Alterations in Large Intestine Innervation during CRC: Comparison
Please note this is a comparison between Version 2 by Rita Xu and Version 1 by Aldona Kasprzak.

Colorectal cancer (CRC), classified as third most prevalent cancer worldwide, remains to be a clinical and research challenge. It is estimated that ~50% of CRC patients die from distant metastases. While, since the 1970s, the consensus is that tumors lack innervation, there are clear evidences of connections between the nervous system and cancer. CRC, as a tumor, possesses nerve fibres from peripheral nervous system (PNS), as part of its microenvironment, as well as axons from both branches of autonomic NS and primary sensory neurons. The structural-functional changes in enteric nervous system innervation of the tumor are important. A connection is suggested between nervous system dysfunctions and a range of neurotransmitters (Nts) (including neuropeptides, NPs), neurotrophins (Ntt) and their receptors in CRC liver metastasis (LM) development. More research is needed to understand the exact mechanisms of communication between the neurons and tumor cells.

  • colorectal cancer, neuropeptides, innervation
 

There is evidence of a direct link between the nervous system and cancer through synapses, non-synapse contacts, or humoral modulation, which contribute to two-way communication and influence cancer metastases. Similar to nerve structures, cancer cells produce Nts/NPs and their receptors [1][2]. In CRC patients, structural and functional changes of large intestine innervation can be observed. Interestingly, in the CRC LM, contrarily to the healthy liver, a lack of autonomic perivascular Protein Gene Product 9.5. (PGP9.5)- and Neuropeptide Y (NPY)-immunoreactive nerves can be observed [3].

There is evidence of a direct link between the nervous system and cancer through synapses, non-synapse contacts, or humoral modulation, which contribute to two-way communication and influence cancer metastases. Similar to nerve structures, cancer cells produce Nts/NPs and their receptors [1,2]. In CRC patients, structural and functional changes of large intestine innervation can be observed. Interestingly, in the CRC LM, contrarily to the healthy liver, a lack of autonomic perivascular Protein Gene Product 9.5. (PGP9.5)- and Neuropeptide Y (NPY)-immunoreactive nerves can be observed [3].

Morphological Changes in Innervation and Neuropeptide Panel in CRC

Structural changes in CRC innervation mostly concern ENS, occurring most commonly in the form of gradual reduction, leading to the total destruction of the nerve structures [4,5,6]. Atrophy of submucosal and MPs within close proximity to the tumor occurs [6,7]. Among the NPs, a decrease in CGRP+ neurons and nerves was observed in both plexus types in the transitional zone between cancerous area and unchanged tissue. The decrease also concerned SP+ nerve fibers in all intramural plexuses [4] and NPY-ergic neurons, as well as the density of nerve fibers in both plexuses [5]. Interestingly, there were no significant quantitative differences in the numbers of SP+, SM+, Vasoactive Intestinal Polypeptide (VIP)-ergic and Pituitary Adenylate Cyclase-activating Peptide (PACAP)-ergic neurons, as well as SM+ nerve fibers in cancer, compared with healthy regions [4,5]. Lower numbers of VIP-ergic and PACAP-ergic nerve fibers were observed in submucosal and MPs than in control sections [5]. In turn, an unchanged density of galanin (Gal)-positive nerve fibers was observed, while the percentage of Gal+ neurons was higher in CRC (46%) than the healthy intestine (35%) [7]. A reduction in the size of Gal+ MPs in the vicinity of the tumor was also reported, as compared with unchanged tissue [6]. Mean Gal content in tumor was lower (9.38 ng/g) than in the morphologically unaltered intestine (12.27 ng/g) [7]. Ultrastructural changes in CRC patients include an increase in the mass of extracellular matrix (ECM), occurrence of myelin-like structures, numerous apoptotic cells, as well as the presence of mast and plasma cells in MPs in the tumor surrounding area [8].

Structural changes in CRC innervation mostly concern ENS, occurring most commonly in the form of gradual reduction, leading to the total destruction of the nerve structures [4][5][6]. Atrophy of submucosal and MPs within close proximity to the tumor occurs [6][7]. Among the NPs, a decrease in CGRP+ neurons and nerves was observed in both plexus types in the transitional zone between cancerous area and unchanged tissue. The decrease also concerned SP+ nerve fibers in all intramural plexuses [4] and NPY-ergic neurons, as well as the density of nerve fibers in both plexuses [5]. Interestingly, there were no significant quantitative differences in the numbers of SP+, SM+, Vasoactive Intestinal Polypeptide (VIP)-ergic and Pituitary Adenylate Cyclase-activating Peptide (PACAP)-ergic neurons, as well as SM+ nerve fibers in cancer, compared with healthy regions [4][5]. Lower numbers of VIP-ergic and PACAP-ergic nerve fibers were observed in submucosal and MPs than in control sections [5]. In turn, an unchanged density of galanin (Gal)-positive nerve fibers was observed, while the percentage of Gal+ neurons was higher in CRC (46%) than the healthy intestine (35%) [7]. A reduction in the size of Gal+ MPs in the vicinity of the tumor was also reported, as compared with unchanged tissue [6]. Mean Gal content in tumor was lower (9.38 ng/g) than in the morphologically unaltered intestine (12.27 ng/g) [7]. Ultrastructural changes in CRC patients include an increase in the mass of extracellular matrix (ECM), occurrence of myelin-like structures, numerous apoptotic cells, as well as the presence of mast and plasma cells in MPs in the tumor surrounding area [8].

The Perineural Invasion (PNI) in CRC

There are ongoing studies on the involvement of perineural invasion (PNI) of cancer cells in the modulation of tumorigenesis [1,2,9]. PNI might be an underestimated mode of metastasis spread, acting in combination with lymphatic and vascular invasion [9,10,11,12], as well as on its own [9]. In CRC, ~16–40% of the patients exhibited PNI characterized by neoplastic invasion of nerves, with altered molecular determinants of the process [9]. It is debated if nerve ablation can delay/inhibit the formation of tumors and/or reduce metastaticity [2].

There are ongoing studies on the involvement of perineural invasion (PNI) of cancer cells in the modulation of tumorigenesis [1][2][9]. PNI might be an underestimated mode of metastasis spread, acting in combination with lymphatic and vascular invasion [9][10][11][12], as well as on its own [9]. In CRC, ~16–40% of the patients exhibited PNI characterized by neoplastic invasion of nerves, with altered molecular determinants of the process [9]. It is debated if nerve ablation can delay/inhibit the formation of tumors and/or reduce metastaticity [2].

 

The markers closely associated with PNI include Nts (e.g., ACh, NE and their receptors: AChR, NE-R), Ntt (NGF, Brain-Derived Neurotrophic Factor (BDNF), Glial Cell line-derived Neurotrophic Factor (GDNF) and their receptors: Neurotrophic Receptor Tropomyosin-related Kinase B (TrKB)), as well as typical NPs (e.g., SP, Gal, NPY/CGRP) [2][9]. PNI is a multistep process, during which a major role is played by the so-called perineural niche, together with numerous signaling molecules (including NPs/NP-Rs) [9]. There is a lack of detailed studies on the mechanisms of nerve–tumor interactions in PNI in CRC, as most of the research concerns different types of cancer (e.g., prostate and gastric cancers, pancreatic ductal adenocarcinoma) [2][9]. However, a prognostic role of PNI was proven in CRC. Defining PNI as a presence of cancer cells inside the nerve sheath, or at least 33% of the nerve periphery surrounded by cancer cells in CRC, shorter 5-year survival rates were observed compared with negative PNI. Additionally, positive correlations between PNI and lymph node metastases, tumor grade depth of invasion, clinical-stage, vessel invasion and tumor growth pattern were observed [13]. PNI was indicated as an independent bad prognostic factor in CRC [9][11][13], affecting both overall survival (OS) [13], cancer-specific survival (CSS) and disease-free survival (DFS) [12]. PNI is also an independent factor in CRC recurrence, points to a more malignant tumor phenotype and, as an important parameter, should be considered in pathological classification of CRC [9]. Recently, a large cohort study indicated that PNI is also more commonly observed in colitis-associated (90%) than in sporadic CRC [12].

The markers closely associated with PNI include Nts (e.g., ACh, NE and their receptors: AChR, NE-R), Ntt (NGF, Brain-Derived Neurotrophic Factor (BDNF), Glial Cell line-derived Neurotrophic Factor (GDNF) and their receptors: Neurotrophic Receptor Tropomyosin-related Kinase B (TrKB)), as well as typical NPs (e.g., SP, Gal, NPY/CGRP) [2,9]. PNI is a multistep process, during which a major role is played by the so-called perineural niche, together with numerous signaling molecules (including NPs/NP-Rs) [9]. There is a lack of detailed studies on the mechanisms of nerve–tumor interactions in PNI in CRC, as most of the research concerns different types of cancer (e.g., prostate and gastric cancers, pancreatic ductal adenocarcinoma) [2,9]. However, a prognostic role of PNI was proven in CRC. Defining PNI as a presence of cancer cells inside the nerve sheath, or at least 33% of the nerve periphery surrounded by cancer cells in CRC, shorter 5-year survival rates were observed compared with negative PNI. Additionally, positive correlations between PNI and lymph node metastases, tumor grade depth of invasion, clinical-stage, vessel invasion and tumor growth pattern were observed [13]. PNI was indicated as an independent bad prognostic factor in CRC [9,11,13], affecting both overall survival (OS) [13], cancer-specific survival (CSS) and disease-free survival (DFS) [12]. PNI is also an independent factor in CRC recurrence, points to a more malignant tumor phenotype and, as an important parameter, should be considered in pathological classification of CRC [9]. Recently, a large cohort study indicated that PNI is also more commonly observed in colitis-associated (90%) than in sporadic CRC [12].

Functional Innervation Disorders in CRC

Functional disorders in CRC and colitis concern mostly changes in interactions between large intestine innervation and the immune system [6]. Interestingly, such alterations occur on the level of NP-Rs, present on most of the immune cells. Anti-inflammatory roles of VIP and CGRP, as well as pro-inflammatory effects of serotonin and NPY, are also often underlined. In turn, SP has both anti- and pro-inflammatory effects. Apart from neurons, the production of Nts: ACh, choline acetyltransferase (ChAT), acetylcholinesterase, and both muscarinic/cholinergic and nicotinic ACh receptors, was also demonstrated on numerous immune cells (e.g., T and B cells, dendritic cells, macrophages), potentially extending the anti-inflammatory action of ACh in the large intestine [14].

Functional disorders in CRC and colitis concern mostly changes in interactions between large intestine innervation and the immune system [6]. Interestingly, such alterations occur on the level of NP-Rs, present on most of the immune cells. Anti-inflammatory roles of VIP and CGRP, as well as pro-inflammatory effects of serotonin and NPY, are also often underlined. In turn, SP has both anti- and pro-inflammatory effects. Apart from neurons, the production of Nts: ACh, choline acetyltransferase (ChAT), acetylcholinesterase, and both muscarinic/cholinergic and nicotinic ACh receptors, was also demonstrated on numerous immune cells (e.g., T and B cells, dendritic cells, macrophages), potentially extending the anti-inflammatory action of ACh in the large intestine [14].

 
Influence of some ANS Nts (e.g., NE, ACh) and their co-transmitters (e.g., NPY, adenosine triphosphate and/or VIP) on the proliferation of Intestinal Epithelial Stem Cells (IESCs) is also often underlined, despite little knowledge on the mechanisms of that process [15]. It seems that regulation of IESCs proliferation occurs with the participation of both branches of ANS, independently of ENS. Due to more numerous IESCs in the deeper regions of intestinal crypts, SNS and Nts can regulate the proliferation of these cells. ACh is also a PNS mediator, initiating a signaling cascade resulting in suppression of cyclin D1 and a downstream decrease in cell proliferation [15]. The role of ANS–IESC interactions is also considered in the context of differentiation of some kinds of colon cancers from somatic SCs, as well as maintenance of IESC-like properties under neoplastic conditions [15,16].
 
 1. Li, S.; Sun, Y.; Gao, D. Role of the nervous system in cancer metastasis. Oncol. Lett. 2013, 5, 1101–1111. [Google Scholar] [CrossRef] [PubMed]
2. Saloman, J.L.; Albers, K.M.; Rhim, A.D.; Davis, B.M. Can stopping nerves, stop cancer? Trends Neurosci. 2016, 39, 880–889. [Google Scholar] [CrossRef] [PubMed]
3. Ashraf, S.; Crowe, R.; Loizidou, M.C.; Turmaine, M.; Taylor, I.; Burnstock, G. The absence of autonomic perivascular nerves in human colorectal liver metastases. Br. J. Cancer 1996, 73, 349–359. [Google Scholar] [CrossRef] [PubMed]
4. Godlewski, J.; Kaleczyc, J. Somatostatin, substance P and calcitonin gene-related peptide-positive intramural nerve structures of the human large intestine affected by carcinoma. Folia Histochem. Cytobiol. 2010, 48, 475–483. [Google Scholar] [CrossRef] [PubMed]
5. Godlewski, J.; Łakomy, I.M. Changes in vasoactive intestinal peptide, pituitary adenylate cyclase-activating polypeptide and neuropeptide Y-ergic structures of the enteric nervous system in the carcinoma of the human large intestine. Folia Histochem. Cytobiol. 2010, 48, 208–216. [Google Scholar] [CrossRef]
6. Kwiatkowski, P.; Godlewski, J.; Kieżun, J.; Kraziński, B.E.; Kmieć, Z. Colorectal cancer patients exhibit increased levels of galanin in serum and colon tissues. Oncol. Lett. 2016, 12, 3323–3329. [Google Scholar] [CrossRef]
7. Godlewski, J.; Pidsudko, Z. Characteristic of galaninergic components of the enteric nervous system in the cancer invasion of human large intestine. Ann. Anat. 2012, 194, 368–372. [Google Scholar] [CrossRef]
8. Zauszkiewicz-Pawlak, A.; Godlewski, J.; Kwiatkowski, P.; Kmiec, Z. Ultrastructural characteristics of myenteric plexus in patients with colorectal Cancer. Folia Histochem. Cytobiol. 2017, 55, 6–10. [Google Scholar] [CrossRef]
9. Chen, S.H.; Zhang, B.Y.; Zhou, B.; Zhu, C.Z.; Sun, L.Q.; Feng, Y.J. Perineural invasion of cancer: A complex crosstalk between cells and molecules in the perineural niche. Am. J. Cancer Res. 2019, 9, 1–21. [Google Scholar]
10. Liebig, C.; Ayala, G.; Wilks, J.A.; Berger, D.H.; Albo, D. Perineural invasion in cancer: A review of the literature. Cancer 2009, 115, 3379–3391. [Google Scholar] [CrossRef]
11. Liebig, C.; Ayala, G.; Wilks, J.; Verstovsek, G.; Liu, H.; Agarwal, N.; Berger, D.H.; Albo, D. Perineural invasion is an independent predictor of outcome in colorectal cancer. J. Clin. Oncol. 2009, 27, 5131–5137. [Google Scholar] [CrossRef]
12. Nozawa, H.; Hata, K.; Ushiku, T.; Kawai, K.; Tanaka, T.; Shuno, Y.; Nishikawa, T.; Sasaki, K.; Emoto, S.; Kaneko, M.; et al. Accelerated perineural invasion in colitis-associated cancer: A retrospective cohort study. Medicine 2019, 98, e17570. [Google Scholar] [CrossRef] [PubMed]
13. Zhou, Y.; Wang, H.; Gong, H.; Cao, M.; Zhang, G.; Wang, Y. Clinical significance of perineural invasion in stages II and III colorectal Cancer. Pathol. Res. Pract. 2015, 211, 839–844. [Google Scholar] [CrossRef]
14. Fujii, T.; Mashimo, M.; Moriwaki, Y.; Misawa, H.; Ono, S.; Horiguchi, K.; Kawashima, K. Expression and function of the cholinergic system in immune cells. Front. Immunol. 2017, 8, 1085. [Google Scholar] [CrossRef] [PubMed]
15. Davis, E.A.; Dailey, M.J. A direct effect of the autonomic nervous system on somatic stem cell proliferation? Am. J. Physiol. Regul. Integr. Comp. Physiol. 2019, 316, R1–R5. [Google Scholar] [CrossRef] [PubMed]
16. Merlos-Suárez, A.; Barriga, F.M.; Jung, P.; Iglesias, M.; Céspedes, M.V.; Rossell, D.; Sevillano, M.; Hernando-Momblona, X.; da Silva-Diz, V.; Muñoz, P.; et al. The intestinal stem cell signature identifies colorectal cancer stem cells and predicts disease relapse. Cell Stem Cell. 2011, 8, 511–524. [Google Scholar] [CrossRef]

Influence of some ANS Nts (e.g., NE, ACh) and their co-transmitters (e.g., NPY, adenosine triphosphate and/or VIP) on the proliferation of Intestinal Epithelial Stem Cells (IESCs) is also often underlined, despite little knowledge on the mechanisms of that process [15]. It seems that regulation of IESCs proliferation occurs with the participation of both branches of ANS, independently of ENS. Due to more numerous IESCs in the deeper regions of intestinal crypts, SNS and Nts can regulate the proliferation of these cells. ACh is also a PNS mediator, initiating a signaling cascade resulting in suppression of cyclin D1 and a downstream decrease in cell proliferation [15]. The role of ANS–IESC interactions is also considered in the context of differentiation of some kinds of colon cancers from somatic SCs, as well as maintenance of IESC-like properties under neoplastic conditions [15][16].

References

  1. Sha Li; Yanlai Sun; Dongwei Gao; Role of the nervous system in cancer metastasis. Oncology Letters 2013, 5, 1101-1111, 10.3892/ol.2013.1168.
  2. Saloman, J.L.; Albers, K.M.; Rhim, A.D.; Davis, B.M. Can stopping nerves, stop cancer? Trends Neurosci. 2016, 39, 880–889.
  3. S Ashraf; R Crowe; Mc Loizidou; M Turmaine; I Taylor; G Burnstock; The absence of autonomic perivascular nerves in human colorectal liver metastases. British Journal of Cancer 1996, 73, 349-359, 10.1038/bjc.1996.60.
  4. Janusz Godlewski; Jerzy Kaleczyc; Somatostatin, substance P and calcitonin gene-related peptide-positive intramural nerve structures of the human large intestine affected by carcinoma. Folia Histochemica et Cytobiologica 2010, 48, 475–483, 10.2478/v10042-010-0079-y.
  5. Janusz Godlewski; Ireneusz Mirosław Łakomy; Changes in vasoactive intestinal peptide, pituitary adenylate cyclase-activating polypeptide and neuropeptide Y-ergic structures of the enteric nervous system in the carcinoma of the human large intestine. Folia Histochemica et Cytobiologica 2010, 48, 208–216, 10.2478/v10042-010-0052-9.
  6. Przemysław Kwiatkowski; Janusz Godlewski; Jacek Kiezun; Bartlomiej Krazinski; Zbigniew Kmieć; Colorectal cancer patients exhibit increased levels of galanin in serum and colon tissues. Oncology Letters 2016, 12, 3323-3329, 10.3892/ol.2016.5037.
  7. Janusz Godlewski; Zenon Pidsudko; Characteristic of galaninergic components of the enteric nervous system in the cancer invasion of human large intestine. Annals of Anatomy - Anatomischer Anzeiger 2012, 194, 368-372, 10.1016/j.aanat.2011.11.009.
  8. Agata Zauszkiewicz-Pawlak; Janusz Godlewski; Przemysław Kwiatkowski; Zbigniew Kmieć; Ultrastructural characteristics of myenteric plexus in patients with colorectal cancer. Folia Histochemica et Cytobiologica 2017, 55, 6-10, 10.5603/fhc.a2017.0003.
  9. Shu-Hai Chen; Bing-Yuan Zhang; Bin Zhou; Cheng-Zhan Zhu; Le-Qi Sun; Yu-Jie Feng; Perineural invasion of cancer: a complex crosstalk between cells and molecules in the perineural niche. American journal of cancer research 2019, 9, 1-21.
  10. Liebig, C.; Ayala, G.; Wilks, J.A.; Berger, D.H.; Albo, D; Perineural invasion in cancer: A review of the literature. Cancer 2009, 115, 3379–3391.
  11. Catherine Liebig; Gustavo Ayala; Jonathan Wilks; Gordana Verstovsek; Hao Liu; Neeti Agarwal; David H. Berger; Daniel Albo; Perineural Invasion Is an Independent Predictor of Outcome in Colorectal Cancer. Journal of Clinical Oncology 2009, 27, 5131-5137, 10.1200/jco.2009.22.4949.
  12. Nozawa, H.; Hata, K.; Ushiku, T.; Kawai, K.; Tanaka, T.; Shuno, Y.; Nishikawa, T.; Sasaki, K.; Emoto, S.; Kaneko, M.; et al. Accelerated perineural invasion in colitis-associated cancer: A retrospective cohort study. Medicine 2019, 98, e17570.
  13. Yi Zhou; Hongyan Wang; Huilin Gong; Meng Cao; Guanjun Zhang; Yi-Li Wang; Clinical significance of perineural invasion in stages II and III colorectal cancer. Pathology - Research and Practice 2015, 211, 839-844, 10.1016/j.prp.2015.09.001.
  14. Takeshi Fujii; Masato Mashimo; Yasuhiro Moriwaki; Hidemi Misawa; Shiro Ono; Kazuhide Horiguchi; Koichiro Kawashima; Expression and Function of the Cholinergic System in Immune Cells. Frontiers in Immunology 2017, 8, 1085, 10.3389/fimmu.2017.01085.
  15. Davis, E.A.; Dailey, M.J. A direct effect of the autonomic nervous system on somatic stem cell proliferation? Am. J. Physiol. Regul. Integr. Comp. Physiol. 2019, 316, R1–R5.
  16. Anna Merlos-Suárez; Francisco M. Barriga; Peter Jung; Mar Iglesias; María Virtudes Céspedes; David Rossell; Marta Sevillano; Xavier Hernando-Momblona; Victoria Da Silva-Diz; Purificación Muñoz; Hans Clevers; Elena Sancho; Ramon Mangues; Eduard Batlle; The Intestinal Stem Cell Signature Identifies Colorectal Cancer Stem Cells and Predicts Disease Relapse. Cell Stem Cell 2011, 8, 511-524, 10.1016/j.stem.2011.02.020.
More