FIG 2 - uploaded by Spyridon Christodoulou
Content may be subject to copyright.
CRH and Ucn mRNA expression in HMC-1 and hCBMC cells. RT-PCR using total RNA extracted from HMC-1 and 10-wk-old hCBMC was used for detection of CRH and Ucn expression as described in Materials and Methods. A, HMC-1 and hCBMC cDNAs express a 122-bp band (arrow) that corresponds to CRH mRNA amplification product. Placental cDNA that was used as positive control confirms that the 122-bp band is the expected PCR product specific for CRH. B, HMC-1 and hCBMC cDNAs express a 145-bp band (arrow) that corresponds to Ucn mRNA amplification product. Whole uterus cDNA that was used as positive control confirms that the 145-bp band is the expected PCR product specific for Ucn. No amplification product was detected in the blank, as expected, in which the cDNA has been substituted with water serving as negative control.

CRH and Ucn mRNA expression in HMC-1 and hCBMC cells. RT-PCR using total RNA extracted from HMC-1 and 10-wk-old hCBMC was used for detection of CRH and Ucn expression as described in Materials and Methods. A, HMC-1 and hCBMC cDNAs express a 122-bp band (arrow) that corresponds to CRH mRNA amplification product. Placental cDNA that was used as positive control confirms that the 122-bp band is the expected PCR product specific for CRH. B, HMC-1 and hCBMC cDNAs express a 145-bp band (arrow) that corresponds to Ucn mRNA amplification product. Whole uterus cDNA that was used as positive control confirms that the 145-bp band is the expected PCR product specific for Ucn. No amplification product was detected in the blank, as expected, in which the cDNA has been substituted with water serving as negative control.

Source publication
Article
Full-text available
Stress activates the hypothalamic-pituitary-adrenal axis through CRH, leading to production of glucocorticoids that down-regulate immune responses. However, acute stress also has proinflammatory effects. We previously showed that restraint stress, as well as CRH and its structurally related urocortin (Ucn), could activate mast cells and trigger mas...

Contexts in source publication

Context 1
... using total RNA extracted from HMC-1 and hCBMC was used for detection of CRH and Ucn expression as described in Materials and Methods. Oligonucleotide prim- ers of previously published sequences for CRH (24) and Ucn (25) were used and yielded the expected bands of 122 bp for CRH ( Fig. 2A) and 145 bp for Ucn (Fig. 2B). Positive controls included placental cDNA and whole uterus cDNA for CRH and Ucn, respectively; they were previously shown to ex- press these PCR products using the same sets of primers. No amplification product was detected in the negative control as shown in Fig. 2 (blank) in which cDNA was substituted ...
Context 2
... using total RNA extracted from HMC-1 and hCBMC was used for detection of CRH and Ucn expression as described in Materials and Methods. Oligonucleotide prim- ers of previously published sequences for CRH (24) and Ucn (25) were used and yielded the expected bands of 122 bp for CRH ( Fig. 2A) and 145 bp for Ucn (Fig. 2B). Positive controls included placental cDNA and whole uterus cDNA for CRH and Ucn, respectively; they were previously shown to ex- press these PCR products using the same sets of primers. No amplification product was detected in the negative control as shown in Fig. 2 (blank) in which cDNA was substituted with water. PCR amplification ...
Context 3
... yielded the expected bands of 122 bp for CRH ( Fig. 2A) and 145 bp for Ucn (Fig. 2B). Positive controls included placental cDNA and whole uterus cDNA for CRH and Ucn, respectively; they were previously shown to ex- press these PCR products using the same sets of primers. No amplification product was detected in the negative control as shown in Fig. 2 (blank) in which cDNA was substituted with water. PCR amplification product using primers for -actin RT-PCR using total RNA extracted from HMC-1 and 10-wk-old hCBMC was used for detection of CRH and Ucn expression as de- scribed in Materials and Methods. A, HMC-1 and hCBMC cDNAs express a 122-bp band (arrow) that corresponds to CRH mRNA am- ...

Similar publications

Article
Full-text available
Stress induces CRH secretion that activates hypothalamic-pituitary-adrenal axis and is also abortogenic. In addition to hypothalamus, CRH and its analog urocortin (Ucn) are also secreted locally outside the brain where they activate mast cells leading to inflammation; however, the level of CRH and Ucn or mast cell mediators has not been examined in...

Citations

... Studies have found that the stimulation of MCs leads to activation of microglia, which are actually involved in ASD [66] (Figure 1). In addition, MCs are also implicated in the regulation of the hypothalamus-pituitary-adrenal (HPA) axis in the skin [168] and brain [169], as histamine, IL-6 [170] and CRH [171] can activate this axis. MCs are usually stimulated by exposure to allergens and binding of IgE to high-affinity receptors (FcERIs), whose aggregation leads to degranulation and the secretion of multiple pre-stored and newly synthesized mediators [155,172]. ...
Article
Full-text available
Autism Spectrum Disorder (ASD) is a disturbance of neurodevelopment with a complicated pathogenesis and unidentified etiology. Many children with ASD have a history of "allergic symp-toms", often in the absence of mast cell (MC)-positive tests. Activation of MCs by various stimuli may release molecules related to inflammation and neurotoxicity, contributing to the development of ASD. The aim of the present paper is to enrich the current knowledge on the relationship between MCs and ASD by discussing key molecules and immune pathways associated with MCs in the pathogenesis of autism. Cytokines, essential marker molecules for MC degranulation and therapeutic targets, are also highlighted. Understanding the relationship between ASD and the activation of MCs, as well as the involved molecules and interactions, are the main points contributing to solving the enigma. Key molecules, associated with MCs, may provide new insights to the discovery of drug targets for modeling inflammation in ASD.
... Receptor selectivity could also be attributed to modifications of different sets of amino acids at positions 30, 31, 33, 34, and 35 and/or to a change in the conformation of peptide by introducing lactam bridges at different positions of CRF [124,125]. For example, Rivier et al. have shown that the introduction of a Glu 32p -Lys 35p lactam bridge into a CRF analog with the eleven N-terminal residues deleted (creating the analog, cyclo (32)(33)(34)(35)[DPhe 12 ,Nle 21,38 ,Glu 32 ,Lys 35 ]-hCRF(12-41)) yielded a CRF 2 R-selective ligand [124]. In marked contrast, the introduction of a Glu 30p -Lys 33p lactam bridge in a CRF truncated analog created the non-selective astressin ([cyclo (30)(31)(32)(33)[DPhe (12),Nle(21),Glu(30), Lys(33),Nle (38)]hCRF((12-41))]) [125]. ...
... For example, Rivier et al. have shown that the introduction of a Glu 32p -Lys 35p lactam bridge into a CRF analog with the eleven N-terminal residues deleted (creating the analog, cyclo (32)(33)(34)(35)[DPhe 12 ,Nle 21,38 ,Glu 32 ,Lys 35 ]-hCRF(12-41)) yielded a CRF 2 R-selective ligand [124]. In marked contrast, the introduction of a Glu 30p -Lys 33p lactam bridge in a CRF truncated analog created the non-selective astressin ([cyclo (30)(31)(32)(33)[DPhe (12),Nle(21),Glu(30), Lys(33),Nle (38)]hCRF((12-41))]) [125]. Furthermore, salt bridges that are formed in the Glu 31p -Glu 34p region of CRF analogs could play a crucial role in peptide selectivity. ...
... Furthermore, salt bridges that are formed in the Glu 31p -Glu 34p region of CRF analogs could play a crucial role in peptide selectivity. The CRF analog Stressin1-A is a CRF 1 R-selective peptide with quite similar sequence as CRF (cyclo (31)(32)(33)(34)[DPhe 12 ,Nle 21,38 ,Glu 31 ,Lys 34 ]Ac-hCRF ). In Stressin1-A, residues Glu 31p and Lys 34p form a lactam bridge resulting in a 130-fold selectivity increase towards the CRF 1 R [126]. ...
Article
Full-text available
Corticotropin-releasing factor or hormone (CRF or CRH) and the urocortins regulate a plethora of physiological functions and are involved in many pathophysiological processes. CRF and urocortins belong to the family of CRF peptides (CRF family), which includes sauvagine, urotensin, and many synthetic peptide and non-peptide CRF analogs. Several of the CRF analogs have shown considerable therapeutic potential in the treatment of various diseases. The CRF peptide family act by interacting with two types of plasma membrane proteins, type 1 (CRF1R) and type 2 (CRF2R), which belong to subfamily B1 of the family B G-protein-coupled receptors (GPCRs). This work describes the structure of CRF peptides and their receptors and the activation mechanism of the latter, which is compared with that of other GPCRs. It also discusses recent structural information that rationalizes the selective binding of various ligands to the two CRF receptor types and the activation of receptors by different agonists.
... The HPA axis, also called the limbic system-hypothalamicpituitary-adrenal axis (LHPA axis), consists of the hypothalamus, the pituitary gland, and the adrenal gland, and is Correspondingly, activated mast cells also help to increase CRH levels in the brain [69]. Therefore, both CRH overactivity and GR hypersensitivity are meaningful models of HPA axis dysfunction in PTSD [45]. ...
Article
Full-text available
Post-traumatic stress disorder (PTSD), gaining increasing attention, is a multifaceted psychiatric disorder that occurs following a stressful or traumatic event or series of events. Recently, several studies showed a close relationship between PTSD and neuroinflammation. Neuroinflammation, a defense response of the nervous system, is associated with the activation of neuroimmune cells such as microglia and astrocytes and with changes in inflammatory markers. In this review, we first analyzed the relationship between neuroinflammation and PTSD: the effect of stress-derived activation of the hypothalamic–pituitary–adrenal (HPA) axis on the main immune cells in the brain and the effect of stimulated immune cells in the brain on the HPA axis. We then summarize the alteration of inflammatory markers in brain regions related to PTSD. Astrocytes are neural parenchymal cells that protect neurons by regulating the ionic microenvironment around neurons. Microglia are macrophages of the brain that coordinate the immunological response. Recent studies on these two cell types provided new insight into neuroinflammation in PTSD. These contribute to promoting comprehension of neuroinflammation, which plays a pivotal role in the pathogenesis of PTSD.
... Activation of mast cells and microglia in the hypothalamus and brain [172] could lead to cognitive dysfunction [173] and neuronal apoptosis ( Figure 1) [173]. In addition, mast cells can activate the hypothalamic-pituitary-adrenal (HPA) axis [174][175][176][177] through the release of histamine [178], IL-6 [179], and CRH [180]. It is interesting that stress has been linked to the possible priming of immune cells thus contributing to neuroinflammation in AD [181,181]. ...
... Activation of mast cells [183] and microglia in the hypothalamus [49] could cause cognitive dysfunction [173] that is also seen in patients with mastocytosis [47, 214,215] and may be related to brain abnormalities [216]. Allergic stimulation of nasal mast cells resulted in stimulation of the HPA axis [174][175][176][177], possibly via mast cell release of histamine [178], IL-6 [178,217], and CRH [180]. The influence of stress on mast cells has also been reviewed [140,218]. ...
Article
Full-text available
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19). About 45% of COVID-19 patients experience several symptoms a few months after the initial infection and develop post-acute sequelae of SARS-CoV-2 (PASC), referred to as “Long-COVID,” characterized by persistent physical and mental fatigue. However, the exact pathogenetic mechanisms affecting the brain are still not well-understood. There is increasing evidence of neurovascular inflammation in the brain. However, the precise role of the neuroinflammatory response that contributes to the disease severity of COVID-19 and long COVID pathogenesis is not clearly understood. Here, we review the reports that the SARS-CoV-2 spike protein can cause blood–brain barrier (BBB) dysfunction and damage neurons either directly, or via activation of brain mast cells and microglia and the release of various neuroinflammatory molecules. Moreover, we provide recent evidence that the novel flavanol eriodictyol is particularly suited for development as an effective treatment alone or together with oleuropein and sulforaphane (ViralProtek®), all of which have potent anti-viral and anti-inflammatory actions.
... The CRH encodes adrenocorticotropin-releasing hormone which may be a downstream gene regulated by PTH. In addition, CRH is usually produced as an autocrine or paracrine inflammatory cytokine at peripheral inflammatory sites (Karalis et al., 1991) and may play a pro-inflammatory role by activating mast cells (Kempuraj et al., 2004). On the other hand, ovarian CRH was found not only in the theca and stroma, but also in the cytoplasm of human oocytes (Mastorakos et al., 1994). ...
Article
Full-text available
The importance of thyroid-related genes has been repeatedly mentioned in the transcriptome studies of poultry with different laying performance, yet there are few systematic studies to unravel the regulatory mechanisms of the thyroid-ovary axis in the poultry egg production process. In this study, we compared the transcriptome profiles in the thyroid and ovarian stroma between high egg production (GP) and low egg production (DP) ducks, and then revealed the pathways and candidate genes involved in the process. We identified 1,114 and 733 differentially expressed genes (DEGs) in the thyroid and ovarian stroma, separately. The Gene Ontology (GO) analysis showed that a total of 504 and 189 GO terms were identified in the thyroid and ovarian stroma (P < 0.05). Three common GO terms were identified from the top 5 GO terms with the highest significant level in two tissues, including extracellular space, calcium ion binding, and integral component of plasma membrane. The enrichment analysis of the Kyoto Encyclopedia of Genes and Genomes (KEGG) showed that 15 and 14 KEGG pathways were significantly (P < 0.05) enriched in the thyroid and ovarian stroma, respectively. And, there were 8 common pathways, including neuroactive ligand-receptor interaction, calcium signaling pathway, ECM-receptor interaction, PPAR signaling pathway, melanogenesis, wnt signaling pathway, vascular smooth muscle contraction, and cytokine-cytokine receptor interaction. Notably, the neuroactive ligand-receptor interaction pathway was the most significantly enriched by the DEGs both in the thyroid and ovarian stroma. The interaction among DEGs enriched in the neuroactive ligand-receptor interaction and ECM-receptor interaction suggested that the thyroid may regulate ovarian development by these genes. Through integrated analysis of the protein-protein interaction (PPI) network and KEGG pathway maps, 9 key DEGs (PTH, THBS2, THBS4, CD36, ADIPOQ, ACSL6, PRKAA2, CRH, and PCK1) were identified, which could play crucial roles in the thyroid to regulate ovarian function and then affect egg-laying performance between GP and DP. This study serves as a basis to explore the molecular mechanism of the thyroid affecting ovarian function and egg production in female ducks and may help to identify molecular markers that can be used for duck genetic selection.
... Intestinal mast cells may be exposed to hypothalamus-derived CRH via the bloodstream or else to CRH released from peripheral nerves, but the relative contribution of each route remains unclear. Furthermore, CRH is released by mast cells themselves 131 and can act in both an autocrine and a paracrine manner to promote an inflammatory tissue environment. ...
Article
Full-text available
Irritable bowel syndrome (IBS) is a chronic functional gastrointestinal disorder that is characterized by abdominal pain and an altered defecation pattern. It affects between 5 and 20% of the general population and can seriously impact quality of life. The pathophysiology of IBS is rather complex and multifactorial including, for example, altered signalling by the gut–brain axis, dysbiosis, abnormal visceral pain signalling and intestinal immune activation. The latter has gained particular interest in recent years, with growing insight into the bidirectional communication between the nervous system and the immune system. In this Review, we detail the current evidence suggesting that immune activation contributes to the pathology seen in patients with IBS and discuss the potential mechanisms involved. Moreover, we describe how immune mediators, particularly those released by mast cells, can directly activate or sensitize pain-transmitting nerves, leading to increased pain signalling and abdominal pain. Finally, we discuss the potential of interventions targeting immune activation as a new therapeutic strategy for patients suffering from IBS. The authors of this Review discuss the current evidence suggesting that pathological immune activation contributes to irritable bowel syndrome (IBS). They explain how immune mediators can contribute to pain signalling and abdominal pain, and highlight the potential for targeting immune pathways in IBS.
... Intestinal mast cells may be exposed to hypothalamus-derived CRH via the bloodstream or else to CRH released from peripheral nerves, but the relative contribution of each route remains unclear. Furthermore, CRH is released by mast cells themselves 131 and can act in both an autocrine and a paracrine manner to promote an inflammatory tissue environment. ...
... On the one hand, MCs express receptors for neuropeptides (eg, MRGPRX2); on the other hand, MCs might also produce neuropeptides such as corticotropin-releasing factor, somatostatin, and SP. 32,61 MCs and MRGPRX2 and CST are colocalized in the lesional skin of patients with CPG, which suggests an autocrine feedback loop of autostimulation of MCs by CST. This has been reported previously for SP, which is thought to be released from both nerve endings and MCs in sickle cell anemia and also acts on MCs themselves, leading to a vicious cycle of MC activation. ...
Article
Full-text available
Background Chronic prurigo (CPG) is characterized by intensive itch and nerve-neuropeptide-mast cell interactions. The role of some neuropeptides such as cortistatin and its receptor MRGPRX2 in CPG is poorly investigated. Objectives We evaluated (i) whether cortistatin activates human skin mast cells (hsMCs), and (ii) whether cortistatin and MRGPRX2 are expressed in the skin of CPG patients and by which cells. Methods Skin prick tests and microdialysis with cortistatin were performed in six and one healthy volunteers, respectively. Degranulation of hsMCs was assessed using β-hexosaminidase and histamine release assays. Skin samples from 10 patients with CPG and 10 control subjects were stained for cortistatin, mast cells and MRGPRX2 (protein and mRNA) using immunohistochemistry, immunofluorescence and/or in situ hybridization. Flow cytometry was used to assess cortistatin in hsMCs. MRGPRX2 levels were measured in serum by ELISA. Results Cortistatin induced concentration-dependent degranulation of hsMCs in vivo and ex vivo. Skin lesions of CPG patients exhibited markedly higher numbers of cortistatin-expressing cells, cortistatin-expressing mast cells, MRGPRX2-expressing cells and MRGPRX2 mRNA-expressing cells than nonlesional skin. Mast cells were the main MRGPRX2 mRNA-expressing cells in the lesions of most CPG patients (70%). Stimulation of hsMCs with anti-IgE led to a release of cortistatin. The number of MRGPRX2-expressing cells correlated with disease severity (r=0.649, p=0.04). MRGPRX2 serum levels in CPG patients correlated with disease severity (r=0.704, p=0.023) and QoL impairment (r=0.687, p=0.028). Conclusions Cortistatin and MRGPRX2 may contribute to the pathogenesis of CPG and should be evaluated in further studies as potential biomarkers and novel therapeutic targets.
... Psychological stress could also lead to increased vascular permeability [135]. This process also contributes to the disruption of the blood-brain barrier (BBB) [149,150] via release of CRH [151] and IL-6 [152], permitting entry into the brain of viral particles, cytokines, or other toxic substances, thus further exacerbating brain inflammation. Breakdown of the BBB has been reported in the developing brain following inflammation [153]. ...
... We showed that stress stimulates mast cells via CRH [135] leading to increased dura vascular permeability, an effect that was absent in mast cell-deficient mice [230]. Moreover, mast cells can activate the hypothalamic-pituitary-adrenal (HPA) axis [142, [231][232][233] via the release of histamine [234], IL-6 [152], and CRH [151]. Moreover, neurotensin [235] and substance P (SP) [236], neuropeptides implicated in inflammation, induced CRHR-1, thus creating an autocrine loop. ...
Article
Full-text available
The prevalence of autism spectrum disorder (ASD) continues to increase, but no distinct pathogenesis or effective treatment are known yet. The presence of many comorbidities further complicates matters, making a personalized approach necessary. An increasing number of reports indicate that inflammation of the brain leads to neurodegenerative changes, especially during perinatal life, “short-circuiting the electrical system” in the amygdala that is essential for our ability to feel emotions, but also regulates fear. Inflammation of the brain can result from the stimulation of mast cells—found in all tissues including the brain—by neuropeptides, stress, toxins, and viruses such as SARS-CoV-2, leading to the activation of microglia. These resident brain defenders then release even more inflammatory molecules and stop “pruning” nerve connections, disrupting neuronal connectivity, lowering the fear threshold, and derailing the expression of emotions, as seen in ASD. Many epidemiological studies have reported a strong association between ASD and atopic dermatitis (eczema), asthma, and food allergies/intolerance, all of which involve activated mast cells. Mast cells can be triggered by allergens, neuropeptides, stress, and toxins, leading to disruption of the blood–brain barrier (BBB) and activation of microglia. Moreover, many epidemiological studies have reported a strong association between stress and atopic dermatitis (eczema) during gestation, which involves activated mast cells. Both mast cells and microglia can also be activated by SARS-CoV-2 in affected mothers during pregnancy. We showed increased expression of the proinflammatory cytokine IL-18 and its receptor, but decreased expression of the anti-inflammatory cytokine IL-38 and its receptor IL-36R, only in the amygdala of deceased children with ASD. We further showed that the natural flavonoid luteolin is a potent inhibitor of the activation of both mast cells and microglia, but also blocks SARS-CoV-2 binding to its receptor angiotensin-converting enzyme 2 (ACE2). A treatment approach should be tailored to each individual patient and should address hyperactivity/stress, allergies, or food intolerance, with the introduction of natural molecules or drugs to inhibit mast cells and microglia, such as liposomal luteolin.
... Thus, the observed increase in the expression of CRH and its receptor on the mast cells of eration of cells and stress response, in both dermis and epidermis, despite the observed decrease in expression ported in psoriasis, where the CRH serum level was found to be increased while skin expression of CRH and CRHR1 was decreased [50,51]. Since it is well established that MS results from the proliferation of transformed, constiof hyperactive MCs to the pool of neuropeptides in the skin could be expected [28,54,55]. In addition, the elevated level of ACTH in the serum of CM/SM patients and its overproduction by NMCs could result in the development of However, in our study, the mRNA levels for all chosen HPA genes, including POMC, were found to be downregulated. ...
Article
Full-text available
Introduction: Mastocytosis is a rare disease characterized by abnormal growth and accumulation of tissue mast cells (MC) in one or more organ systems and is classified as being either cutaneous mastocytosis (CM) or systemic mastocytosis (SM). In the pioneer studies of Slominski's group, a fully functional hypothalamic-pituitary-adrenal axis equivalent has been discovered in various tissues, including skin. Aim: In the present study we investigated potential involvement of hypothalamus-pituitary-adrenal (HPA) cutaneous equivalent in ongoing mastocytosis. Material and methods: The expression of HPA elements: CRH, UCN1, UCN2, UCN3, CRHR1, POMC, MC1R, MC2R and NR3C1 was assessed for their mRNA level in skin biopsies of adult patients with mastocytosis and healthy donors (n = 16 and 19, respectively), while CRH, UCN1, CRHR1, ACTH and MC1R were selected for immunostaining assay (n = 13 and 7, respectively). The expression of CRH receptor 1 (CRHR1) isomers was investigated by RT-PCR. The ELISA was used for detection of cortisol, CRH, UCN and ACTH in the serum. Results: The decrease in the expression of HPA element of skin equivalent was observed on both mRNA and protein levels, however quantification of immunohistochemical data was impeded due to melanin in epidermis. Furthermore, we observed infiltration of dermis with HPA elements overexpressing mononuclear cells, which is in the agreement with an in vitro study showing a high expression of HPA elements by mast cells. Conclusions: Taken together, it was confirmed that the expression elements of HPA was modulated in mastocytosis, thus the potential involvement of general and local stress responses in its pathogenesis should be postulated and further investigated.