Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Biol Reprod ; 90(6): 140, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24829028

ABSTRACT

The luteal microenvironment is thought to direct the function of resident immune cells to facilitate either luteal function or regression. To determine if luteal cells from functional (Days 10-12) and regressing (8 h after administration of prostaglandin F2alpha) corpora lutea (CL) induce different responses in γδ T cells, luteal cells were cocultured with autologous γδ T cells isolated from peripheral blood. Proliferation, functional phenotypes, and cytokine synthesis were analyzed by flow cytometry. To determine if the luteal cells from functional CL induce hyporesponsiveness in γδ T cells, γδ(+) cells were cocultured with midcycle luteal cells and further stimulated with concanavalin A. Coculture of γδ(+) cells with midcycle luteal cells did not inhibit concanavalin A-induced proliferation. In a proliferation assay, luteal cells from midcycle CL predominantly induced proliferation of γδ(+) WC1(-) cells (P < 0.05), while luteal cells from regressing CL predominantly induced proliferation of γδ(+)WC1(+) cells (P < 0.05). Analysis of intracellular cytokines indicated that midcycle luteal cells increased the proportion of γδ(+) cells containing interleukin 10 (P < 0.05), but reduced the proportion of γδ(+) cells containing interferon gamma (IFNG; P < 0.05). There were no changes in the proportions of γδ(+) cells synthesizing interleukin 4 or tumor necrosis factor. Unexpectedly, coculture of γδ(+) cells with luteal cells from regressing CL had no effect on any of the cytokines analyzed. These data support the hypothesis that the function of resident T cells is differentially modulated depending on the status of the CL.


Subject(s)
Cattle Diseases/immunology , Cattle/immunology , Infertility/veterinary , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , Animals , Cell Communication/immunology , Corpus Luteum/cytology , Corpus Luteum/immunology , Cytokines/immunology , Cytokines/metabolism , Female , Fertility/immunology , Immunophenotyping , Infertility/immunology , Luteolysis/immunology , Luteolysis/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
2.
Reproduction ; 146(6): 527-37, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24019509

ABSTRACT

Secreted phosphoprotein 1 (SPP1) in the bovine corpus luteum (CL) regulates cell function during the transitional periods of luteinization and luteal regression. The objectives were to i) characterize SPP1 expression in the CL throughout the estrous cycle, ii) determine factors that regulate SPP1 expression in luteal cells, and iii) examine the role of SPP1 in lymphocyte chemotaxis, proliferation, and function. SPP1 mRNA was greater in fully functional (d10) CL and late cycle (d18) CL compared with developing (d4) CL. Additionally, SPP1 mRNA increased within 1 h and remained elevated 4 and 8 h following induction of luteolysis with prostaglandin (PG)F2α. Expression of the SPP1 receptor, ß3 integrin, was not different throughout the estrous cycle but decreased following induction of luteolysis. Expression of CD44 increased during the estrous cycle but did not change during luteal regression. In cultured luteal cells, SPP1 mRNA was upregulated by PGF2α and/or tumor necrosis factor α. Western blots revealed the presence of both full-length SPP1 and multiple cleavage products in cultured luteal cells and luteal tissue. Depletion of endogenous SPP1 did not hinder luteal cell-induced lymphocyte proliferation or lymphocyte phenotype but did inhibit lymphocyte migration toward luteal cells. Based on these data, it is concluded that SPP1 is initially activated to establish and maintain cellular interactions between steroidogenic and nonsteroidogenic cells during the development of the CL. Upon induction of luteolysis, SPP1 serves as a signaling molecule to recruit or activate immune cells to facilitate luteal regression and tissue degradation.


Subject(s)
Chemotaxis, Leukocyte/genetics , Corpus Luteum/immunology , Corpus Luteum/metabolism , Osteopontin/genetics , T-Lymphocytes/physiology , Animals , Cattle , Cell Proliferation/drug effects , Cells, Cultured , Chemotaxis, Leukocyte/drug effects , Corpus Luteum/cytology , Corpus Luteum/drug effects , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Gene Knockdown Techniques , Luteolysis/drug effects , Luteolysis/genetics , Luteolysis/immunology , Osteopontin/metabolism , RNA, Small Interfering/pharmacology , T-Lymphocytes/drug effects
3.
PLoS One ; 7(1): e29054, 2012.
Article in English | MEDLINE | ID: mdl-22235260

ABSTRACT

Prostaglandin F(2α) (PGF(2α)) induces luteolysis within a few days in cows, and immune cells increase in number in the regressing corpus luteum (CL), implying that luteolysis is an inflammatory-like immune response. We investigated the rapid change in polymorphonuclear neutrophil (PMN) numbers in response to PGF(2α) administration as the first cells recruited to inflammatory sites, together with mRNA of interleukin-8 (IL-8: neutrophil chemoattractant) and P-selectin (leukocyte adhesion molecule) in the bovine CL. CLs were collected by ovariectomy at various times after PGF(2α) injection. The number of PMNs was increased at 5 min after PGF(2α) administration, whereas IL-8 and P-selectin mRNA increased at 30 min and 2 h, respectively. PGF(2α) directly stimulated P-selectin protein expression at 5-30 min in luteal endothelial cells (LECs). Moreover, PGF(2α) enhanced PMN adhesion to LECs, and this enhancement by PGF(2α) was inhibited by anti-P-selectin antibody, suggesting that P-selectin expression by PGF(2α) is crucial in PMN migration. In conclusion, PGF(2α) rapidly induces the accumulation of PMNs into the bovine CL at 5 min and enhances PMN adhesion via P-selectin expression in LECs. It is suggested that luteolytic cascade by PGF(2α) may involve an acute inflammatory-like response due to rapidly infiltrated PMNs.


Subject(s)
Corpus Luteum/physiology , Dinoprost/pharmacology , Luteolysis/drug effects , Neutrophils/cytology , Neutrophils/drug effects , Animals , Cattle , Cell Adhesion/drug effects , Cell Count , Chemotaxis, Leukocyte/drug effects , Corpus Luteum/immunology , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Gene Expression Regulation/drug effects , Interleukin-8/genetics , Luteolysis/immunology , Neutrophils/metabolism , P-Selectin/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Time Factors
4.
J Reprod Immunol ; 93(1): 28-37, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22186103

ABSTRACT

Knowledge on the regulation of corpus luteum (CL) function in the mare is scarce. In this study, the presence of cytokines tumor necrosis factor alpha (TNF) and interferon gamma (IFNG), and their receptors (TNFRI, TNFRII and IFNRI), was investigated in equine CL throughout the luteal phase. The effects of TNF and IFNG on secretory function and viability of luteal cells were defined in vitro. Cytokine ligands and receptors were present in steroidogenic and endothelial cells. Protein expression for TNF was greater in mid-phase and regressing CL, while TNFRI was increased in regressing CL and TNFRII did not change. IFNG and IFNRI showed the highest expression in regressing CL. Transcription of mRNA for TNF increased from mid-phase to regressing CL and both TNFRI and TNFRII decreased from early to regressing CL. Transcription of mRNA for IFNG was lower in CL from early phase than in mid or regressing luteal phase, while IFNRI expression was not changed. In the early CL, TNF acted to increase P(4) and PGE(2) but decrease PGF(2α) secretion. In the mid luteal phase, TNF increased PGF(2α) secretion and TNF+IFNG decreased PGE(2) secretion. In the regressing luteal phase, TNF, IFNG and TNF+IFNG decreased P(4) and PGE(2) secretion, but TNF and TNF+IFNG increased PGF(2α) secretion by luteal cells. Cell viability was reduced by TNF+IFNG in regressing CL. These data show the presence of cytokines TNF and IFNG, and their receptors, in the equine CL and indicate their potential involvement in regulation of luteal function.


Subject(s)
Autocrine Communication , Corpus Luteum/immunology , Horses/immunology , Interferon-gamma/metabolism , Luteolysis/genetics , Paracrine Communication , Tumor Necrosis Factor-alpha/metabolism , Animals , Cells, Cultured , Corpus Luteum/pathology , Dinoprost/genetics , Dinoprost/metabolism , Dinoprostone/genetics , Dinoprostone/metabolism , Female , Gene Expression Regulation/immunology , Interferon-gamma/genetics , Luteal Cells/immunology , Luteal Cells/metabolism , Luteal Cells/pathology , Luteolysis/immunology , Oligopeptides/genetics , Oligopeptides/metabolism , Receptors, Interferon/genetics , Receptors, Interferon/metabolism , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Receptors, Tumor Necrosis Factor, Type II/genetics , Receptors, Tumor Necrosis Factor, Type II/metabolism , Tumor Necrosis Factor-alpha/genetics
5.
Mol Cell Endocrinol ; 338(1-2): 38-45, 2011 May 16.
Article in English | MEDLINE | ID: mdl-21371521

ABSTRACT

Progress in understanding the background of structural luteolysis depends on insights into the physiological function of innate immunity (INIM), in particular the presence of dendritic cells (DCs) in the corpus luteum (CL). For this reason, the cultures of five endothelial cell-like phenotypes derived from the bovine CL and their long-lasting analysis (morphology, function, and origin) become important. Types 1 and 2 represent microvascular endothelial cells with cytokeratin (CK) expression, assumed to be danger-sensing cells. Types 3 and 4 express features of common endothelial cells. Type 5 indicates a steroidogenic cell type, which could be derived from steroidogenic CK(+) cells in the CL of development after loss of CK expression. Type 5 is a promising candidate to become a mature DC. It might act with the microvascular CK(+) cell/type 1 like a luteovascular unit, which connects INIM with adaptive/cell-mediated immunity (ADIM) in structural luteolysis.


Subject(s)
Corpus Luteum/cytology , Dendritic Cells/immunology , Endothelial Cells/metabolism , Luteolysis/genetics , Animals , Cattle , Corpus Luteum/blood supply , Corpus Luteum/immunology , Endothelial Cells/immunology , Female , Immunity, Innate , Keratins/metabolism , Luteolysis/immunology , Microvessels/cytology , Microvessels/immunology , Phenotype
6.
Am J Reprod Immunol ; 64(4): 275-86, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20712810

ABSTRACT

The dynamic changes that characterize the female reproductive system are regulated by hormones. However, local cell-to-cell interactions may mediate responsiveness of tissues to hormonal signals. The corpus luteum (CL) is an excellent model for understanding how immune cells are recruited into tissues and the role played by those cells in regulating tissue homeostasis or demise. Leukocytes are recruited into the CL throughout its lifespan, and leukocyte-derived cytokines have been found in corpora lutea of all species examined. The proinflammatory cytokines inhibit gonadotropin-stimulated steroidogenesis, profoundly stimulate prostaglandin synthesis by luteal cells, and promote apoptosis. However, there is mounting evidence that leukocytes and luteal cells communicate in different ways to maintain homeostasis within the functional CL. Domestic animals have provided important information regarding the presence and role of immune cells in the CL.


Subject(s)
Animals, Domestic/physiology , Corpus Luteum/immunology , Corpus Luteum/physiology , Leukocytes/physiology , Models, Animal , Ovary/physiology , Animals , Animals, Domestic/immunology , Apoptosis , Cell Communication , Corpus Luteum/cytology , Cytokines/metabolism , Female , Homeostasis , Humans , Luteal Cells/physiology , Luteinization , Luteolysis/immunology , Ovarian Follicle/physiology , Ovary/immunology , Ovulation , Pregnancy
7.
Biol Reprod ; 70(2): 473-80, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14561657

ABSTRACT

The process of luteolysis requires very subtly modulated coordination of different factors and regulation systems. Immune cells and cytokines were shown to be relevant for bovine luteolysis. The aim of this study was to investigate the detailed pattern of mRNA expression of the pro-inflammatory cytokines tumor necrosis factor alpha (TNFalpha), TNF receptor type 1 (TNF-R1), interleukin 1beta (IL-1beta), and interferon gamma (IFNgamma), and of the inducible nitric oxide synthase (iNOS) and the basic fibroblast growth factor (FGF-2) during prostaglandin (PG) F(2alpha)-induced luteolysis in the bovine corpus luteum (CL). In addition, the mRNA expression for the LH-receptor (LH-R) and the steroidogenic enzyme p450scc was determined. Cows in the midluteal phase (Days 8-12) were injected with the PGF(2alpha) analogue cloprostenol, and CL were collected by transvaginal ovariectomy before and 2, 4, 12, 48, and 64 h after PGF(2alpha) injection. Conventional and real-time reverse transcription polymerase chain reaction RT-PCR (LightCycler) using SYBR Green I detection were employed to determine the mRNA expression for the investigated factors. All cytokines were significantly up-regulated during induced luteolysis. LH-R and p450scc mRNA were down-regulated (P < 0.05) during structural luteolysis (after 12 h), and p450scc in addition at 2 h after PGF(2alpha) (P < 0.05). FGF-2 expression increased (P < 0.001) during functional luteolysis (until 12 h after PGF(2alpha)) and diminished thereafter. The mRNA expression for iNOS decreased (P < 0.05) after induction of luteolysis. In conclusion, cytokines may be involved not only in structural but also in functional luteolysis and the deprivation of luteal survival factors, leading to a situation where apoptosis can occur. FGF-2 may participate in the suppression of cytokine-induced iNOS mRNA expression and in the prevention of an inflammatory reaction in the surrounding tissues.


Subject(s)
Corpus Luteum/immunology , Cytokines/genetics , Dinoprost/genetics , Fibroblast Growth Factor 2/genetics , Luteolysis/immunology , Animals , Antigens, CD/genetics , Cattle , Cholesterol Side-Chain Cleavage Enzyme/genetics , Female , Gene Expression/immunology , Interferon-gamma/genetics , Interleukin-1/genetics , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Progesterone/blood , RNA, Messenger/analysis , Receptors, LH/genetics , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor, Type I , Tumor Necrosis Factor-alpha/genetics
8.
J Exp Zool A Comp Exp Biol ; 296(2): 152-9, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12658721

ABSTRACT

Immune cells, which constitute a significant cell mass in the corpora lutea (CLs), are considered to play critical roles in luteolysis, but the details are not fully understood. We histochemically investigated the changes in distribution and cell density of macrophages and T lymphocytes and in tumor necrosis factor (TNF)-alpha and interferon (IFN)-gamma, which can induce apoptosis in the luteal cells in murine CLs during luteal regression. No macrophages or T lymphocytes were observed in functionally healthy CLs. Abundant macrophages and increasing T lymphocytes were demonstrated in CLs at the functional regression stage (early stage of regression). At the structural regression stage (late stage of regression), abundant T lymphocytes but no macrophages were demonstrated in the CLs. A moderate amount of TNF-alpha was detected in all CLs at all stages. No IFN-gamma was detected in either healthy or early regressing CLs, but a large amount of IFN-gamma was detected at the late regression stage. Moreover, in cultured luteal cells, reactivity against Fas-ligand (FasL) was caused by pretreatment with TNF-alpha and IFN-gamma and apoptosis was induced by FasL treatment. These findings support the hypothesis that macrophages initiate T lymphocyte aggregation at the early stage of luteal regression, and then T lymphocytes induce apoptosis on luteal cells, which in turn develop sensitivity against FasL by TNF-alpha and IFN-gamma.


Subject(s)
Corpus Luteum/cytology , Corpus Luteum/immunology , Interferon-gamma/metabolism , Luteolysis/physiology , Macrophages/metabolism , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Apoptosis/drug effects , Cell Count , Cell Survival/drug effects , Cells, Cultured , Corpus Luteum/drug effects , Fas Ligand Protein , Female , Immunohistochemistry , Interferon-gamma/pharmacology , Luteolysis/immunology , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/pharmacology , Mice , Tumor Necrosis Factor-alpha/pharmacology
9.
Reproduction ; 122(5): 665-76, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11690526

ABSTRACT

The corpus luteum produces progesterone, which is essential for the maintenance of pregnancy. In the absence of a viable embryo, the corpus luteum must regress rapidly to allow for development of new ovulatory follicles. In many species, luteal regression is initiated by uterine release of PGF(2alpha), which inhibits steroidogenesis and may launch a cascade of events leading to the ultimate demise of the tissue. Immune cells, primarily macrophages and T lymphocytes, are present in the corpus luteum, particularly at the time of luteolysis. The macrophages are important for ingestion of cellular remnants that result from the death of luteal cells. However, it has also been hypothesized that immune cells are involved directly in the destruction of luteal cells, as well as in the loss of steroidogenesis; this hypothesis is reviewed in the first part of this article. An alternative hypothesis is also presented, namely that immune cells serve to abate an inflammatory response generated by dead and dying luteal cells, in effect, preventing a response that would otherwise damage surrounding ovarian tissues. Finally, the changes in immune cells that accompany maternal recognition of pregnancy and rescue of the corpus luteum are discussed briefly. Inhibition of immune cells in the corpus luteum during early pregnancy may be due to embryonic or uterine signals, or to maintenance of high progesterone concentrations within the luteal tissue.


Subject(s)
Corpus Luteum/physiology , Luteolysis/immunology , Macrophages/immunology , Mammals/immunology , Models, Immunological , T-Lymphocytes/immunology , Animals , Apoptosis/physiology , Corpus Luteum/metabolism , Cytokines/physiology , Dinoprost/metabolism , Embryo Implantation/immunology , Female , Immunity, Cellular , Phagocytosis/physiology , Pregnancy , Progesterone/biosynthesis , Progesterone/physiology
10.
J Reprod Fertil ; 115(1): 87-96, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10341726

ABSTRACT

Immune cells and their cytokine products have powerful local effects within body tissues. There has been great interest in the potential role of these cells, not only during destruction of the corpus luteum but also during its functional lifespan. In this study, lymphocytes, macrophages and major histocompatibility complex class II molecules were quantified using immunohistochemistry and the reverse transcription-polymerase chain reaction was used to detect mRNA for tumour necrosis factor alpha and interferon gamma within corpora lutea from three groups of cows: (1) corpora lutea collected at an abattoir and assessed visually into four stages (stage I (days 1-5), stage II (days 6-12), stage III (days 13-18) and stage IV (days 19-21) of the oestrous cycle); (2) corpora lutea collected around natural luteolysis (days 14-20); and (3) corpora lutea collected 6, 12 and 24 h after prostaglandin F 2 alpha-induced luteolysis. The numbers of T lymphocytes (CD5+ and CD8+) were significantly higher (P < 0.05) at stage IV and from day 16 onwards, before functional luteolysis. There were significantly higher numbers (P < 0.01) of macrophages at stages I, III and IV compared with stage II in visually staged tissue. Major histocompatibility complex class II molecules were increased (P < 0.05) at stages I and IV compared to stage II and at all times after induced luteolysis. Using reverse transcription-polymerase chain reaction, mRNA encoding tumour necrosis factor alpha and interferon gamma was detected in all luteal tissue collected around natural luteolysis and after induced luteolysis. These findings, particularly the increase in T lymphocytes before functional luteolysis, provide further evidence of a significant role for the immune system in affecting reproductive function in cows.


Subject(s)
Cattle/immunology , Corpus Luteum/immunology , Cytokines/biosynthesis , Estrus/immunology , Animals , Cell Count , Female , Histocompatibility Antigens Class II/analysis , Immunohistochemistry , Interferon-gamma/genetics , Luteolysis/immunology , Macrophages/cytology , Pregnancy , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Statistics, Nonparametric , T-Lymphocytes/cytology , Tumor Necrosis Factor-alpha/genetics
11.
Hum Reprod ; 13(8): 2186-91, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9756294

ABSTRACT

We investigated, using an image analysis system, the immunohistochemical localization of leukocyte subpopulations and human leukocyte antigen (HLA)-DR in 30 normal-cycling human ovaries in order to better understand local immunological events in human ovaries. All subtypes of T lymphocytes examined (CD3+, CD4+ and CD8+ cells) were sporadically observed in the stroma and theca layers of follicles throughout the menstrual cycle (ranging from 4.32 to 6.25 cells/10(-7) m2, 1.67 to 3.33 cells/10(-7) m2 and 2.33 to 3.44 cells/10(-7) m2, respectively for the three subtypes), and subsequently, increased in number in atretic follicles (78.70 +/- 6.90, 31.13 +/- 2.54 and 43.31 +/- 3.35). After ovulation, the number of T lymphocytes was markedly low in the early and mid corpus luteum (13.88 +/- 1.62, 4.18 +/- 0.50 and 6.53 +/- 0.45). The number increased in the late corpus luteum, and was highest in the late degenerating corpus luteum (255.67 +/- 27.10, 102.12 +/- 7.80 and 137.34 +/- 12.50). HLA-DR was sporadically positive in fibroblasts in the stroma and theca layers of follicles (means ranged from 1.25 to 1.82 cells/10(-7) m2), and increased in atretic follicles (24.68 +/- 2.25). HLA-DR+ cells were markedly low in the early and mid corpus luteum (2.16 +/- 0.88), increased in the late corpus luteum, and reached a plateau in the late degenerating corpus luteum (121.84 +/- 17.73). The great majority of these increased HLA-DR+ cells were macrophages. Results of our study suggest that T lymphocytes and/or macrophages play important roles in luteal regression and follicular atresia in normal-cycling human ovaries.


Subject(s)
Leukocytes/cytology , Leukocytes/immunology , Ovary/cytology , Ovary/immunology , Adult , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Female , Follicular Atresia/immunology , HLA-DR Antigens/metabolism , Humans , Immunohistochemistry , Luteolysis/immunology , Macrophages/cytology , Macrophages/immunology , Menstrual Cycle/immunology , Middle Aged , Neutrophils/cytology , Neutrophils/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology
12.
Am J Reprod Immunol ; 36(6): 327-41, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8985508

ABSTRACT

PROBLEM: We have recently observed that the regression of corpora lutea (CL) in women during the reproductive period of life is accompanied by a diminution of Thy-1 differentiation protein release from vascular pericytes and an accumulation of T lymphocytes and activated macrophages among both degenerating granulosa lutein cells (GLC) and theca lutein cells. These data suggest that the immune system and other stromal factors, representing components of the "tissue control system," may play a role in regression of the CL. We investigated degenerating CL from climacteric women to address the possibility that the decline of immune functions with advancing age may result in incomplete regression of luteal tissue. This could contribute to the altered hormonal profiles and abnormal uterine bleeding that frequently occur during the climacteric. METHOD: Immunoperoxidase staining and image analysis were used to localize Thy-1 differentiation protein of vascular pericytes, cytokeratin staining of GLC, neural cell adhesion molecule expression by theca lutein cells, CD15 of neutrophils, CD4, CD14, CD68, and leukocyte common antigens of macrophages, and CD3 and CD8 determinants of T lymphocytes. We also investigated the expression of luteinizing hormone receptor (LH receptor) and mitogen activated protein kinases (MAP kinases) in luteal cells. Samples of regressing luteal tissue were obtained during the follicular phase from perimenopausal women (age 45-50) who exhibited prolonged or irregular cycles. For comparison, luteal tissues from women with regular cycles (age 29-45) and CL of pregnancy were also investigated. RESULTS: Corpora lutea of the climacteric women exhibited irregular regression of luteal tissue characterized by a lack of cytoplasmic vacuolization and nuclear pyknosis in GLC, and by a persistence of theca lutein cells exhibiting hyperplasia and adjacent theca externa layers. This was accompanied by a continuing release of Thy-1 differentiation protein from vascular pericytes. Persisting GLC lacked surface expression of macrophage markers (CD4, CD14, CD68 and leukocyte common antigen) as well as nuclear granules exhibiting CD15 of neutrophils, detected in regularly regressing GLC. In addition, such persisting GLC showed weak or no LH receptor expression, and retained the expression of cytokeratin. They also exhibited enhanced staining for MAP kinases. Strong cytoplasmic MAP kinase expression with occasional nuclear translocation was also detected in persisting theca lutein cells, indicating high metabolic activity of these cells. T lymphocytes, although occasionally present in luteal stroma within luteal convolutions, did not invade among persisting GLC and were virtually absent from layers of theca externa and theca lutein cells. CONCLUSIONS: These data indicate that the regressing CL in climacteric women may exhibit persistence of luteal cells, perhaps because of age-induced alterations of the immune system and other local stromal homeostatic mechanisms involved in the elimination of luteal cells. Persisting GLC and/or theca lutein cells may exhibit abnormal hormonal secretion that contributes to the alteration of target tissues, such as the endometrium, resulting in abnormal uterine bleeding, hyperplasia, and neoplasia.


Subject(s)
Aging/immunology , Climacteric/immunology , Luteolysis/immunology , Organ Specificity/immunology , Adult , Antigens, CD/analysis , Antigens, Differentiation, Myelomonocytic/analysis , Endometrium/immunology , Female , Humans , Immunohistochemistry , Keratins/analysis , Lewis X Antigen/analysis , Middle Aged , Neural Cell Adhesion Molecules/analysis , Ovary/immunology , Thy-1 Antigens/analysis
13.
Biol Reprod ; 53(6): 1373-84, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8562694

ABSTRACT

Factors determining the life span of the human corpus luteum (CL) are not known. In addition to being determined by hormonal factors, such as hCG, the life of luteal cells may be determined by the preservation of luteal vascularization. Furthermore, the CL represents an immunologically unique tissue, as it is formed after menarche, long after adaptation of the immune system toward self. Thus, CL regression may be immunologically mediated. To determine what role the vasculature and immune system play in human CL development and regression, we examined immunohistochemically 1) the expression of Thy-1 differentiation protein by vascular pericytes, 2) the expression of major histocompatibility complex (MHC) class I and class II molecules in granulosa lutein cells (GLC), and 3) infiltration of the CL by macrophages and T lymphocytes. LH receptor (LHR) and cytokeratin 18 expression were also studied. In developing CL, the pericytes of luteal microvasculature released Thy-1 differentiation protein among the endothelial cells of proliferating vessels. In mature CL, Thy-1 released from vascular pericytes accumulated on the surface of GLC, and these cells exhibited LHR immunoreactivity (LHRI). Overall LHRI increased during the luteal phase and was strongest at the beginning of the late luteal phase. Although vascular pericytes showed strong LHRI, no staining of endothelium was detected during the luteal phase. GLC exhibited strong cytokeratin staining and moderate staining for MHC class I and MHC class II antigens; numerous macrophages were detected in luteal tissue. During pregnancy, the staining pattern was similar to that seen in the mature CL at the end of the midluteal phase. During the late luteal phase, surface expression of MHC class I and MHC class II antigens by GLC was substantially enhanced, and some T cells invaded among luteal cells. By the end of the cycle, an acute regression of vasculature and luteal tissue was observed along the fibrous septa. The remaining GLC showed only surface and no cytoplasmic LHRI. During the subsequent cycle, in the presence of numerous T cells, regressing GLC exhibited strong surface expression of various macrophage markers, such as CD4, CD14, CD68, and leukocyte common antigen, a feature not detected in the CL during the luteal phase nor described in other tissues. A complete loss of cytokeratin staining in GLC was observed. In regressing CL, strong LHRI was present in the endothelium of small and large luteal vessels. In conclusion, vascular pericytes and macrophages may stimulate the development and senescence of luteal tissue. The senescence of GLC may be inconsistent with preservation of luteal vasculature, and T lymphocytes appear to participate in terminal regression of the CL. Regression of luteal tissue therefore resembles immunologic rejection of a transplant. During pregnancy, the aging process of GLC appears to be interrupted, possibly due to the temporary acceptance of the CL "graft."


Subject(s)
Immunity , Luteolysis/immunology , Receptors, LH/analysis , Adult , Corpus Luteum/blood supply , Corpus Luteum/chemistry , Corpus Luteum/immunology , Endothelium, Vascular/chemistry , Female , Granulosa Cells/chemistry , Granulosa Cells/immunology , Histocompatibility Antigens Class I/analysis , Histocompatibility Antigens Class II/analysis , Humans , Immunoenzyme Techniques , Keratins/analysis , Macrophages/immunology , Middle Aged , Pregnancy , T-Lymphocytes/immunology , Thy-1 Antigens/analysis
14.
J Reprod Fertil Suppl ; 49: 365-77, 1995.
Article in English | MEDLINE | ID: mdl-7623327

ABSTRACT

Primary cultures of luteal cells have been used to determine both acute and chronic effects of cytokines on luteal cell function and viability. Gonadotrophin-stimulated progesterone production is inhibited by interleukin 1 beta (IL-1 beta), tumour necrosis factor alpha (TNF-alpha), or gamma-interferon (IFN-gamma), the last two cytokine being more effective than IL-1. In contrast, all three cytokines are potent stimulators of prostaglandin production by these cells. The mechanism by which prostaglandin synthesis is enhanced may differ slightly for each cytokine. In luteal cells, TNF-alpha appears to act primarily through stimulation of phospholipase A2, whereas IL-1 beta may activate phospholipase C and prostaglandin endoperoxide synthase (PGS) in addition to phospholipase A2. The mechanism of action of IFN-gamma has not yet been determined. In addition to the observed functional effects, cytokines may also promote cell death during luteal regression. Although the three cytokines mentioned have little or no effect on viability of cultured luteal cells when administered separately, combined treatment with TNF-alpha and IFN-gamma results in a substantial decrease in the number of viable cells. Inhibition of cytokine-stimulated prostaglandin production does not alter the cytotoxic effect of these cytokines. Expression of major histocompatibility (MHC) class I molecules on luteal cells is enhanced, and MHC class II molecules are induced, by exposure to IFN-gamma. This is especially intriguing, as MHC class II expression increases before luteal regression in vivo, and is suppressed in early pregnancy. In summary, evidence is rapidly accumulating that supports the hypothesis that the function or structural integrity of luteal cells may be modulated by resident immune cells. Future research will probably address how these local events are hormonally controlled, and if they can be modified to regulate corpus luteum function.


Subject(s)
Corpus Luteum/immunology , Cytokines/physiology , Luteolysis/immunology , Lymphocytes/physiology , Animals , Cattle , Female , Progesterone/physiology , Sheep
15.
J Reprod Fertil ; 96(1): 347-53, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1432967

ABSTRACT

The potential role of macrophages and T lymphocytes in the destruction of the corpus luteum at the end of the luteal phase was investigated by treating pseudopregnant rabbits with the immunosuppressant glucocorticoid methylprednisolone. Eleven specific pathogen-free New Zealand White rabbits were injected with pregnant mares' serum gonadotrophin (40 iu, i.m.), followed 2 days later by human chorionic gonadotrophin (40 iu, i.v.) to stimulate ovulation. The following day (day 1 of pseudo-pregnancy) all animals had an oestradiol-filled Silastic capsule implanted s.c., to ensure that oestradiol, the luteotrophic hormone in this species, would not be limiting. From day 10 of pseudopregnancy, three animals were injected with a low dose of methylprednisolone (2 mg kg-1 per day) until day 20. Three other animals were injected with a higher dose of methylprednisolone (20 mg kg-1 per day) from day 13 of pseudopregnancy until day 19. Five animals served as control, vehicle-injected animals. Blood samples were taken at intervals and assayed for progesterone. Immunofluorescence was used to stain luteal tissue for macrophages, T lymphocytes and class II antigens, and positive cells were counted under high-power magnification. Methylprednisolone treatment reduced (by about 70%), but did not eliminate, the macrophages in the regressing corpora lutea. In contrast, the high dose of methylprednisolone essentially eliminated T lymphocytes, and reduced (by about 90%) the number of cells expressing class II antigen in the luteal tissue. Despite the effects of methylprednisolone on these cells, serum progesterone profiles were not altered by treatment with methylprednisolone, and pseudopregnancy was of normal duration.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Corpus Luteum/immunology , Luteolysis/immunology , Macrophages/immunology , Methylprednisolone/pharmacology , Pseudopregnancy/immunology , T-Lymphocytes/immunology , Animals , Female , Fluorescent Antibody Technique , Immunosuppression Therapy/methods , Progesterone/blood , Pseudopregnancy/blood , Rabbits
SELECTION OF CITATIONS
SEARCH DETAIL
...