinal smooth muscle: re-evaluating vagal mechanoreceptor electrophysiology. Brain Res Brain Res Rev. 2000;34:1–26.
    1. Phillips RJ, Powley TL. As the gut ages: timetables for aging of innervation vary by organ in the Fischer 344 rat. J Comp Neurol. 2001;434:358–377.
    1. Phillips RJ, Kieffer EJ, Powley TL. Aging of the myenteric plexus: neuronal loss is specific to cholinergic neurons. Auton Neurosci. 2003;106:69–83.
    1. Phillips RJ, Hargrave SL, Rhodes BS, Zopf DA, Powley TL. Quantification of neurons in the myenteric plexus: an evaluation of putative pan-neuronal markers. J Neurosci Methods. 2004a;133:99–107.
    1. Phillips RJ, Kieffer EJ, Powley TL. Loss of glia and neurons in the myenteric plexus of the aged Fischer 344 rat. Anat Embryol (Berl) 2004b;209:19–30.
    1. Phillips RJ, Pairitz JC, Powley TL. Age-related neuronal loss in the submucosal plexus of the colon of Fischer 344 rats. Neurobiol Aging. 2006a doi: 10.1016/j.neurobiolaging 2006.05.019.
    1. Phillips RJ, Rhodes BS, Powley TL. Effects of age on sympathetic innervation of the myenteric plexus and gastrointestinal smooth muscle of Fischer 344 rats. Anat Embryol (Berl) 2006b;211:673–683.
    1. Powley TL, Phillips RJ. Musings on the wanderer: what’s new in our understanding of vago-vagal reflexes? I. Morphology and topography of vagal afferents innervating the GI tract. Am J Physiol Gastrointest Liver Physiol. 2002;283:G1217–1225.
    1. Powley TL, Phillips RJ. Advances in neural tracing of vagal afferent nerves and terminals. In: Undem BJ, Weinreich D, editors. Methods and New Frontiers in Neuroscience: Advances in Vagal Afferent Neurobiology. Vol. 28. CRC Press; Boca Raton, Florida: 2005. pp. 123–145.
    1. Powley TL, Holst MC, Boyd DB, Kelly JB. Three-dimensional reconstructions of autonomic projections to the gastrointestinal tract. Microsc Res Tech. 1994;29:297–309.
    1. Ruhl A. Glial cells in the gut. Neurogastroenterol Motil. 2005;17:777–790.
    1. Ruhl A, Nasser Y, Sharkey KA. Enteric glia. Neurogastroenterol Motil. 2004;16(Suppl 1):44–49.
    1. Saffrey MJ. Ageing of the enteric nervous system. Mech Ageing Dev. 2004;125:899–906.
    1. Santer RM. Survival of the population of NADPH-diaphorase stained myenteric neurons in the small intestine of aged rats. J Auton Nerv Syst. 1994;49:115–121.
    1. Santer RM, Baker DM. Enteric neuron numbers and sizes in Auerbach’s plexus in the small and large intestine of adult and aged rats. J Auton Nerv Syst. 1988;25:59–67.
    1. Santer RM, Baker DM. Enteric system. In: Amenta F, editor. Aging of the Autonomic Nervous System. CRC Press; Baca Raton, Florida: 1993. pp. 213–225.
    1. Santer RM, Dering MA, Ranson RN, Waboso HN, Watson AH. Differential susceptibility to ageing of rat preganglionic neurones projecting to the major pelvic ganglion and of their afferent inputs. Auton Neurosci. 2002;96:73–81.
    1. Schliebs R, Arendt T. The significance of the cholinergic system in the brain during aging and in Alzheimer’s disease. J Neural Transm. 2006;113:1625–1644.
    1. Schmidt RE. Age-related sympathetic ganglionic neuropathology: human pathology and animal models. Auton Neurosci. 2002a;96:63–72.
    1. Schmidt RE. Neuropathology and pathogenesis of diabetic autonomic neuropathy. Int Rev Neurobiol. 2002b;50:257–292.
    1. Schmidt RE, Dorsey DA, Beaudet LN, Plurad SB, Parvin CA, Bruch LA. Vacuolar neuritic dystrophy in aged mouse superior cervical sympathetic ganglia is strain-specific. Brain Res. 1998;806:141–151.
    1. Schmidt RE, Chae HY, Parvin CA, Roth KA. Neuroaxonal dystrophy in aging human sympathetic ganglia. Am J Pathol. 1990;136:1327–1338.
    1. Schwaller B, Meyer M, Schiffmann S. ‘New’ functions for ‘old’ proteins: the role of the calcium-binding proteins calbindin D-28k, calretinin and parvalbumin, in cerebellar physiology. Studies with knockout mice. Cerebellum. 2002;1:241–258.
    1. Soltanpour N, Baker DM, Santer RM. Neurons and microvessels of the nodose (vagal sensory) ganglion in young adult and aged rats: morphometric and enzyme histochemical studies. Tissue Cell. 1996;28:593–602.
    1. Soltanpour N, Santer RM. Vagal nuclei in the medulla oblongata: structure and activity are maintained in aged rats. J Auton Nerv Syst. 1997;67:114–117.
    1. Spangeus A, El-Salhy M. Myenteric plexus of obese diabetic mice (an animal model of human type 2 diabetes) Histol Histopathol. 2001;16:159–165.
    1. Spangeus A, Kand M, El-Salhy M. Gastrointestinal endocrine cells in an animal model for human type 2 diabetes. Dig Dis Sci. 1999;44:979–985.
    1. Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes R, Goedert M. Alpha-synuclein in Lewy bodies. Nature. 1997;388:839–840.
    1. Sprott RL. Development of animal models of aging at the National Institute of Aging. Neurobiol Aging. 1991;12:635–638.
    1. Steinkamp M, Geerling I, Seufferlein T, von Boyen G, Egger B, Grossmann J, Ludwig L, Adler G, Reinshagen M. Glial-derived neurotrophic factor regulates apoptosis in colonic epithelial cells. Gastroenterology. 2003;124:1748–1757.
    1. Sternini C, Anderson K. Calcitonin gene-related peptide-containing neurons supplying the rat digestive system: differential distribution and expression pattern. Somatosens Mot Res. 1992;9:45–59.
    1. Sternini C, Reeve JR, Jr, Brecha N. Distribution and characterization of calcitonin gene-related peptide immunoreactivity in the digestive system of normal and capsaicin-treated rats. Gastroenterology. 1987;93:852–862.
    1. Sturrock RR. A comparison of age-related changes in neuron number in the dorsal motor nucleus of the vagus and the nucleus ambiguus of the mouse. J Anat. 1990;173:169–176.
    1. Takahashi T, Qoubaitary A, Owyang C, Wiley JW. Decreased expression of nitric oxide synthase in the colonic myenteric plexus of aged rats. Brain Res. 2000;883:15–21.
    1. Talley NJ, O’Keefe EA, Zinsmeister AR, Melton LJ., 3rd Prevalence of gastrointestinal symptoms in the elderly: a population-based study. Gastroenterology. 1992;102:895–901.
    1. Thrasivoulou C, Soubeyre V, Ridha H, Giuliani D, Giaroni C, Michael GJ, Saffrey MJ, Cowen T. Reactive oxygen species, dietary restriction and neurotrophic factors in age-related loss of myenteric neurons. Aging Cell. 2006;5:247–257.
    1. Turturro A, Witt WW, Lewis S, Hass BS, Lipman RD, Hart RW. Growth curves and survival characteristics of the animals used in the Biomarkers of Aging Program. J Gerontol A Biol Sci Med Sci. 1999;54:B492–501.
    1. Vanner S. Corelease of neuropeptides from capsaicin-sensitive afferents dilates submucosal arterioles in guinea pig ileum. Am J Physiol. 1994;267:G650–655.
    1. Wade PR. Aging and neural control of the GI tract. I. Age-related changes in the enteric nervous system. Am J Physiol Gastrointest Liver Physiol. 2002;283:G489–495.
    1. Wade PR, Cowen T. Neurodegeneration: a key factor in the ageing gut. Neurogastroenterol Motil. 2004;16(Suppl 1):19–23.
    1. Wade PR, Hornby PJ. Age-related neurodegenerative changes and how they affect the gut. Sci Aging Knowledge Environ. 2005;2005:pe8.
    1. Wakabayashi K, Takahashi H. Neuropathology of autonomic nervous system in Parkinson’s disease. Eur Neurol. 1997;38(Suppl 2):2–7.
    1. Wakabayashi K, Takahashi H, Takeda S, Ohama E, Ikuta F. Parkinson’s disease: the presence of Lewy bodies in Auerbach’s and Meissner’s plexuses. Acta Neuropathol (Berl) 1988;76:217–221.
    1. Wakabayashi K, Takahashi H, Takeda S, Ohama E, Ikuta F. Lewy bodies in the enteric nervous system in Parkinson’s disease. Arch Histol Cytol. 1989;52(Suppl):191–194.
    1. Wakabayashi K, Takahashi H, Ohama E, Ikuta F. Parkinson’s disease: an immunohistochemical study of Lewy body-containing neurons in the enteric nervous system. Acta Neuropathol (Berl) 1990;79:581–583.
    1. Wakabayashi K, Takahashi H, Ohama E, Takeda S, Ikuta F. Lewy bodies in the visceral autonomic nervous system in Parkinson’s disease. Adv Neurol. 1993;60:609–612.
    1. Wakabayashi K, Toyoshima Y, Awamori K, Anezaki T, Yoshimoto M, Tsuji S, Takahashi H. Restricted occurrence of Lewy bodies in the dorsal vagal nucleus in a patient with late-onset parkinsonism. J Neurol Sci. 1999;165:188–191.
    1. Wang ZJ, Neuhuber WL. Intraganglionic laminar endings in the rat esophagus contain purinergic P2X2 and P2X3 receptor immunoreactivity. Anat Embryol (Berl) 2003;207:363–371.
    1. Wang FB, Powley TL. Topographic inventories of vagal afferents in gastrointestinal muscle. J Comp Neurol. 2000;421:302–324.
    1. Wiley JW. Aging and neural control of the GI tract: III. Senescent enteric nervous system: lessons from extraintestinal sites and nonmammalian species. Am J Physiol Gastrointest Liver Physiol. 2002;283:G1020–1026.
    1. Wong HC, Tache Y, Lloyd KC, Yang H, Sternini C, Holzer P, Walsh JH. Monoclonal antibody to rat alpha-CGRP: production, characterization, and in vivo immunoneutralization activity. Hybridoma. 1993;12:93–106.
    1. Wu M, Van Nassauw L, Kroese AB, Adriaensen D, Timmermans JP. Myenteric nitrergic neurons along the rat esophagus: evidence for regional and strain differences in age-related changes. Histochem Cell Biol. 2003;119:395–403.
    1. Yu BP, Masoro EJ, McMahan CA. Nutritional influences on aging of Fischer 344 rats: I. Physical, metabolic, and longevity characteristics. J Gerontol. 1985;40:657–670.

Source: PubMed

3
Prenumerera