Cellular pathogenesis of gastroparesis





Abbreviations



Ano-1


anoctamin-1


BH4


tetrahydrobiopterin


BMP-2


bone morphogenetic protein-2


BMPR


BMP receptor


ChAT


choline acetyltransferase


CSF-1


colony stimulating fator-1


CX3CR1


CX3C chemokine receptor 1


DG


diabetic gastroparesis


DM


diabetes mellitus


ENS


enteric nervous system


GI


gastrointestinal


HO-1


heme oxygenase 1


ICC


interstitial cells of Cajal


IG


idiopathic gastroparesis


IGF-1


insulin like growth factor 1


IL-1β


interleukin 1β


iNOS


inhibitory nitric oxide synthase


IP3


inositol triphosphate


MHCII


major histocompatibility complex II


nNOS


neuronal nitric oxide synthase


NO


nitric oxide


NOD


non-obese diabetic


NPY


neuropeptide Y


op/op


osteopetrotic mouse


PKC


protein kinase C


SMCs


smooth muscle cells


SP


substance P


STZ


streptozotocin


TGF-β


transforming growth factor β


TNF-α


tumor necrosis factor α


VIP


vasoactive intestinal polypeptide.



Introduction


Delayed emptying of gastric contents in the absence of any mechanical obstruction of the gastric outlet is pathognomonic for gastroparesis . As opposed to normal GI function, delay in gastric emptying may be due to antral dysmotility , impaired fundic accommodation , and/or pyloric dysfunction . The cardinal symptoms of this chronic disorder include early satiety, postprandial fullness, abdominal pain, nausea and vomiting . These result in significant impairment of quality of life and high healthcare expenditures .


Although, diabetes has been considered to be the leading cause of gastroparesis, a long-term follow-up study accounted for only 29% patients harboring diabetic gastroparesis (DG), while gastroparesis acquired after a surgery (post-surgical gastroparesis) constituted 13% of the cases . A majority of patients (>50%) presented with an unknown primary cause and were categorized as idiopathic gastroparesis (IG) . However, a recently published cross-sectional study in a large and diverse US population, only 28% of patients represented IG whereas, 72% patients exhibited DG (16% type 1 diabetes mellitus and 55% type 2 diabetes mellitus) . These differences may be attributed to the diagnostic code-based approach for the identification of gastroparesis in the recent compared to gastric scintigraphy confirmed gastroparesis in the prior study. Other commonly observed etiologies of gastroparesis include connective tissue disorders, Parkinson’s disease, or medication-induced .


Gastric emptying is considered to be abnormal/delayed when >60% of administered contents are retained at 2 hours and/or >10% at 4 hours on a solid meal gastric scintigraphy . The severity of symptoms associated with gastroparesis is assessed using the Gastroparesis Cardinal Symptom Index (GCSI) which is based on three subscales: early satiety/postprandial fullness, nausea/vomiting, and bloating . Even though the abdominal pain is not included in the list of GCSI subscales, pain is observed in >80% patients with gastroparesis . Interestingly, a very recent report by Hasler et al. indicates the use of opioids is common in both DG and IG patients and associates with worse cardinal symptoms of gastroparesis and increased hospitalization .


Decades of research in the field of gastroparesis using complimentary animal models and human biopsy specimens, has significantly helped in understanding the cellular and molecular changes in disease pathogenesis. In this chapter, we aim to highlight the research advancements in understanding the gastroparesis pathophysiology and strategies for minimally invasive procurement and use of human full thickness biopsy specimens for future research.


Complexity of gastric emptying


Emptying of gastric contents into the small bowel is a highly complex physiological process involving the synergetic interactions of the neuro-muscular apparatus of the stomach. Normally, emptying of gastric contents occurs by coordinated and appropriate movements of the fundus, body, antrum, and pylorus. These movements occur by the critical roles played by extrinsic nerve innervations, excitatory and inhibitory components of the enteric (intrinsic) nervous system (ENS), interstitial cells of Cajal (ICC), fibroblast-like cells and smooth muscle cells (SMC) . These different cell types are located deep within the muscular and myenteric layers of the stomach. In addition, different parts of the stomach and the feedback loop between the stomach and small intestine also play important roles in mediating gastric emptying . For instance, the fundus and the upper body portions of the stomach generate a basal pressure by producing low frequency, sustained contractions. Similarly, the lower body portion and the antrum of the stomach generate contractions by producing strong peristaltic waves. These tonic contractions assist in pushing gastric contents towards the pylorus while, the digestible solids larger than 2 mm are bounced backwards. This process of mixing and churning occurring due to phasic contractions completes the process of homogenizing the ingested food. The digested food is then emptied into the small bowel as the increased amplitude of peristaltic waves obliterates the lumen of the pylorus, delivering the gastric contents in small volumes of liquid and chyme .


Gastric motility is controlled by a set of neural and hormonal signals. GI tract stands distinctive by having its own intrinsic neuroglial circuits (the ENS). Excitatory and inhibitory neurons are categorized based on the expression pattern of specific neurotransmitters. For example, excitatory neurons express choline acetyltransferase (ChAT) , substance P (SP) and neurokinins (NKA, NKB & neuropeptide Y) whereas, the inhibitory neurons express neuronal nitric oxide synthase (nNOS) – which generates nitric oxide (NO) , and vasoactive intestinal polypeptide (VIP) . In addition, specialized functions like fundic accommodation and pyloric relaxation are mediated by inhibitory nitrergic nerves .


ICC are the mesenchymal cells located within the musculature of the GI tract and are well known for their intrinsic ‘pacemaker’ activity . They play a vital role in generating bioelectrical slow wave potential in a spatiotemporal manner , driving rhythmic contraction of SMC . The syncytial network of ICC is known to mediate sequential excitatory and inhibitory neuro transmissions and also does play a role in mechanotransduction – the critical components of normal gastrointestinal motility . Populations of ICC are recognized typically by their surface expression of receptor tyrosine kinase (Kit) and anoctamin-1 (Ano-1) – a calcium activated chloride channel . NO has been shown to be a survival factor for ICC in mouse models and NO also promotes ICC proliferation in vitro , indicating the association of nitrergic neurons with ICC. Changes in the expression and splicing of Ano-1 have been associated with symptoms of DG .


Fibroblast-like cells are a second population of interstitial cells located in the tunica muscularis of the GI tract and impart additional regulatory control in the ENS . These cells are shown to express cell surface receptor tyrosine kinase – platelet-derived growth factor alpha (PDGFRα) and not Kit , hence are popularly known as PDGFRα + cells. Ultrastructure studies have shown fibroblast-like cells exhibiting features of fibroblasts and also have connections with circular and longitudinal SMC via gap junctions . In addition, these fibroblast-like cells are also shown to be closely associated with intramuscular ICC and enteric nerve fibers . The ICC, fibroblast-like cells and SMC together form an electrically coupled unit termed SIP syncytium – an important unit for normal GI motility .


Smooth muscle of GI tract is required to crush, grind and mix the intraluminal contents, liquefying them to form chyme. The chyme is then pushed through the pyloric canal into the duodenum establishing gastric emptying. This process of gastric emptying is accomplished by phasic contractions of SMC . The SMC themselves do not originate slow waves – a rate limiting step of gastric emptying. Instead, the SMC are linked to the neighboring ICC via gap junctions creating a syncytium , which drives coordinated contraction and relaxation of SMC by producing bioelectrical slow waves . These phasic contractions and relaxations of the SMC are also regulated by NO from nitrergic neurons and the mechanism is mediated by NO-sensitive guanylyl cyclase (NO-GC) .


Macrophages play a pivotal role in maintaining tissue homeostasis by responding to internal and external stimuli. Resident macrophages are highly heterogeneous in nature and the tissue microenvironment guides the macrophages to attain a specific phenotype. Each of these phenotypes is known to have variable gene expression profiles . Variability is often seen in the expression pattern of well-established macrophage markers between mouse and humans ; for example, iNOS, Arginase-1 and Ym1 are predominantly expressed by mouse but not by human macrophages . Indeed, variability in macrophage gene expression profiles can also be observed when compared not only between the species but, also within a tissue from the same host. For example, the gene expression profile of macrophages in the GI tract varies from mucosa, submucosa, muscularis and serosa in mouse . Typically, muscularis macrophages of mouse predominantly express CX3CR1 (hi), MHCII (hi) and CD11c (lo), whereas, lamina propria macrophages express CD11c (hi) . Likewise, high levels of CD11b and CD14 are expressed by human muscularis macrophages but, a reduced level of their expression is seen in mucosal macrophages . Phenotypically, macrophages located in the circular muscle and longitudinal muscle are of bipolar shape, whereas, myenteric macrophages are stellate shaped .


In the context of GI tract, the studies concerning macrophages are mostly focused on the lamina propria macrophages. Wherein, these macrophages play defensive roles by acting against harmful gut microbial invasion, engulfing and clearing luminal antigens that occasionally breach mucosal barrier. Recently, muscularis macrophages are gaining attention of the researchers as they stand unique transcriptionally and also morphologically from the neighboring macrophage counterparts of mucosa and lamina propria . There is a close and likely symbiotic interaction between the macrophages and ENS in the myenteric plexus . Macrophages secrete a protein belonging to TGF-β superfamily called bone morphogenetic protein-2 (BMP-2) . Enteric neurons express BMP receptor (BMPR) and binding of BMP-2 to BMPR leads to oligomerization of type I and type II serine kinases, followed by phosphorylation and nuclear translocation of SMAD proteins . Activation of BMPR promotes nitrergic enteric neuronal differentiation and helps in regulating gastrointestinal motility . And also, BMP-2 has been shown to promote nNOS expression in myenteric neurons through SMAD1 phosphorylation . In turn, enteric neurons produce colony stimulating fator-1 (CSF-1) – a growth factor essential for the differentiation and maintenance of muscularis macrophages and this is evident from the osteopetrotic ( op/op ) mice, which harbor a mutation in their CSF-1 gene, also lack muscularis macrophages and display a disordered ENS architecture . In addition, ICC has been identified as a non-neuronal source of CSF-1 in mice lacking enteric neurons (RetKO mice) . A recent report by Cipriani et al. shows an increased number of neurons in Csf1 op/op mice expressing an overlap between NOS1 and ChAT–a chemical code overlap that is known to reduce as the neurons mature . Thus, cross-talk between ENS, ICC, fibroblasts like cells, SMC and immune cells play an indispensable role in maintaining normal gastric physiology.


Pathogenesis of gastroparesis is associated with cellular alterations


Research so far in delineating the pathophysiological alterations associated with gastroparesis has been successful in finding several key anomalies associated with some of the important cell types ( Fig. 6.1 ). In addition, very recently, transcriptomic and proteomic analyses have been successful in identifying the molecular changes occurring during the pathogenesis of gastroparesis. Here we describe some of the important studies that have impacted our understanding of the pathological and molecular basis of gastroparesis.




Figure 6.1


Approximate percentage of gastroparetic patients exhibiting cellular abnormalities.

The pie chart represents the approximate percentage of gastroparetic patients exhibiting altered number of represented cell types. ~50% of gastroparetic patients exhibit loss of ICC as determined by reduced Kit immunoreactivity while, at least 80% of patients display signs of ICC degeneration as depicted by ultrastructural studies showing vacuoles, lamellar bodies and swollen mitochondria. The second most common abnormality observed in about 40–50% of patients is the dysregulation of immune cells. The abnormalities associated with enteric nerves are represented only in a very small subset of patient population varying from 0% to 15% depending on the type of enteric nerves (excitatory, inhibitory, extrinsic innervation) and there were signs of degeneration on ultrastructural studies. Trichrome staining of tissue sections shows mild fibrosis to be present in ≤10% of patient population. Abnormalities associated with SMC are found to be in ~20% of patients as determined by reduced Smoothelin immunoreactivity. ~30% of patients do not show any marked abnormalities in these immunochemical assessments when compared with the controls.

This data is based on a previously published report by Grover et al. Grover M, Farrugia G, Lurken MS, et al. Cellular changes in diabetic and idiopathic gastroparesis. Gastroenterology 2011;140(5):1575–85, e8.


Alterations in ENS associated with gastroparesis


Paul Kassander stands first to describe diabetic gastroparesis . In his initial report involving case studies of six patients with diabetes, asymptomatic gastric retention of meal was observed on X-ray findings with no abnormalities in small intestine. This disturbance in the expulsion of gastric contents was attributed to the vagus nerve, and the radiographic results resembled to those of gastric hypotonia after vagotomy . However, two decades later, a case study by Norman Soler involving two diabetic patients showed gastroparesis even in the absence of extrinsic diabetic neuropathy . Around the same time, a report by Shellito & Warshaw described a syndrome of recurrent intermittent gastric atony in five non-diabetic patients experiencing the symptoms of gastroparesis , which gave rise to the entity of IG . A study by Yoshida et al., on 16 patients with long-standing diabetes involving five patients with gastroparesis, did not show any changes in the number and appearance of neurons or axons, and also no abnormalities associated with vagus nerve were observed in the myenteric plexus . However, the major drawback of this study was the use of only conventional histological staining techniques for visualizing myenteric plexus and the vagus nerve.


The initial studies were focused only on the extrinsic nervous system however; subsequent research exhibited defects in intrinsic nervous system in animal models of diabetic gastroparesis. Using streptozotocin (STZ) induced rat model of diabetes (without delayed gastric emptying), Belai et al., have observed a consistent increase in VIP-like immunoreactivity in nerve fibers, and more frequent intensely stained cell bodies in the myenteric plexus and circular muscle layer of both ileum and proximal colon ( P <0.001). Whereas, compared to controls, no significant changes were observed in the SP innervation in myenteric plexus and circular muscle of both ileum and colon of diabetic rats . Similarly, increased VIP-like immunoreactivity was also observed in STZ induced diabetic rats with delayed gastric emptying . Interestingly, continuous insulin administration in an in vitro setting reversed these effects, suggesting that, the availability of a steady level of insulin influences some of the changes occurring during early stages of diabetes . In a similar study by Watkins et al. , non-obese diabetic (NOD) mice and STZ induced diabetic mice showed defects mimicking the mice harboring a deletion of the nNOS by showing defects in gastric emptying and pyloric muscle relaxation . Insulin administration or intraperitoneal injection of phosphodiesterase inhibitor (Sildenafil) to these mice resulted in an increase in the levels of NO and also restored gastric emptying . Interestingly, chronic depletion of NO derived from nNOS resulted in delayed emptying of solids and liquids from the stomach but, depletion of NO derived from eNOS – a constitutive enzyme produced by endothelial cells, had no effect , suggesting the importance of nNOS and NO signaling in pyloric relaxation. Chronic estrogen deficiency in Follicle-stimulating hormone receptor knockout (FORKO) mouse has been reported to affect the function of tetrahydrobiopterin – a co-factor essentially required for nNOS enzyme activity, and thus contributing for the development of gastroparesis . In a report by Choi et al. delayed gastric emptying in NOD mouse model of DG was restored by exogenous administration of IL-10 and this effect was independent of nNOS activity . Further research aiming to delineate the pathways associated with regulation of nNOS expression would help in considering it as a molecular target to treat gastroparesis.


Alterations in expression of enteric nerve markers have also been observed in biopsy specimens from gastroparetic patients. In a case report of a 38-year-old type 1 DM patient, full thickness jejunal biopsy specimen showed a significant decrease in nerve fiber content (PGP9.5) in the circular muscle when compared with the control tissues from six subjects ( P <0.05) . In addition, the nNOS, VIP, and PACAP were also found to be decreased significantly . Yet another case report of 32 year-old IG patient showed similar findings where, there was 69% reduction in the number of nerve cell bodies (PGP9.5) in the stomach corpus sections . A report by Iwasaki et al. involving male gastric cancer patients with and without type 2 DM showed significant reduction in the expression of nNOS (p<0.01) and SP ( P <0.01) in the stomach antrum of patients with type 2 DM but not in patients without type 2 DM, indicating that the expression levels of nNOS & SP are associated with the pathogenesis of DG . In a case report of two gastroparetic patients with type 1 DM, Pasricha et al. have shown reduced expression of nNOS, nerve fibers and myenteric neurons (PGP9.5) in the full thickness gastric biopsy of a patient with long standing, poorly controlled diabetes. Although, both the patients were presented with similar symptoms, the other patient with well controlled diabetes over a short period showed no significant abnormalities, when compared with the controls . These findings suggest the association of histopathological alterations in the ENS with poorly controlled diabetes. In a study by Chandrasekharan et al. enhanced apoptosis and loss of peripherin, nNOS, neuropeptide Y (NPY) and ChAT neurons were observed along with a significant reduction in the ganglion size in the colons of patients with diabetes . In another study by Harberson et al. , aiming to histologically evaluate the pathologies in full-thickness antral biopsies from patients with refractory gastroparesis (14 each of type 1 DM & idiopathic gastroparesis), showed a substantial reduction in the numbers of ganglia, NOS + and NOS nerve cells when compared to controls . However, in the largest study by the NIDDK GpCRC defining the cellular changes associated with gastroparesis using gastric biopsies obtained from control, DG and IG patients (20 patients in each group), only a 14–18% decrease in the expression of PGP9.5 in gastroparesis as compared to the controls . Although, nNOS was found to be decreased in 20% of patients with DG and 40% of patients with IG, no statistically significant difference in nNOS was observed when compared between the groups . Likewise, 20% of patients with DG and 15% of patients with IG displayed a reduction in the expression of VIP immunolabelling but, no statistical difference was found between the three groups . Immunolabeling for SP did not show any statistically significant difference either, when compared between the groups . The results of this largest study to date, involving more number of subjects in each group clearly indicates the changes in the number of enteric nerves and neurons observed may only be present in a subset of patients with gastroparesis. The changes may also be associated with the etiology, disease duration and site of assessment in the stomach. Remarkably, Faussone-Pellegrini et al. using transmission electron microscopy (TEM) of full-thickness gastric biopsies showed evidence for cellular damage to the nerves, even in patients with no apparent histological changes on light microscopy . The observed alterations include, loss of synaptic vesicles, thickened basal lamina and fibrosis around nerves. Interestingly, IG patients showed a more severe damage as compared to DG .


Loss of ICC associated with gastroparesis


In the context of diabetic gastroparesis, the very first study using an animal model of diabetes (NOD/LtJ mice), depletion of gastric ICC networks resulted in a significant delay in emptying of the gastric contents . ICC loss in antral muscle tissues by 50% was associated with reduced Kit-like immunoreactivity. In addition, lack of association between ICC and enteric nerve terminals was also noted . This report suggested the alteration in pacemaker ICC content being the basis of DG. A subsequent study by Wang et al. employing STZ induced rat model of diabetes showed reduction in the density of ICC in antrum and the effect was also accompanied with the loss of synaptic connections and delayed gastric emptying .


Reports concerning the loss of ICC in human biopsy specimens of jejunum , colon and antrum were reported in DG and IG patients. A report by Horváth et al. using organotypic cultures of murine gastric muscle showed that, the loss of ICC was not associated with diabetes induced hyperglycemia; instead, the effect was due to reduced insulin and insulin like growth factor 1 (IGF-1) signaling . In a study by the NIDDK GpCRC , more than 25% of ICC loss was documented in 50% of patients irrespective of DG or IG as shown by reduction in Kit expression. Loss was ICC was similar in both DG and IG accounting for ~50% mean ICC count when compared with controls. Ultrastructural studies using TEM displayed damage associated alterations in ICC structure in the tissues of both DG and IG patients. The observed alterations include apoptotic features, intracytoplasmatic vacuoles, swollen mitochondria and extended rough endoplasmic reticulum. A physical separation of ICC from other ICC and nerves was also observed when compared with the controls. In a similar study focusing on the ultrastructural differences between DG and IG, both intramuscular and myenteric ICC had similar changes . A study on investigating the associations between the status of ICC and electrogastrogram (EGG) parameters on gastric emptying and its symptoms in a large cohort of 41 patients with gastroparesis by Lin et al., showed, severe loss of ICCs in 36% of patients while, rest of the patients had no visible ICCs identified . EGG recordings showed significantly reduced normal slow waves in patients with severe loss of ICC both under fasting and fed states and these results correlated significantly with the loss of ICC . In a similar study by GpCRC, >60% loss of ICC was documented in the enrolled patients . All these studies clearly suggest that, loss of ICC as a hallmark feature of gastroparesis in humans.


Changes in fibroblast-like cells associated with gastroparesis


In a report aiming to determine the morphology and distribution of fibroblast-like cells (PDGFRα + ) in human gastric body biopsies obtained from control subjects (n=5) showed PDGFRα + cells having slender cell bodies with long and thin processes . And these PDGFRα + cells were more predominantly located in longitudinal muscle layers than circular muscle layer. In addition, in the myenteric plexus, these PDGFRα + cells displayed morphology with smaller and round cell bodies possessing 3–4 processes, forming a network around the ganglia . Although, fibroblast-like cells are a identified to be a sub-population of interstitial cells , no overlapping cell bodies of fibroblast-like cells with the ICC were observed despite, being located in close proximity to each other . In an attempt to identify their alterations in patients with DG (n=10) and IG (n=10), no changes in morphology, distribution or the number of cells were observed in patients with gastroparesis when compared with the controls . Interestingly, in a study involving a large cohort of patients with DM (n=32; without gastroparesis) and control subjects (n=26), significant number of fibroblast-like cells were reduced in the fundus and the body of stomach but, not in antrum of patients with DM when compared with the control group, as identified by PDGFRα + immunoreactivity . Hence, this creates an avenue for an in-depth analysis of changes in the population of PDGFRα + cells during DG and IG, and also to delineate their actual role in gastroparesis.


Alterations in smooth muscle cells associated with gastroparesis


A study on the effects of transmural nerve stimulation, acetylcholine, norepinephrine, or K-free solution on the electrical responses of gastric smooth muscles by Xue et al., , the STZ induced diabetic rats showed functional impairments associated with neuromuscular transmission. This abnormality was associated with reduced adrenoceptor (norepinephrine) sensitivity and Na-K pump activity whereas, increased sensitivity to muscarinic receptor (acetylcholine) was observed . These results suggests that, the alterations in SMC leading to the development of delayed gastric emptying is a result of diabetes .


Forkhead transcription factors are shown to be involved in smooth muscle proliferation, migration and apoptosis . Knocking out the forkhead transcription factors – FOXF1 and FOXF2, in SMC of adult mice has been shown to cause delayed gastric emptying . In addition, the authors have also observed a significant reduction in the expression levels of these transcription factors in full-thickness gastric biopsies obtained from DG and IG patients, suggesting the role of FOXF1 and FOXF2 in normal gastric function .


Smooth muscle degeneration and fibrosis with eosinophilic inclusion bodies has been reported by Ejskjaer et al. in four DG patients with a symptom of intractable vomiting and this indicates the presence of gastromyopathy. In a case study of two patients, one exhibiting poorly controlled long standing diabetes and the other with well controlled diabetes over a short duration reported by Pasricha et al. showed, extensive fibrosis in the myenteric plexus, circular and longitudinal muscle layers of full thickness gastric biopsy obtained from the patient with poorly controlled diabetes over long duration but, not in patient with well controlled diabetes over a short duration . This clearly indicates the SMC changes associated with the status and clinical course of DM. Unfortunately, in a large cohort study by NIDDK GpCRC involving both DG and IG patients, no such fibrosis was observed in the full-thickness stomach body specimens when compared with matched controls . In the same study, 23% of patients exhibited reduced expression of smoothelin-A – a specific marker of SMC. However, study using TEM, failed to show any marked abnormalities in SMC in the tissue specimens of both DG and IG patients .


Alterations in macrophage milieu associated with gastroparesis


Determining the role of macrophage dysregulation in the pathogenesis of gastroparesis is an active area of research. This is because; macrophages are prone to acquiring different phenotypes in response to subtle changes in the tissue microenvironment. Various stimuli and cytokines are known to polarize macrophages into M1 (pro-inflammatory) or M2 (anti-inflammatory) phenotype . A number of disease states have been associated with an alteration in the macrophage milieu . M1 and M2 spectrum of macrophages are differentiated based on the expression profiles of different pro- and anti-inflammatory markers expressed on their surface. For example, M1 macrophages typically express the classical pro-inflammatory markers TNF-α, IL-1β and iNOS. On the other hand, M2 macrophages predominantly express the CD206 or mannose receptor, Arginase-1 and TGF-β .


In the context of gastroparesis, the CD206+ macrophages are gaining much importance as the studies show alterations in the overall population of these anti-inflammatory macrophages within the muscular layers of the GI tract. CD206+ macrophages express heme oxygnase 1 (HO-1) – an enzyme well known for its role in cellular defense mechanism against oxidative stress . In a study on NOD mouse model by Choi et al., during early diabetes, CD206+ muscularis macrophages showed increased expression of HO-1 and these levels were consistently maintained in the animals that were resistant to develop delayed gastric emptying . However, loss of CD206+ macrophages was observed in mice developing delayed gastric emptying, and this loss was also associated with loss of HO-1 expression in addition to the loss of Kit (ICC) . Due to the reduced expression of HO-1, an increase in the oxidative stress was also observed and this effect was reversed upon treating these mice either with hemin or IL-10 . This provides evidence that HO-1 expressing CD206+ macrophages may be able to prevent or treat DG.


Despite being severely diabetic, CSF1 op/op mice that lack muscularis macrophages do not develop delayed gastric emptying . Supplementing these mice with an intraperitoneal injection of CSF1 restores the muscularis macrophages, rendering them susceptible to acquire gastroparesis upon induction of diabetes . Interestingly, subjecting the cultured ICC with M1 macrophage conditioned medium reduced the ICC cell counts by 41% while, no such effect was observed when the ICC were treated with conditioned medium from M2 macrophages . In an attempt to identify possible paracrine mediators, immunoblot assay showed, 12 pro-inflammatory cytokine and chemokine markers in M1 macrophage conditioned medium . This suggests a potential role for paracrine signaling in macrophage driven Cajalopathy. Further studies delineating the molecular mechanisms associated with macrophage-ICC interactions are needed.


Concerning human studies, an initial report by Harberson et al. on full thickness antrum biopsies from patients with DG (n=14) and IG (n=14), showed mild lymphocytic infiltrate positive for CD45 and CD3 in the myenteric plexus . Further studies by NIDDK GpCRC on full thickness gastric biopsies of the antrum and body obtained from DG and IG patients showed a significant reduction in the total number of CD206+ macrophages in the sections of antrum but not the body of the stomach. However, total population of immune cells (CD45+) was unchanged . Remarkably, the loss of CD206+ macrophages correlated significantly with the loss of ICC in circular muscle of gastric antrum , suggesting the importance of CD206+ macrophages in maintaining ICC population.


Transcriptomic signatures in gastroparesis


To date, most of the studies focused on understanding the pathological basis of gastroparesis have reported valuable information on the cellular abnormalities associated with the disease pathogenesis. However, the reports detailing changes occurring at the molecular level are limited. A recent study by the NIDDK GpCRC reported a list of differentially expressing genes (log 2 fold difference | ≥ 2|, FDR< 5%) by deep transcriptomic sequencing of RNA isolated from full-thickness biopsies of gastric body from patients with both DG and IG. It shows, 111 and 181 genes expressing differentially in DG and IG respectively, of which sixty-five genes were commonly expressed between DG and IG. As determined by Ingenuity Pathway Analysis (IPA), these differentially expressing genes were belonging to the top five canonical pathways associated with immune signaling . In addition, immune profile analysis using CIBERSORT showed the genes associated with the pro-inflammatory M1 macrophages to be enriched in the tissues isolated from IG patients, when compared with the controls. A recent report by Herring et al., describes the increased expression of mRNAs encoding contractile proteins (MYH11, MYLK1 and ACTA2) in smooth muscle tissues of IG patients as compared to lean controls . And the authors have also observed a reduced expression of PDGFRα and also its ligand – PDGFβ . However, in our previous work on gastric body specimens, the numbers and distribution of PDGFRα + cells was not altered in gastroparesis .


Very recently, we reported proteomic expression profiles in full-thickness gastric antrum biopsies of DG and IG patients as determined by aptamer-based SomaLogic tissue scan that is known to quantitatively identify 1300 human proteins . We found, 73 proteins differentially expressing in DG and 133 proteins differentially expressing in IG. Of these differentially expressing proteins, 40 proteins were common between both DG and IG. The results of IPA analysis showed “Role of Macrophages, Fibroblasts and Endothelial Cells” as the most statistically significant altered pathway and, this complements the results of our deep transcriptomic profiling study . In summary, both RNA- and protein-based molecular profiling results complement the histological studies in suggesting the involvement of immune based dysregulation in ENS injury leading to gastroparesis.


Correlation of pathological anomalies with clinical symptoms of gastroparesis


A large cohort study showed ICC loss to positively correlating with the gastric retention of solids in patients with DG . In another study, ICC counts in the myenteric plexus were found to be associated with delayed gastric emptying . In a study by Forster et al. , severe tachygastria was observed in gastroparesis patients exhibiting depleted or no ICC but, not in patients with relatively normal number of ICC . These patients were also found to be associated with reduced postprandial rhythmic function. In addition, the total symptom score of gastroparesis was also found to be significantly higher in patients with depleted or total loss of ICC at baseline and also after three months of using gastric electrical stimulator, when compared with patients having adequate ICC . Similar results were reported subsequently in a larger cohort by the same group . The study from the NIDDK GpCRC, also found correlation between the abundance of myenteric immune cells with nausea and the overall symptom severity . Use of next generation sequencing technology followed by targeted validation can help in identifying molecules that can serve as biomarkers and allow target discovery for treatment of gastroparesis.


Future directions for the effective use of gastric biopsies in research


Research works on full thickness gastric biopsies so far have helped us tremendously in understanding the pathological changes associated with human gastroparesis. Most of the reports have found several similarities in cellular alterations between DG and IG. The most important shift in our understanding of the pathology associated with gastroparesis is the identification of the involvement of innate immune system, which drives ENS and ICC damage . However, the biggest challenge to target the macrophages as the culprits is a lack of atlas describing the subtypes of macrophages present in the gastric muscle layers and this challenge holds good for other important components like the ENS and ICC. Although, much of our understanding on the disease pathology is from the animal models of diabetic gastroparesis, the hurdle is that, the expression pattern of genotypic markers in mouse and human immune cells are not same . Future studies aiming to identify the cellular and molecular atlases of immune cell, ENS and ICC from healthy subjects would help in identifying the key factors altered in patients with gastroparesis. From a diagnostic standpoint, we also need to further develop minimally invasive methods for procuring gastric full-thickness tissue. In a recent study by Rajan et al. , adequate tissue was procured (~1 cm) by performing a no-hole gastric body biopsy using an over-the-scope clip and which is sufficient enough for ENS assessment. Endoscopic ultrasound based approaches have also been tried but the yield for full-thickness assessment of the ENS is low . From a therapeutic standpoint, strategies to influence macrophage polarization to acquire anti-inflammatory phenotype or to restore gastric emptying may act as treatment options for gastroparesis. Paradigms aiming to identify exact pathological alterations in gastroparesis patients followed by targeted therapeutic approach would help in finding a cure rather than, focusing solely on symptom alleviation.



References

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Feb 4, 2021 | Posted by in GASTROENTEROLOGY | Comments Off on Cellular pathogenesis of gastroparesis

Full access? Get Clinical Tree

Get Clinical Tree app for offline access