Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Adv Exp Med Biol ; 1383: 229-241, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36587162

RESUMO

Years ago gastrointestinal motility was thought to be due to interactions between enteric nerves and smooth muscle cells (SMCs) in the tunica muscularis. Thus, regulatory mechanisms controlling motility were either myogenic or neurogenic. Now we know that populations of interstitial cells, c-Kit+ (interstitial cells of Cajal or ICC), and PDGFRα+ cells (formerly "fibroblast-like" cells) are electrically coupled to SMCs, forming the SIP syncytium. Pacemaker and neurotransduction functions are provided by interstitial cells through Ca2+ release from the endoplasmic reticulum (ER) and activation of Ca2+-activated ion channels in the plasma membrane (PM). ICC express Ca2+-activated Cl- channels encoded by Ano1. When activated, Ano1 channels produce inward current and, therefore, depolarizing or excitatory effects in the SIP syncytium. PDGFRα+ cells express Ca2+-activated K+ channels encoded by Kcnn3. These channels generate outward current when activated and hyperpolarizing or membrane-stabilizing effects in the SIP syncytium. Inputs from enteric and sympathetic neurons regulate Ca2+ transients in ICC and PDGFRα+ cells, and currents activated in these cells conduct to SMCs and regulate contractile behaviors. ICC also serve as pacemakers, generating slow waves that are the electrophysiological basis for gastric peristalsis and intestinal segmentation. Pacemaker types of ICC express voltage-dependent Ca2+ conductances that organize Ca2+ transients, and therefore Ano1 channel openings, into clusters that define the amplitude and duration of slow waves. Ca2+ handling mechanisms are at the heart of interstitial cell function, yet little is known about what happens to Ca2+ dynamics in these cells in GI motility disorders.


Assuntos
Células Intersticiais de Cajal , Células Intersticiais de Cajal/fisiologia , Receptor alfa de Fator de Crescimento Derivado de Plaquetas , Músculo Liso/fisiologia , Trato Gastrointestinal/fisiologia , Intestino Delgado/metabolismo
2.
Int J Mol Sci ; 23(9)2022 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-35563399

RESUMO

Metalloendopeptidase ADAM-Like Decysin 1 (ADAMDEC1) is an anti-inflammatory peptidase that is almost exclusively expressed in the gastrointestinal (GI) tract. We have recently found abundant and selective expression of Adamdec1 in colonic mucosal PDGFRα+ cells. However, the cellular origin for this gene expression is controversial as it is also known to be expressed in intestinal macrophages. We found that Adamdec1 mRNAs were selectively expressed in colonic mucosal subepithelial PDGFRα+ cells. ADAMDEC1 protein was mainly released from PDGFRα+ cells and accumulated in the mucosal layer lamina propria space near the epithelial basement membrane. PDGFRα+ cells significantly overexpressed Adamdec1 mRNAs and protein in DSS-induced colitis mice. Adamdec1 was predominantly expressed in CD45- PDGFRα+ cells in DSS-induced colitis mice, with only minimal expression in CD45+ CD64+ macrophages. Additionally, overexpression of both ADAMDEC1 mRNA and protein was consistently observed in PDGFRα+ cells, but not in CD64+ macrophages found in human colonic mucosal tissue affected by Crohn's disease. In summary, PDGFRα+ cells selectively express ADAMDEC1, which is localized to the colon mucosa layer. ADAMDEC1 expression significantly increases in DSS-induced colitis affected mice and Crohn's disease affected human tissue, suggesting that this gene can serve as a diagnostic and/or therapeutic target for intestinal inflammation and Crohn's disease.


Assuntos
Proteínas ADAM , Colite , Doença de Crohn , Doenças Inflamatórias Intestinais , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Animais , Biomarcadores , Colite/induzido quimicamente , Colite/genética , Colite/metabolismo , Colo/citologia , Colo/metabolismo , Doença de Crohn/metabolismo , Doenças Inflamatórias Intestinais/diagnóstico , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/metabolismo , Mucosa Intestinal/metabolismo , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo
3.
FASEB J ; 34(4): 5563-5577, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32086857

RESUMO

Transcriptome data revealed α1 adrenoceptors (ARs) expression in platelet-derived growth factor receptor α+ cells (PDGFRα+ cells) in murine colonic musculature. The role of PDGFRα+ cells in sympathetic neural regulation of murine colonic motility was investigated. Norepinephrine (NE), via α1A ARs, activated a small conductance Ca2+ -activated K+ (SK) conductance, evoked outward currents and hyperpolarized PDGFRα+ cells (the α1A AR-SK channel signal pathway). α1 AR agonists increased intracellular Ca2+ transients in PDGFRα+ cells and inhibited spontaneous phasic contractions (SPCs) of colonic muscle through activation of a SK conductance. Sympathetic nerve stimulation inhibited both contractions of distal colon and propulsive contractions represented by the colonic migrating motor complexes (CMMCs) via the α1A AR-SK channel signal pathway. Postsynaptic signaling through α1A ARs in PDGFRα+ cells is a novel mechanism that conveys part of stress responses in the colon. PDGFRα+ cells appear to be a primary effector of sympathetic neural regulation of murine colonic motility.


Assuntos
Colo/fisiologia , Músculo Liso/fisiologia , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/fisiologia , Receptores Adrenérgicos alfa 1/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Sistema Nervoso Simpático/fisiologia , Potenciais Sinápticos , Trifosfato de Adenosina , Animais , Cálcio/metabolismo , Colo/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso/citologia , Transdução de Sinais , Sistema Nervoso Simpático/citologia
4.
J Physiol ; 598(20): 4509-4521, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32735345

RESUMO

KEY POINTS: ß-Nicotinamide adenine dinucleotide (ß-NAD) is a key inhibitory neurotransmitter in the colon. The neuroeffector junction in the gut consists of enteric motor neurons and SIP syncytium, including smooth muscle cells (SMCs), interstitial cells of Cajal (ICC), and cells expressing platelet-derived growth factor receptor α (PDGFRα+ cells). Measuring metabolism of 1,N6 -etheno-NAD (eNAD) in colonic tunica muscularis and in SMCs, ICC and PDGFRα+ cells with HPLC-FLD, we report that (1) in tissues, eNAD is degraded to eADP-ribose, eAMP and e-adenosine (eADO) by CD38, ENPP1 and NT5E, (2) with SMCs and PDGFRα+ cells, eNAD is metabolized to eADO by ENPP1 and NT5E, (3) eNAD is not metabolized by ICC, (4) NT5E is expressed chiefly by SMCs and moderately by PDGFRα+ cells, (5) SIP cells are not the primary location of CD38. These data argue that the duration and strength of purinergic neurotransmission can be modulated by targeting multiple enzymes with specialized cellular distribution in the colon. ABSTRACT: Prior studies suggest that ß-nicotinamide adenine dinucleotide (ß-NAD) is an important inhibitory motor neurotransmitter in the enteric nervous system. Metabolism of ß-NAD at the neuroeffector junction (NEJ) is likely to be necessary for terminating inhibitory neurotransmission and may also produce bioactive metabolites. The enteric NEJ consists of enteric neurons and postjunctional cells of the SIP syncytium, including smooth muscle cells (SMCs), interstitial cells of Cajal (ICC), and cells expressing platelet-derived growth factor receptor α (PDGFRα+ cells). We examined possible specialized functions of the NEJ in ß-NAD metabolism by determining the degradation of 1,N6 -etheno-NAD (eNAD) in colonic tunica muscularis of wild-type, Cd38-/- , Nt5e-/- , Enpp1-/- and Cd38-/- /Nt5e-/- mice and in SIP cells from mice expressing cell-specific fluorescent reporters purified by fluorescence activated cell sorting (FACS). We measured eNAD and its metabolites eADP-ribose (eADPR), eAMP and e-adenosine (eADO) from tissues and sorted SIP cells using liquid chromatography. eNAD exposed to colonic muscularis of wild-type mice produced eADPR, eAMP and eADO. CD38 mediated the conversion of eNAD to eADPR, whereas ENPP1 mediated degradation of eNAD and eADPR to eAMP. NT5E (aka CD73) was the primary enzyme forming eADO from eAMP. PDGFRα+ cells and SMCs were involved in production of eADO from eNAD, and ICC were not involved in extracellular metabolism of eNAD. CD38 mediated the eNAD metabolism in whole tissues, but CD38 did not appear to be functionally expressed by SMCs or ICC. NT5E was expressed in SMCs > PDGFRα+ cells. Our data show that extracellular metabolism of ß-NAD in the colon is mediated by multiple enzymes with cell-specific expression.


Assuntos
Células Intersticiais de Cajal , NAD , Animais , Colo , Camundongos , Músculo Liso , Neurotransmissores
5.
Proc Natl Acad Sci U S A ; 111(44): 15821-6, 2014 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-25341729

RESUMO

Enteric purinergic motor neurotransmission, acting through P2Y1 receptors (P2Y1R), mediates inhibitory neural control of the intestines. Recent studies have shown that NAD(+) and ADP ribose better meet criteria for enteric inhibitory neurotransmitters in colon than ATP or ADP. Here we report that human and murine colon muscles also release uridine adenosine tetraphosphate (Up4A) spontaneously and upon stimulation of enteric neurons. Release of Up4A was reduced by tetrodotoxin, suggesting that at least a portion of Up4A is of neural origin. Up4A caused relaxation (human and murine colons) and hyperpolarization (murine colon) that was blocked by the P2Y1R antagonist, MRS 2500, and by apamin, an inhibitor of Ca(2+)-activated small-conductance K(+) (SK) channels. Up4A responses were greatly reduced or absent in colons of P2ry1(-/-) mice. Up4A induced P2Y1R-SK-channel-mediated hyperpolarization in isolated PDGFRα(+) cells, which are postjunctional targets for purinergic neurotransmission. Up4A caused MRS 2500-sensitive Ca(2+) transients in human 1321N1 astrocytoma cells expressing human P2Y1R. Up4A was more potent than ATP, ADP, NAD(+), or ADP ribose in colonic muscles. In murine distal colon Up4A elicited transient P2Y1R-mediated relaxation followed by a suramin-sensitive contraction. HPLC analysis of Up4A degradation suggests that exogenous Up4A first forms UMP and ATP in the human colon and UDP and ADP in the murine colon. Adenosine then is generated by extracellular catabolism of ATP and ADP. However, the relaxation and hyperpolarization responses to Up4A are not mediated by its metabolites. This study shows that Up4A is a potent native agonist for P2Y1R and SK-channel activation in human and mouse colon.


Assuntos
Colo/metabolismo , Fosfatos de Dinucleosídeos/farmacologia , Motilidade Gastrointestinal/efeitos dos fármacos , Agonistas do Receptor Purinérgico P2Y/farmacologia , Receptores Purinérgicos P2Y1/metabolismo , Difosfato de Adenosina/farmacologia , Animais , Antineoplásicos/farmacologia , Colo/inervação , Nucleotídeos de Desoxiadenina/farmacologia , Humanos , Camundongos , Camundongos Knockout , Relaxamento Muscular/efeitos dos fármacos , Músculo Liso/metabolismo , Receptores Purinérgicos P2Y1/genética , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Suramina/farmacologia , Difosfato de Uridina/farmacologia
6.
Am J Physiol Gastrointest Liver Physiol ; 311(6): G1037-G1046, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27742704

RESUMO

Interstitial cells of Cajal (ICC) generate electrical slow waves by coordinated openings of ANO1 channels, a Ca2+-activated Cl- (CaCC) conductance. Efflux of Cl- during slow waves must be significant, as there is high current density during slow-wave currents and slow waves are of sufficient magnitude to depolarize the syncytium of smooth muscle cells and PDGFRα+ cells to which they are electrically coupled. We investigated how the driving force for Cl- current is maintained in ICC. We found robust expression of Slc12a2 (which encodes an Na+-K+-Cl- cotransporter, NKCC1) and immunohistochemical confirmation that NKCC1 is expressed in ICC. With the use of the gramicidin permeabilized-patch technique, which is reported to not disturb [Cl-]i, the reversal potential for spontaneous transient inward currents (ESTICs) was -10.5 mV. This value corresponds to the peak of slow waves when they are recorded directly from ICC in situ. Inhibition of NKCC1 with bumetanide shifted ESTICs to more negative potentials within a few minutes and reduced pacemaker activity. Bumetanide had no direct effects on ANO1 or CaV3.2 channels expressed in HEK293 cells or L-type Ca2+ currents. Reducing extracellular Cl- to 10 mM shifted ESTICs to positive potentials as predicted by the Nernst equation. The relatively rapid shift in ESTICs when NKCC1 was blocked suggests that significant changes in the transmembrane Cl- gradient occur during the slow-wave cycle, possibly within microdomains formed between endoplasmic reticulum and the plasma membrane in ICC. Recovery of Cl- via NKCC1 might have additional consequences on shaping the waveforms of slow waves via Na+ entry into microdomains.


Assuntos
Potenciais de Ação , Cloretos/metabolismo , Células Intersticiais de Cajal/metabolismo , Membro 2 da Família 12 de Carreador de Soluto/metabolismo , Animais , Bumetanida/farmacologia , Canais de Cálcio Tipo T/metabolismo , Células Cultivadas , Células HEK293 , Humanos , Células Intersticiais de Cajal/efeitos dos fármacos , Células Intersticiais de Cajal/fisiologia , Camundongos , Periodicidade , Inibidores de Simportadores de Cloreto de Sódio e Potássio/farmacologia , Membro 2 da Família 12 de Carreador de Soluto/genética
7.
Am J Physiol Cell Physiol ; 307(6): C561-70, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25055825

RESUMO

Enteric inhibitory neurotransmission is an important feature of the neural regulation of gastrointestinal motility. Purinergic neurotransmission, via P2Y1 receptors, mediates one phase of inhibitory neural control. For decades, ATP has been assumed to be the purinergic neurotransmitter and smooth muscle cells (SMCs) have been considered the primary targets for inhibitory neurotransmission. Recent experiments have cast doubt on both of these assumptions and suggested that another cell type, platelet-derived growth factor receptor-α-positive (PDGFRα(+)) cells, is the target for purinergic neurotransmission. We compared responses of PDGFRα(+) cells and SMCs to several purine compounds to determine if these cells responded in a manner consistent with enteric inhibitory neurotransmission. ATP hyperpolarized PDGFRα(+) cells but depolarized SMCs. Only part of the ATP response in PDGFRα(+) cells was blocked by MRS 2500, a P2Y1 antagonist. ADP, MRS 2365, ß-NAD, and adenosine 5-diphosphate-ribose, P2Y1 agonists, hyperpolarized PDGFRα(+) cells, and these responses were blocked by MRS 2500. Adenosine 5-diphosphate-ribose was more potent in eliciting hyperpolarization responses than ß-NAD. P2Y1 agonists failed to elicit responses in SMCs. Small hyperpolarization responses were elicited in SMCs by a small-conductance Ca(2+)-activated K(+) channel agonist, cyclohexyl-[2-(3,5-dimethyl-pyrazol-1-yl)-6-methyl-pyrimidin-4-yl]-amine, consistent with the low expression and current density of small-conductance Ca(2+)-activated K(+) channels in these cells. Large-amplitude hyperpolarization responses, elicited in PDGFRα(+) cells, but not SMCs, by P2Y1 agonists are consistent with the generation of inhibitory junction potentials in intact muscles in response to purinergic neurotransmission. The responses of PDGFRα(+) cells and SMCs to purines suggest that SMCs are unlikely targets for purinergic neurotransmission in colonic muscles.


Assuntos
Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Colo/metabolismo , Músculo Liso/metabolismo , Miócitos de Músculo Liso/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Animais , Colo/citologia , Colo/efeitos dos fármacos , Colo/inervação , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Potenciais da Membrana , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Músculo Liso/citologia , Músculo Liso/efeitos dos fármacos , Músculo Liso/inervação , Miócitos de Músculo Liso/efeitos dos fármacos , Inibição Neural , Técnicas de Patch-Clamp , Agonistas do Receptor Purinérgico P2Y/farmacologia , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptores Purinérgicos P2Y1/efeitos dos fármacos , Receptores Purinérgicos P2Y1/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Baixa/agonistas , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo
8.
J Physiol ; 592(21): 4733-45, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-25217377

RESUMO

Four types of electrical activity were recorded and related to cell structure by intracellular recording and dye injection into impaled cells in muscles of rabbit small intestine. The specific cell types from which recordings were made were longitudinal smooth muscle cells (LSMCs), circular smooth muscle cells (CSMCs), interstitial cells of Cajal distributed in the myenteric region (ICC-MY) and fibroblast-like cells (FLCs). Slow waves (slow wavesSMC) were recorded from LSMCs and CSMCs. Slow waves (slow wavesICC) were of greatest amplitude (>50 mV) and highest maximum rate of rise (>10 V s(-1)) in ICC-MY. The dominant activity in FLCs was spontaneous transient hyperpolarizations (STHs), with maximum amplitudes above 30 mV. STHs were often superimposed upon small amplitude slow waves (slow wavesFLC). STHs displayed a cyclical pattern of discharge irrespective of background slow wave activity. STHs were inhibited by MRS2500 (3 µm), a P2Y1 antagonist, and abolished by apamin (0.3 µm), a blocker of small conductance Ca(2+)-activated K(+) channels. Small amplitude STHs (<15 mV) were detected in smooth muscle layers, whereas STHs were not resolved in cells identified as ICC-MY. Electrical field stimulation evoked purinergic inhibitory junction potentials (IJPs) in CSMCs. Purinergic IJPs were not recorded from ICC-MY. These results suggest that FLCs may regulate smooth muscle excitability in the rabbit small intestine via generation of rhythmic apamin-sensitive STHs. Stimulation of P2Y1 receptors modulates the amplitudes of STHs. Our results also suggest that purinergic inhibitory motor neurons regulate the motility of the rabbit small intestine by causing IJPs in FLCs that conduct to CSMCs.


Assuntos
Potenciais da Membrana/fisiologia , Miócitos de Músculo Liso/fisiologia , Animais , Células Intersticiais de Cajal/fisiologia , Masculino , Purinas/metabolismo , Coelhos , Transmissão Sináptica/fisiologia
9.
Am J Physiol Gastrointest Liver Physiol ; 304(9): G823-34, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23429582

RESUMO

Recently platelet-derived growth factor-α-positive cells (PDGFRα(+) cells), previously called "fibroblast-like" cells, have been described in the muscle layers of the gastrointestinal tract. These cells form networks and are involved in purinergic motor neurotransduction. Examination of colon from mice with enhanced green fluorescent protein (eGFP) driven from the endogenous Pdgfra (PDGFRα-eGFP mice) revealed a unique population of PDGFRα(+) cells in the mucosal layer of colon. We investigated the phenotype and potential role of these cells, which have not been characterized previously. Expression of PDGFRα and several additional proteins was surveyed in human and murine colonic mucosae by immunolabeling; PDGFRα(+) cells in colonic mucosa were isolated from PDGFRα-eGFP mice, and the gene expression profile was analyzed by quantitative polymerase chain reaction. We found for the first time that PDGFRα was expressed in subepithelial cells (subepithelial PDGFRα(+) cells) forming a pericryptal sheath from the base to the tip of crypts. These cells were in close proximity to the basolateral surface of epithelial cells and distinct from subepithelial myofibroblasts, which were identified by expression of α-smooth muscle actin and smooth muscle myosin. PDGFRα(+) cells also lay in close proximity to varicose processes of nerve fibers. Mouse subepithelial PDGFRα(+) cells expressed Toll-like receptor genes, purinergic receptor genes, 5-hydroxytryptamine (5-HT) 4 receptor gene, and hedgehog signaling genes. Subepithelial PDGFRα(+) cells occupy an important niche in the lamina propria and may function in transduction of sensory and immune signals and in the maintenance of mucosal homeostasis.


Assuntos
Colo/citologia , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Animais , Colo/metabolismo , Desmina/biossíntese , Humanos , Mucosa Intestinal/citologia , Camundongos , Miofibroblastos/metabolismo , Miosinas/metabolismo , Vimentina/biossíntese
10.
J Cell Mol Med ; 16(7): 1397-404, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22225616

RESUMO

An obstacle to understanding motor pathologies of the gastrointestinal (GI) tract is that the physiology of some of the cellular components of the gut wall is not understood. Morphologists identified fibroblast-like cells in the tunica muscularis many years ago, but little is known about these interstitial cells because of inadequate techniques to identify these cells. Recent findings have shown that fibroblast-like cells express platelet-derived growth factor receptor α (PDGFRα) in mice and that antibodies for these receptors can be used to label the cells. We used immunohistochemical techniques to study the phenotype and intercellular relationships of fibroblast-like cells in the human colon. Fibroblast-like cells are labelled specifically with antibodies to PDGFRα and widely distributed through the tunica muscularis of human colon. These cells form discrete networks in the region of the myenteric plexus and within the circular and longitudinal muscle layers. Platelet-derived growth factor receptor α(+) cells are distinct from c-Kit(+) interstitial cells of Cajal and closely associated with varicose processes of neurons expressing substance P (excitatory motor neurons) or neuronal nitric oxide synthase (nNOS) (inhibitory motor neurons). Platelet-derived growth factor receptor α(+) cells express small conductance Ca(2+)-activated K(+) channels (SK3), which are likely to mediate purinergic neural regulation of colonic muscles. Our data suggest that PDGFRα(+) cells may have an important role in transducing inputs from enteric motor neurons. This study identifies reagents and techniques that will allow investigation of this class of interstitial cells and help develop an understanding of the role of PDGFRα(+) cells in the human GI tract in health and disease.


Assuntos
Colo/citologia , Músculo Liso/citologia , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Idoso , Colo/metabolismo , Fibroblastos , Humanos , Imuno-Histoquímica , Células Intersticiais de Cajal/metabolismo , Pessoa de Meia-Idade , Neurônios Motores/metabolismo , Músculo Liso/metabolismo , Plexo Mientérico/metabolismo , Óxido Nítrico Sintase Tipo I/genética , Óxido Nítrico Sintase Tipo I/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Baixa/genética , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Substância P/metabolismo
11.
Cell Mol Gastroenterol Hepatol ; 14(2): 357-373, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35569815

RESUMO

BACKGROUND & AIMS: Platelet-derived growth factor receptor α (PDGFRα)-positive interstitial cells (PIC) are interposed between enteric nerve fibers and smooth muscle cells (SMCs) in the tunica muscularis of the gastrointestinal tract. PIC have robust expression of small conductance Ca2+ activated K+ channels 3 (SK3 channels) and transduce inhibitory inputs from purinergic and sympathetic nerves in mouse and human colon. We investigated whether PIC also express pituitary adenylate cyclase-activating polypeptide (PACAP) receptors, PAC1 (PAC1R), and are involved in mediating inhibitory regulation of colonic contractions by PACAP in mouse and human colons. METHODS: Gene expression analysis, Ca2+ imaging, and contractile experiments were performed on mouse colonic muscles. Ca2+ imaging, intracellular electrical recordings, and contractile experiments were performed on human colonic muscles. RESULTS: Adcyap1r1 (encoding PAC1R) is highly expressed in mouse PIC. Interstitial cells of Cajal (ICC) and SMCs expressed far lower levels of Adcyap1r. Vipr1 and Vipr2 were expressed at low levels in PIC, ICC, and SMCs. PACAP elicited Ca2+ transients in mouse PIC and inhibited spontaneous phasic contractions via SK channels. In human colonic muscles, PAC1R agonists elicited Ca2+ transients in PIC, hyperpolarized SMCs through SK channels and inhibited spontaneous phasic contractions. CONCLUSIONS: PIC of mouse and human colon utilize PAC1R-SK channel signal pathway to inhibit colonic contractions in response to PACAP. Effects of PACAP are in addition to the previously described purinergic and sympathetic inputs to PIC. Thus, PIC integrate inhibitory inputs from at least 3 neurotransmitters and utilize several types of receptors to activate SK channels and regulate colonic contractile behaviors.


Assuntos
Células Intersticiais de Cajal , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase , Animais , Colo/metabolismo , Humanos , Células Intersticiais de Cajal/metabolismo , Camundongos , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais
12.
PLoS One ; 17(5): e0261743, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35560163

RESUMO

Subepithelial platelet-derived growth factor receptor alpha (PDGFRα)+ cells found in the colonic mucosal tissue come in close contact with epithelial cells, immune cells, neurons, capillaries, and lymphatic networks. Mucosal subepithelial PDGFRα+ cells (MuPαC) are important regulators in various intestinal diseases including fibrosis and inflammation. However, the transcriptome of MuPαC has not yet been elucidated. Using Pdgfra-eGFP mice and flow cytometry, we isolated colonic MuPαC and obtained their transcriptome data. In analyzing the transcriptome, we identified three novel, and selectively expressed, markers (Adamdec1, Fin1, and Col6a4) found in MuPαC. In addition, we identified a unique set of MuPαC-enriched genetic signatures including groups of growth factors, transcription factors, gap junction proteins, extracellular proteins, receptors, cytokines, protein kinases, phosphatases, and peptidases. These selective groups of genetic signatures are linked to the unique cellular identity and function of MuPαC. Furthermore, we have added this MuPαC transcriptome data to our Smooth Muscle Genome Browser that contains the transcriptome data of jejunal and colonic smooth muscle cells (SMC), interstitial cells of Cajal (ICC), and smooth muscle resident PDGFRα+ cells: (https://med.unr.edu/physio/transcriptome). This online resource provides a comprehensive reference of all currently known genetic transcripts expressed in primary MuPαC in the colon along with smooth muscle resident PDGFRα cells, SMC, and ICC in the murine colon and jejunum.


Assuntos
Células Intersticiais de Cajal , Receptor alfa de Fator de Crescimento Derivado de Plaquetas , Animais , Biomarcadores/metabolismo , Colo/metabolismo , Perfilação da Expressão Gênica , Células Intersticiais de Cajal/metabolismo , Mucosa Intestinal/metabolismo , Camundongos , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo
13.
J Physiol ; 589(Pt 3): 697-710, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21173079

RESUMO

Smooth muscles, as in the gastrointestinal tract, are composed of several types of cells. Gastrointestinal muscles contain smooth muscle cells, enteric neurons, glial cells, immune cells, and various classes of interstitial cells. One type of interstitial cell, referred to as 'fibroblast-like cells' by morphologists, are common, but their function is unknown. These cells are found near the terminals of enteric motor neurons, suggesting they could have a role in generating neural responses that help control gastrointestinal movements. We used a novel mouse with bright green fluorescent protein expressed specifically in the fibroblast-like cells to help us identify these cells in the mixture of cells obtained when whole muscles are dispersed with enzymes. We isolated these cells and found they respond to a major class of inhibitory neurotransmitters - purines. We characterized these responses, and our results provide a new hypothesis about the role of fibroblast-like cells in smooth muscle tissues.


Assuntos
Fibroblastos/citologia , Fibroblastos/fisiologia , Trato Gastrointestinal/fisiologia , Músculo Liso/fisiologia , Transmissão Sináptica/fisiologia , Difosfato de Adenosina/farmacologia , Trifosfato de Adenosina/farmacologia , Animais , Apamina/farmacologia , Cálcio/farmacologia , Membrana Celular/efeitos dos fármacos , Membrana Celular/fisiologia , Charibdotoxina/farmacologia , Nucleotídeos de Desoxiadenina/farmacologia , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Fenômenos Eletrofisiológicos/fisiologia , Fibroblastos/efeitos dos fármacos , Trato Gastrointestinal/citologia , Expressão Gênica/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios Motores/citologia , Neurônios Motores/metabolismo , Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/fisiologia , NAD/farmacologia , Óxido Nítrico Sintase Tipo I/metabolismo , Técnicas de Patch-Clamp , Antagonistas do Receptor Purinérgico P2Y/farmacologia , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores Purinérgicos P2Y1/genética , Canais de Potássio Ativados por Cálcio de Condutância Baixa/antagonistas & inibidores , Canais de Potássio Ativados por Cálcio de Condutância Baixa/genética , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Proteínas Vesiculares de Transporte de Acetilcolina/metabolismo
14.
Cell Tissue Res ; 344(1): 17-30, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21337122

RESUMO

Interstitial cells of Cajal (ICC) have been shown to participate in nitrergic neurotransmission in various regions of the gastrointestinal (GI) tract. Recently, fibroblast-like cells, which are positive for platelet-derived growth factor receptor α (PDGFRα(+)), have been suggested to participate additionally in inhibitory neurotransmission in the GI tract. The distribution of ICC and PDGFRα(+) cell populations and their relationship to inhibitory nerves within the mouse internal anal sphincter (IAS) are unknown. Immunohistochemical techniques and confocal microscopy were therefore used to examine the density and arrangement of ICC, PDGFRα(+) cells and neuronal nitric-oxide-synthase-positive (nNOS(+)) nerve fibers in the IAS of wild-type (WT) and W/W ( v ) mice. Of the total tissue volume within the IAS circular muscle layer, 18% consisted in highly branched PDGFRα(+) cells (PDGFRα(+)-IM). Other populations of PDGFRα(+) cells were observed within the submucosa and along the serosal and myenteric surfaces. Spindle-shaped intramuscular ICC (ICC-IM) were present in the WT mouse IAS but were largely absent from the W/W ( v ) IAS. The ICC-IM volume (5% of tissue volume) in the WT mouse IAS was significantly smaller than that of PDGFRα(+)-IM. Stellate-shaped submucosal ICC (ICC-SM) were observed in the WT and W/W ( v ) IAS. Minimum surface distance analysis revealed that nNOS(+) nerve fibers were closely aligned with both ICC-IM and PDGFRα(+)-IM. An even closer association was seen between ICC-IM and PDGFRα(+)-IM. Thus, a close morphological arrangement exists between inhibitory motor neurons, ICC-IM and PDGFRα(+)-IM suggesting that some functional interaction occurs between them contributing to inhibitory neurotransmission in the IAS.


Assuntos
Canal Anal/inervação , Canal Anal/ultraestrutura , Células Intersticiais de Cajal/citologia , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/análise , Animais , Fibroblastos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo I/análise
15.
Differentiation ; 77(3): 277-89, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19272526

RESUMO

Bone morphogenetic protein (BMP) signaling is essential for normal development of the gastrointestinal (GI) tract. BMPs also play multiple roles in vascular smooth muscle cells; however, the BMP signaling in the development of the GI musculature remains to be clarified. We investigated the expression of BMPs and their receptors in mouse embryonic GI tracts by immunohistochemistry and in situ hybridization. We demonstrated that BMP2, BMP receptor Ib and BMP receptor II were expressed in the smooth muscle progenitors from E12 to E13 for the first time. BMP signaling on smooth muscle differentiation was examined by implantation of agarose beads soaked with BMPs in the in vitro developmental model that is gut-like structures from mouse embryonic stem (ES) cells. BMP2 rather than BMP4 beads enhanced smooth muscle differentiation, and increased gut-like structures showing spontaneous contractions and expressing intensive alpha-smooth muscle actin immunoreactivity. This increase was confirmed by up-regulation of SM22 mRNA shown by real-time PCR. By addition of noggin beads or noggin to the medium at BMP2 bead implantation, the ratio of contractive gut-like structures decreased. Implantation of BMP2 beads at EB7 (EB--embryoid bodies) (corresponding to E12 or E13 of mouse embryo) showed the highest effects and up-regulation of transcription factors msx-1 after 24h. This increase was blocked by noggin, and msx-1 decreased to almost the control level after 60 h. BMP2 beads at EB7 increased platelet-derived growth factor-A (PDGF-A) in the differentiating smooth muscle cells. We have recently reported that PDGF-A is expressed in the developing inner circular smooth muscle and is crucial for the longitudinal smooth muscle differentiation. Taken together, BMP signaling was expressed for a short window in the smooth muscle progenitors and the signal, especially BMP2, plays an essential role in smooth muscle differentiation in cooperation with PDGF signaling.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Diferenciação Celular , Intestinos/citologia , Miócitos de Músculo Liso , Transdução de Sinais , Células-Tronco , Animais , Proteínas Morfogenéticas Ósseas/classificação , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento , Imuno-Histoquímica , Camundongos , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/citologia , Células-Tronco/metabolismo
16.
Cell Mol Gastroenterol Hepatol ; 10(3): 658-671.e1, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32376421

RESUMO

BACKGROUND & AIMS: Colonic musculature contain smooth muscle cells (SMC), interstitial cells of Cajal (ICC), and platelet-derived growth factor receptor α+ cells (PDGFRα+ cells), which are electrically coupled and operate together as the SIP syncytium. PDGFRα+ cells have enriched expression of small conductance Ca2+-activated K+ (SK) channels. Purinergic enteric neural input activates SK channels in PDGFRα+ cells, hyperpolarizes SMC, and inhibits colonic contractions. Recently we discovered that PDGFRα+ cells in mouse colon have enriched expression of α1A adrenoceptors (ARs), which coupled to activation of SK channels and inhibited colonic motility, and α1A ARs were principal targets for sympathetic regulation of colonic motility. Here we investigated whether PDGFRα+ cells in human colon express α1A ARs and share the roles as targets for sympathetic regulation of colonic motility. METHODS: Isometric tension recording, intracellular recording, and Ca2+ imaging were performed on muscles of the human colon. Responses to α1 ARs agonists or electric field stimulation with AR antagonists and neuroleptic reagents were studied. RESULTS: Exogenous or endogenous norepinephrine released from nerve fibers inhibited colonic contractions through binding to α1A ARs or enhanced colonic contractions by acting on α1D ARs. Inhibitory responses were blocked by apamin, an antagonist of SK channels. Phenylephrine, α1 AR agonists, or norepinephrine increased intracellular [Ca2+] in PDGFRα+ cells, but not in ICC, and hyperpolarized SMCs by binding to α1 ARs expressed by PDGFRα+ cells. CONCLUSIONS: Human colonic contractions are inhibited by α1A ARs expressed in PDGFRα+ cells and activated by α1D ARs expressed in SMC.


Assuntos
Colo/efeitos dos fármacos , Contração Muscular/efeitos dos fármacos , Norepinefrina/farmacologia , Receptores Adrenérgicos alfa 1/metabolismo , Agonistas de Receptores Adrenérgicos alfa 1/farmacologia , Antagonistas de Receptores Adrenérgicos alfa 1/farmacologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Colo/citologia , Colo/metabolismo , Feminino , Trânsito Gastrointestinal/efeitos dos fármacos , Trânsito Gastrointestinal/fisiologia , Humanos , Masculino , Pessoa de Meia-Idade , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo
17.
Nagoya J Med Sci ; 69(3-4): 133-7, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18351232

RESUMO

Mouse embryonic stem (ES) cells are pluripotent and retain their potential to form cells, tissues and organs originated from three embryonic germ layers. Recently, we developed in vitro organ--gut-like structures--from mouse ES cells. They had basically similar morphological features to a mouse gastrointestinal tract in vivo composed of three distinct layers (i.e., epithelium, connective tissue and musculature). Gut-like structures showed spontaneous contractions derived from pacemaker cells (interstitial cells of Cajal) in the musculature. We also examined their formation process and expression pattern of transcription factors crucial for gut organogenesis such as Id2, Sox17, HNF3beta/Foxa2 and GATA4. We found that they mimic the development of embryonic gut in vivo and showed a similar expression pattern of common transcription factors. They also maintain their developmental potential after transplantation to a renal capsule. Therefore, gut-like structures are suitable for in vitro models of gastrointestinal tracts and their development. In addition, we pointed out several unique features different from gut in vivo that provide useful and advantageous tools to investigate the developmental mechanism of the gastrointestinal tract.


Assuntos
Células-Tronco Embrionárias/citologia , Trato Gastrointestinal/citologia , Trato Gastrointestinal/embriologia , Técnicas de Cultura de Órgãos/métodos , Animais , Técnicas de Cultura de Células , Células Cultivadas , Camundongos
18.
PLoS One ; 12(8): e0182265, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28806761

RESUMO

Platelet-derived growth factor receptor alpha (PDGFRα)+ cells are distributed into distinct morphological groups within the serosal, muscular, and submucosal layers as well as the myenteric and deep muscular plexi. PDGFRα+ cells directly interact with interstitial cells of Cajal (ICC) and smooth muscle cells (SMC) in gastrointestinal smooth muscle tissue. These three cell types, SMC, ICC, and PDGFRα+ cells (SIP cells), form an electrical syncytium, which dynamically regulates gastrointestinal motility. We have previously reported the transcriptomes of SMC and ICC. To complete the SIP cell transcriptome project, we obtained transcriptome data from jejunal and colonic PDGFRα+ cells. The PDGFRα+ cell transcriptome data were added to the Smooth Muscle Genome Browser that we previously built for the genome-scale gene expression data of ICC and SMC. This browser provides a comprehensive reference for all transcripts expressed in SIP cells. By analyzing the transcriptomes, we have identified a unique set of PDGFRα+ cell signature genes, growth factors, transcription factors, epigenetic enzymes/regulators, receptors, protein kinases/phosphatases, and ion channels/transporters. We demonstrated that the low voltage-dependent T-type Ca2+ channel Cacna1g gene was particularly expressed in PDGFRα+ cells in the intestinal serosal layer in mice. Expression of this gene was significantly induced in the hyperplasic PDGFRα+ cells of obstructed small intestine in mice. This gene was also over-expressed in colorectal cancer, Crohn's disease, and diverticulitis in human patients. Taken together, our data suggest that Cacna1g exclusively expressed in serosal PDGFRα+ cells is a new pathological marker for gastrointestinal diseases.


Assuntos
Canais de Cálcio Tipo T/metabolismo , Perfilação da Expressão Gênica , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Animais , Canais de Cálcio Tipo T/genética , Desdiferenciação Celular , Proliferação de Células/genética , Separação Celular , Regulação da Expressão Gênica , Genoma , Humanos , Hiperplasia , Hipertrofia , Canais Iônicos/genética , Canais Iônicos/metabolismo , Jejuno/metabolismo , Camundongos , Músculo Liso/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
19.
Nihon Shokakibyo Gakkai Zasshi ; 99(8): 917-24, 2002 Aug.
Artigo em Japonês | MEDLINE | ID: mdl-12229164

RESUMO

BACKGROUND: Usefulness of p53 staining for the differentiation between adenoma and DALM has been reported recently, so recognizable lesions stained positively can be diagnosed as DALMs. For the cases with DALMs, total colectomy has been thought to be necessary. METHODS: Immunohistochemical staining for p53 was performed in 4 adenocarcinomas and 4 adenomas in ulcerative colitis. RESULTS: Three carcinomas and 3 adenomas were positive. One carcinoma (protruded mucosal cancer) and 3 adenomas (1 flat elevated lesion and 2 laterally spreading tumors) stained positively for p53 were treated only by polypectomy or local excision. The patients have been under surveillance for periods ranging from 1 to 10 years, during which no metachronous dysplasia has developed. CONCLUSIONS: These findings suggest that some groups of the polypoid lesions can be resected locally even if stained positively by p53 immunohistochemistry.


Assuntos
Colite Ulcerativa/complicações , Lesões Pré-Cancerosas/cirurgia , Proteína Supressora de Tumor p53/análise , Adulto , Idoso , Colite Ulcerativa/patologia , Colite Ulcerativa/cirurgia , Pólipos do Colo/etiologia , Pólipos do Colo/cirurgia , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Lesões Pré-Cancerosas/patologia
20.
Stem Cells ; 24(12): 2618-26, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16888283

RESUMO

Recently, we reported the formation of gut-like structures from mouse ESCs in vitro. To determine whether ESCs provide an in vitro model of gastrointestinal (GI) tracts and their organogenesis, we investigated the morphological features, formation process, cellular development, and regional location within the GI tract by immunohistochemistry, electron microscopy, and reverse transcription-polymerase chain reaction. We also examined the developmental potential by transplantation into kidney capsules. The results demonstrated that Id2-expressing epithelium developed first, alpha-smooth muscle actin appeared around the periphery, and finally, the gut-like structures were formed into a three-layer organ with well-differentiated epithelium. A connective tissue layer and musculature with interstitial cells of Cajal developed, similar to organogenesis of the embryonic gut. Enteric neurons appeared underdeveloped, and blood vessels were absent. Many structures expressed intestinal markers Cdx2 and 5-hydroxytryptamine but not the stomach marker H(+)/K(+) ATPase. Transplants obtained blood vessels and extrinsic nerve growth from the host to prolong life, and even grafts of premature structures did not form teratoma. In conclusion, gut-like structures were provided with prototypical tissue components of the GI tract and are inherent in the intestine rather than the stomach. The formation process was basically same as in gut organogenesis. They maintain their developmental potential after transplantation. Therefore, gut-like structures provide a unique and useful in vitro system for development and stem cell studies of the GI tract, including transplantation experiments.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/transplante , Trato Gastrointestinal/embriologia , Organogênese , Animais , Biomarcadores , Vasos Sanguíneos , Cápsula Glomerular/citologia , Feminino , Feto/embriologia , Trato Gastrointestinal/irrigação sanguínea , Trato Gastrointestinal/inervação , Trato Gastrointestinal/ultraestrutura , Regulação da Expressão Gênica , Camundongos , Camundongos SCID , Modelos Biológicos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA