Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Neuroendocrinol ; 27(12): 872-86, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26442444

ABSTRACT

Ghrelin, a gut hormone originating from the post-translational cleavage of preproghrelin, is the endogenous ligand of growth hormone secretagogue receptor 1a (GHS-R1a). Within the growth hormone (GH) axis, the biological activity of ghrelin requires octanoylation by ghrelin-O-acyltransferase (GOAT), conferring selective binding to the GHS-R1a receptor via acylated ghrelin. Complete loss of preproghrelin-derived signalling (through deletion of the Ghrl gene) contributes to a decline in peak GH release; however, the selective contribution of endogenous acyl-ghrelin to pulsatile GH release remains to be established. We assessed the pulsatile release of GH in ad lib. fed male germline goat(-/-) mice, extending measures to include mRNA for key hypothalamic regulators of GH release, and peripheral factors that are modulated relative to GH release. The amount of GH released was reduced in young goat(-/-) mice compared to age-matched wild-type mice, whereas pulse frequency and irregularity increased. Altered GH release did not coincide with alterations in hypothalamic Ghrh, Srif, Npy or Ghsr mRNA expression, or pituitary GH content, suggesting that loss of Goat does not compromise canonical mechanisms that contribute to pituitary GH production and release. Although loss of Goat resulted in an irregular pattern of GH release (characterised by an increase in the number of GH pulses observed during extended secretory events), this did not contribute to a change in the expression of sexually dimorphic GH-dependent liver genes. Of interest, circulating levels of insulin-like growth factor (IGF)-1 were elevated in goat(-/-) mice. This rise in circulating levels of IGF-1 was correlated with an increase in GH pulse frequency, suggesting that sustained or increased IGF-1 release in goat(-/-) mice may occur in response to altered GH release patterning. Our observations demonstrate that germline loss of Goat alters GH release and patterning. Although the biological relevance of altered GH secretory patterning remains unclear, we propose that this may contribute to sustained IGF-1 release and growth in goat(-/-) mice.


Subject(s)
Acyltransferases/deficiency , Acyltransferases/physiology , Growth Hormone/metabolism , Acyltransferases/genetics , Animals , Growth Hormone-Releasing Hormone/biosynthesis , Hypothalamus/metabolism , Insulin-Like Growth Factor I/metabolism , Male , Membrane Proteins , Mice , Mice, Knockout , Neuropeptide Y/biosynthesis , Receptors, Ghrelin/biosynthesis , Somatostatin/biosynthesis
2.
Endocr Relat Cancer ; 12(4): 839-50, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16322325

ABSTRACT

While oestrogen, progesterone and growth factors, including growth hormone (GH), are clearly implicated in the pathogenesis of breast cancer, there is now evidence that the newly described ghrelin axis is also involved. The aims of this study were to investigate the expression of the ghrelin axis in breast cancer tissues and cell lines and to examine the effect of ghrelin on breast cancer cell proliferation in vitro. Ghrelin and its functional receptor, the growth hormone secretagogue receptor (GHSR) type 1a, were expressed in normal breast tissue and breast cancer specimens and cell lines. In contrast, the truncated GHSR type 1b isoform was exclusively expressed in breast carcinoma, suggesting that it has potential as a diagnostic marker. Ghrelin treatment significantly increases the proliferation of the MDA-MB-435 and MDA-MB-231 breast cancer cell lines in vitro. In addition, we have described the expression of a human preproghrelin isoform, exon 3-deleted preproghrelin, which encodes mature ghrelin plus a novel C-terminal peptide. Quantitative RT-PCR was used to demonstrate that this mRNA isoform is highly expressed in the MDA-MB-435 metastatic breast cancer cell line relative to the benign MCF-10A breast epithelial cell line. The unique C-terminal peptide of exon 3-deleted preproghrelin is expressed in the glandular epithelium of breast cancer tissues, with high-grade carcinoma exhibiting the strongest immunoreactivity. The data presented here suggest that components of the ghrelin axis may represent novel markers for breast cancer and potential therapeutic targets.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma/metabolism , Peptide Hormones/metabolism , Peptide Hormones/pharmacology , Peptide Hormones/physiology , Amino Acid Sequence , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/diagnosis , Breast Neoplasms/pathology , Carcinoma/diagnosis , Carcinoma/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Ghrelin , Humans , Immunohistochemistry , Molecular Sequence Data , Peptide Hormones/analysis , Peptide Hormones/genetics , Protein Isoforms/analysis , Protein Isoforms/genetics , RNA, Messenger/analysis , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/analysis , Receptors, G-Protein-Coupled/genetics , Receptors, Ghrelin , Sequence Deletion , Transcription, Genetic
3.
Endocrinology ; 146(1): 432-40, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15471962

ABSTRACT

Ghrelin, an n-octanoylated 28-amino-acid peptide capable of inducing GH secretion and food intake in humans and rats, is the endogenous ligand for the GH secretagogue receptor (GHS-R). Here we describe the expression and tissue distribution of the ghrelin/GHS-R axis in the mouse. We also report for the first time the identification of a novel mouse ghrelin mRNA variant in which there is a complete deletion of exon 4. Translation of this variant mRNA yields a protein containing ghrelin and an alternative C-terminal domain with a unique C-terminal peptide sequence. RT-PCR with primers specific for mouse ghrelin was used to demonstrate the mRNA expression of the full preproghrelin transcript and the exon 4-deleted variant in multiple mouse tissues. Real-time PCR was also employed to quantitate mRNA expression of ghrelin, the novel isoform and a previously reported ghrelin gene variant, ghrelin gene-derived transcript. We also demonstrated the tissue expression of the functional GHS-R in the mouse. Immunohistochemistry, employing antibodies raised against the mature human n-octanoylated ghrelin peptide and the putative C-terminal peptide encoded by the exon 4-deleted proghrelin variant, was used to demonstrate protein expression of ghrelin and the variant in multiple mouse tissues including stomach, kidney, and reproductive tissues. The coexpression of ghrelin and its receptor in a wide range of murine tissues suggests varied autocrine/paracrine roles for these peptides. Exon 4-deleted proghrelin, a novel mouse proghrelin isoform with a unique C-terminal peptide sequence, is also widely expressed in the mouse and thus may possess biological activity in these tissues.


Subject(s)
Exons , Gene Deletion , Gene Expression , Genetic Variation , Peptide Hormones/genetics , Protein Precursors/genetics , Amino Acid Sequence , Animals , Base Sequence , Blotting, Western , Computer Systems , Gastric Mucosa/metabolism , Ghrelin , Immunohistochemistry , Mice , Mice, Inbred Strains , Molecular Sequence Data , Peptide Fragments/genetics , Peptide Hormones/metabolism , Polymerase Chain Reaction , Protein Precursors/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Ghrelin , Sequence Homology, Amino Acid , Tissue Distribution
4.
J Clin Endocrinol Metab ; 90(3): 1798-804, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15585554

ABSTRACT

Ghrelin, the endogenous ligand of the GH secretagogue receptor (GHS-R), is a newly identified, ubiquitously expressed molecule that has been involved in a wide array of endocrine and nonendocrine functions, including cell proliferation. In this context, our group recently reported the expression of ghrelin and its functional receptor, the GHS-R type 1a, in the human ovary and testis as well as several testicular tumors. Ovarian malignancies, however, remain unexplored. Notably, a vast majority of ovarian tumors derive from the surface epithelium, which originates from the celomic epithelium. Considering the proven expression of ghrelin in the human ovary, and its reported effects in the proliferative activity of different cancer cell lines, we aimed at evaluating whether the ovarian surface epithelium as well as related reproductive structures and tumors are potential targets of ghrelin. To this end, expression of GHS-R1a was analyzed by immunohistochemistry in a panel of normal, metaplastic, and neoplastic tissues. Uniform GHS-R1a immunostaining was detected throughout the ovarian surface epithelium. Likewise, ciliated cells within the fallopian tube epithelium showed strong GHS-R1a expression. In contrast, other celomic derivatives, such as endometrium and endocervix, were negative for GHS-R1a immunoreactivity. In keeping with data from normal tissues, inclusion cysts from the surface epithelium expressed GHS-R1a. Similarly, benign serous tumors resembling fallopian tube epithelium were also positive, whereas serous cystadenocarcinomas showed GHS-R1a expression only in highly differentiated specimens. In contrast, other neoplasms, such as mucinous cystadenomas and cystadenocarcinomas, endometrioid tumors, clear cell carcinomas, and Brenner tumors, did not express GHS-R1a. In conclusion, our results demonstrate that the ovarian surface epithelium and related tumors are potential targets for systemic or locally produced ghrelin because they express the functional type 1a GHS-R. Considering the relevant role of the ovarian surface epithelium in key physiological events (such as ovulation) and neoplastic transformation of the ovary, the potential actions of ghrelin in those phenomena merit further investigation.


Subject(s)
Fallopian Tubes/metabolism , Ovarian Neoplasms/metabolism , Ovary/metabolism , Receptors, G-Protein-Coupled/metabolism , Cervix Uteri/embryology , Cervix Uteri/metabolism , Endometrium/embryology , Endometrium/metabolism , Epithelial Cells/metabolism , Fallopian Tubes/embryology , Female , Humans , Immunohistochemistry , Mullerian Ducts/embryology , Ovarian Neoplasms/pathology , Ovary/embryology , Receptors, Ghrelin
5.
J Clin Endocrinol Metab ; 89(1): 400-9, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14715878

ABSTRACT

Ghrelin, the endogenous ligand for the GH secretagogue receptor (GHS-R), has been primarily linked to the central neuroendocrine regulation of GH secretion and food intake, although additional peripheral actions of ghrelin have also been reported. In this context, the expression of ghrelin and its cognate receptor has been recently demonstrated in rat testis, suggesting a role for this molecule in the direct control of male gonadal function. However, whether this signaling system is present in human testis remains largely unexplored. In this study we report the expression and cellular location of ghrelin and its functional receptor, the type 1a GHS-R, in adult human testis. In addition, evaluation of ghrelin and GHS-R1a immunoreactivity in testicular tumors and dysgenetic tissue is presented. The expression of the mRNAs encoding ghrelin and GHS-R1a was demonstrated in human testis specimens by RT-PCR, followed by direct sequencing. In normal testis, ghrelin immunostaining was demonstrated in interstitial Leydig cells and, at lower intensity, in Sertoli cells within the seminiferous tubules. In contrast, ghrelin was not detected in germ cells at any stage of spermatogenesis. The cognate ghrelin receptor showed a wider pattern of cellular distribution, with detectable GHS-R1a protein in germ cells, mainly in pachytene spermatocytes, as well as in somatic Sertoli and Leydig cells. Ghrelin immunoreactivity was absent in poorly differentiated Leydig cell tumor, which retained the expression of GHS-R1a peptide. In contrast, highly differentiated Leydig cell tumors expressed both the ligand and the receptor. The expression of ghrelin and GHS-R1a was also detected in dysgenetic Sertoli cell-only seminiferous tubules, whereas germ cell tumors (seminoma and embryonal carcinoma) were negative for ghrelin and were weakly positive for GHS-R1a. In conclusion, our results demonstrate that ghrelin and the type 1a GHS-R are expressed in adult human testis and testicular tumors. Overall, the expression of ghrelin and its functional receptor in human and rat testis, with roughly similar patterns of cellular distribution, is highly suggestive of a conserved role for this newly discovered molecule in the regulation of mammalian testicular function.


Subject(s)
Gene Expression , Peptide Hormones/genetics , Receptors, G-Protein-Coupled/genetics , Testicular Neoplasms/chemistry , Testis/chemistry , Adult , Aged , Carcinoma, Embryonal/chemistry , Ghrelin , Humans , Immunohistochemistry , Leydig Cell Tumor/chemistry , Leydig Cells/chemistry , Male , Middle Aged , Peptide Hormones/analysis , RNA, Messenger/analysis , Receptors, G-Protein-Coupled/analysis , Receptors, Ghrelin , Reverse Transcriptase Polymerase Chain Reaction , Seminiferous Tubules/chemistry , Seminoma/chemistry , Sertoli Cells/chemistry
6.
J Clin Endocrinol Metab ; 88(2): 879-87, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12574228

ABSTRACT

Ghrelin is a novel 28-amino acid peptide identified as the endogenous ligand for the GH secretagogue receptor (GHS-R). Besides its hallmark central neuroendocrine effects in the control of GH secretion and food intake, an unexpected reproductive facet of ghrelin has recently emerged because expression of this molecule and its cognate receptor has been demonstrated in rat testis. However, whether this signaling system is present in human gonads remains to be evaluated. In this study, we have assessed the presence and cellular location of ghrelin and its functional receptor, namely the type 1a GHS-R, in the cyclic human ovary by means of immunohistochemistry using specific polyclonal antibodies. Strong ghrelin immunostaining was demonstrated in ovarian hilus interstitial cells. In contrast, ghrelin signal was not detected in ovarian follicles at any developmental stage, nor was it present in newly formed corpora lutea (CL) at very early development. However, specific ghrelin immunoreactivity was clearly observed in young and mature CL, whereas expression of the peptide disappeared in regressing luteal tissue. Concerning the cognate receptor, ovarian expression of GHS-R1a protein showed a wider pattern of tissue distribution, with detectable specific signal in oocytes as well as somatic follicular cells; luteal cells from young, mature, old, and regressing CL; and interstitial hilus cells. Of particular note, follicular GHS-R1a peptide expression paralleled follicle development with stronger immunostaining in granulosa and theca layers of healthy antral follicles. In conclusion, our results are the first to demonstrate that ghrelin and its functional type 1a receptor are expressed in the cyclic human ovary with distinct patterns of cellular location. The presence of both components (ligand and receptor) of the ghrelin signaling system within the human ovary opens up the possibility of a potential regulatory role of this novel molecule in ovarian function under physiological and pathophysiological conditions.


Subject(s)
Ovarian Follicle/chemistry , Peptide Hormones/analysis , Receptors, Cell Surface/analysis , Receptors, G-Protein-Coupled , Antibody Specificity , Female , Ghrelin , Humans , Menstrual Cycle/physiology , Peptide Hormones/immunology , Receptors, Cell Surface/immunology , Receptors, Ghrelin
7.
Biol Reprod ; 68(5): 1631-40, 2003 May.
Article in English | MEDLINE | ID: mdl-12606422

ABSTRACT

Recent evidence from our research suggested the direct role of ghrelin in the control of testicular function. However, the pattern of expression and hormonal regulation of the gene encoding its cognate receptor (i.e., the growth hormone-secretagogue receptor [GHS-R]) in the male gonad remains to be fully elucidated. In this paper, overall expression of GHS-R mRNA in rat testis was compared with that of the functional receptor form, namely GHS-R type 1a, in different developmental and experimental settings. In addition, cellular distribution of GHS-R within adult testis tissue was assessed. Our analyses demonstrated persistent expression of the GHS-R gene in rat testis throughout postnatal development. In contrast, testicular expression of GHS-R type 1a mRNA remained undetectable before puberty and sharply increased thereafter. In adult testis, GHS-R1a mRNA expression presented a scattered pattern of cellular distribution, including Sertoli and Leydig cells that also showed specific GHS-R1a immunoreactivity. Expression of total GHS-R and specific GHS-R1a mRNAs was detected in isolated seminiferous tubule preparations, with varying levels throughout the defined stages of the spermatogenic cycle. In addition, testicular expression of total GHS-R and GHS-R1a mRNAs was up-regulated by exposure to ghrelin in vitro and after stimulation with FSH in vivo. In conclusion, our data demonstrate that expression of the GHS-R gene in rat testis takes place in a developmental, stage-specific, and hormonally regulated manner. Divergent expression of total GHS-R and type 1a specific mRNAs was detected at certain stages of postnatal development and spermatogenic cycle, thus raising the possibility that, in addition to net changes in GHS-R gene expression, the balance between receptor subtypes may represent a novel mechanism for the tuning of ghrelin sensitivity in rat testis.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Hormones/physiology , RNA, Messenger/biosynthesis , Receptors, G-Protein-Coupled/biosynthesis , Testis/growth & development , Testis/metabolism , Animals , Ghrelin , Gonadotropins/metabolism , In Situ Hybridization , Male , Peptide Hormones/metabolism , RNA, Messenger/genetics , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/genetics , Receptors, Ghrelin , Reverse Transcriptase Polymerase Chain Reaction , Seminiferous Tubules/metabolism , Signal Transduction/physiology , Spermatogenesis/physiology , Testis/cytology
8.
Aust Vet J ; 80(1-2): 32-6, 2002.
Article in English | MEDLINE | ID: mdl-12180875

ABSTRACT

An 8-year-old mare, with a foal at foot, was inseminated on foal heat with frozen semen, with the resultant pregnancy lost between days 34 and 41. The right ovary developed a large anovulatory follicle that was non-responsive to multiple doses of ovulating agents. The follicle eventually appeared to luteinise, although plasma progesterone concentrations did not reflect this. Another follicle developed, responded to GnRH and resulted in a pregnancy from frozen semen that went to term with a healthy foal. When the mare was examined after foaling, the structure on the right ovary appeared to be a granulosa cell tumour; the left ovary was smaller than normal and non-functional. Surgical removal of the right ovary before increasing photoperiod resulted in a return to function of the left ovary and a pregnancy to frozen semen on the second cycle following removal. Figures showing concentrations of inhibin, progesterone, androstenedione, oestradiol and testosterone are presented for this entire period. Unusual ovarian activity in the mare might be a prelude to the development of a granulosa cell tumour.


Subject(s)
Granulosa Cell Tumor/veterinary , Horse Diseases/diagnosis , Ovarian Neoplasms/veterinary , Ovary/metabolism , Animals , Diagnosis, Differential , Female , Granulosa Cell Tumor/blood , Granulosa Cell Tumor/diagnosis , Hormones/blood , Horse Diseases/pathology , Horse Diseases/surgery , Horses , Ovarian Neoplasms/blood , Ovarian Neoplasms/diagnosis
9.
Growth Horm IGF Res ; 12(2): 126-36, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12175650

ABSTRACT

Prostate cancer is a significant cause of morbidity and mortality in Western males. While it is known that androgens play a central role in prostate cancer development and progression, other hormones and growth factors are also involved in prostate growth. Insulin-like growth factor-I (IGF-I) plasma levels have been associated with prostate cancer risk, and growth hormone (GH), a major factor regulating IGF levels, also appears to have a role in prostate cancer cell growth. Most significantly, GH has been shown to increase the rate of cell proliferation in prostate cancer cell lines. We have now demonstrated the co-expression of GH and GH receptor (GHR) mRNA isoforms in the ALVA41, PC3, DU145, LNCaP prostate cancer cells by reverse transcription polymerase chain reaction. Sequence analysis has confirmed that these cell lines express the pituitary form of GH mRNA and also the placental mRNA isoform. These prostate cancer cell lines also express the full-length mRNA for the GHR and the exon 3 deleted isoform. We have also demonstrated the presence of GH and GHR proteins in these cell lines by immunohistochemistry. GH expression has not been described previously in human prostate cancer cells. The co-expression of GH and its receptor would enable an autocrine-paracrine pathway to exist in the prostate that would be capable of stimulating prostate growth, either directly via the GHR or indirectly via IGF production. The GH axis in the prostate could therefore be an important additional target for the future development of prostate cancer therapies.


Subject(s)
Growth Hormone/biosynthesis , Growth Hormone/chemistry , Prostatic Neoplasms/metabolism , Receptors, Somatotropin/biosynthesis , Receptors, Somatotropin/chemistry , Blotting, Northern , Blotting, Southern , Cell Division , Cell Nucleus/metabolism , Culture Media, Conditioned/pharmacology , DNA Primers/pharmacology , DNA, Complementary/metabolism , Exons , Humans , Immunohistochemistry , Male , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
10.
J Endocrinol ; 172(3): R7-11, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11874717

ABSTRACT

This study has examined the expression of two new facets of the growth hormone axis, the growth hormone secretagogue receptor (GHS-R) and its recently identified putative natural ligand ghrelin, in prostate cancer cells. GHS-R 1a and 1b isoforms and ghrelin mRNA expression were detected by RT-PCR in the ALVA-41, LNCaP, DU145 and PC3 prostate cancer cell lines. A normal prostate cDNA library expressed GHS-R1a, but not the 1b isoform or ghrelin. Immunohistochemical staining for the GHS-R 1a isoform and ghrelin was positive in the four cell lines studied. PC3 cells showed increased cell proliferation in vitro in response to ghrelin to levels 33% above untreated controls, implying a potential tumour-promoting role for ghrelin in this tissue. This study is the first to demonstrate the co-expression of the GHS-R and ghrelin in prostate cancer cells. It is also the first study to provide evidence that a previously unrecognised autocrine/paracrine pathway involving ghrelin, that is capable of stimulating growth, exists in prostate cancer.


Subject(s)
Peptide Hormones , Peptides/metabolism , Prostatic Neoplasms/metabolism , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled , Ghrelin , Humans , Male , Peptides/genetics , Protein Isoforms/metabolism , RNA, Messenger/analysis , Receptors, Cell Surface/genetics , Receptors, Ghrelin , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
11.
Prostate ; 49(2): 116-21, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11582590

ABSTRACT

BACKGROUND: Recent studies have shown that GHRH antagonists inhibit prostate tumour growth and IGF-II production both in vivo and in vitro. The mechanism underlying these observations is unknown, but may involve an interaction with a prostatic GHRH receptor (GHRH-R), raising the possibility of an autocrine pathway for the GHRH axis in the prostate. METHODS: GHRH and GHRH-R mRNA expression was examined by RT-PCR in human prostate cancer cell lines, and the authenticity of PCR products was confirmed by Southern analysis and cDNA sequencing. Immunohistochemical techniques were used to examine the expression of GHRH protein in prostate cancer cell lines. RESULTS: GHRH-R (mRNA) and GHRH (mRNA and protein) are co-expressed in the ALVA-41, DU145, LNCaP and PC3 human prostate cancer cell lines. CONCLUSIONS: These observations suggest the presence of an intact prostatic GHRH autocrine pathway which may stimulate prostate cell proliferation. This pathway may be disrupted by GHRH antagonists.


Subject(s)
Growth Hormone-Releasing Hormone/biosynthesis , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Neuropeptide/biosynthesis , Receptors, Pituitary Hormone-Regulating Hormone/biosynthesis , Blotting, Southern , Cell Division/physiology , DNA, Neoplasm/biosynthesis , DNA, Neoplasm/genetics , Growth Hormone-Releasing Hormone/genetics , Humans , Immunohistochemistry , Male , Prostatic Neoplasms/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Neuropeptide/genetics , Receptors, Pituitary Hormone-Regulating Hormone/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Tumor Cells, Cultured
12.
J Morphol ; 223(2): 191-201, 1995 Feb.
Article in English | MEDLINE | ID: mdl-29865304

ABSTRACT

Ultrastructural descriptions of the dipnoan heart are lacking. Many ultrastructural features of the heart of the Australian lungfish, Neoceratodus forsteri, resemble those of other lower vertebrates. The epicardial cells appear to be adapated for the exchange of material with the pericardial fluid. The most prominent features of the endocardial cells are numerous moderately electron-dense vesicles found within the cytoplasm. These organelles might have an endocrine function. The myocardiocytes are typically small. The banding pattern of the sarcomere is shared with most fish. The intercalated disc has a convoluted path and consists of desmosomes and fascia adherens. Caveolae are a prominent feature of the sarcoplasm. The sarcoplasmic reticulum is sparse, and T-tubules are lacking. Atrial myocardial dense bodies occur in vast numbers throughout the atrium and are occasionally seen in the ventricle. These vesicles are chromaffin-positive but fail to show catecholamine fluorescence. They are likely to contain peptides related to ANP. Subendothelial cells exhibiting catecholamine-specific fluorescence are scattered throughout the atrium. Ultrastructurally these cells contain many chromaffin-positive granules. Chromaffin cells represent another cell type with a probable endocrine function within the heart of N. forsteri. © 1995 Wiley-Liss, Inc.

SELECTION OF CITATIONS
SEARCH DETAIL
...