Endocrinology Basic and Clinical Principles - part 3 pot

45 305 0
Endocrinology Basic and Clinical Principles - part 3 pot

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

78 Part II / Hormone Secretion and Action through heterotrimeric G proteins and other specialized signaling partners that are integral to plasma membrane. In the case of some hormonal responses, interaction at the surface membrane may itself be sufficient to elicit an alteration in cell function. For example, estra- diol can directly stimulate PKC activity in membranes isolated from chondrocytes, and the steroid also modu- lates calcium-dependent eNOS activity associated with its receptor in isolated plasma membranes from endothelial cells. Moreover, estrogens may enhance growth of mammary tumor cells, largely independent of ERE-dependent transcription, by stimulating mem- brane-associated MAPK pathways. In ER-negative cells, transfection of transcriptionally inactive, mutant forms of ERα allows full stimulation of DNA synthe- sis by estradiol. Ligand-independent activation of ste- roid hormone receptors also occurs and may represent a more primitive response pathway, whereby cross- communication with peptide signaling systems in the cell can directly modulate the activity of steroid hor- mone receptors. For example, ER can be activated in the absence of estradiol through phosphorylation by Fig. 8. Schematic representation of time course of responses of uterus to E 2 . Times shown on the logarithmic scale refer to the onset of unequivocal change from baseline values. Thus, times indicated are dependent in part on sensitivities of the various analytic methods applied and on the somewhat arbitrary selection of initial time points for observation in the several experimental protocols. GMP = guanosine 5´-monophosphate; PI 3 -Kinase = phosphatidylinositol 3-kinase. (Reprinted with permission from Szego and Pietras [1984], wherein additional details and sources are given.) Chapter 5 / Plasma Membrane Receptors for Steroids 79 EGF-stimulated MAPK. These signaling pathways may also allow for phosphorylation of important regu- latory molecules, such as the steroid receptor coacti- vators and corepressors that play a crucial role in steroid hormone action. As is readily appreciated, the shuttling of phosphate groups in and out of proteins critical to the signal transduction cascade is a powerful means of modifying their structure, with the immedi- ate result of altering their folding patterns and/or rela- tive degree of their interactions with neighboring molecules. Thus, such apparently minimal changes in composition provide a means of augmenting or attenu- ating their catalytic functions in a virtually instanta- neous manner. It is not surprising that this efficient mechanism is so widely conserved in so many biologic contexts and across so wide an evolutionary spectrum. Any comprehensive model of steroid hormone action must account for these important cellular interactions. Accordingly, the functions of the surface membrane– associated receptors are likely twofold. Both lead to coordination of the activities of more downstream cel- lular organelles. One such function is complementary to the more distal and time-delayed events at the genome, through communication of information from the extra- cellular compartment. The second function supplements the more deferred and metabolically expensive activi- ties at the genome, through exclusion of the latter. Instead, the cascade of signals, transduced from binding of steroids at the cell surface, are themselves converted into immediate and more readily reversible stimuli, such as those eliciting acute ion shifts and changes in vaso- motor dynamics—these being of evolutionary signifi- cance for survival. In the case of some hormone responses, such primary interactions at the plasma mem- brane may be sufficient to trigger a cascade of intracel- lular signals that lead to specifically altered cell function. Thus, within seconds of estrogen binding by surface receptors of the target cell, widespread changes are communicated to the cytoarchitecture involving striking alterations in the localized assembly and disas- sembly of the microtubule-microfilament scaffolding of the cell. These abrupt, but transitory, changes in the subcellular cytoskeleton may allow enhanced exchanges between membrane and nuclear compart- ments to promote redistribution of matériel in the pretranscriptional stage of the estrogen response cas- cade. Indeed, these dual capacities of membrane recep- tor activation underlie adaptation of the target cell to processing of information from its external environment on two independent/synergistic pathways: acute and delayed. Some selected examples of the role of mem- brane receptors for steroids in health and disease states are provided in the following sections. 5. MEMBRANE-ASSOCIATED STEROID RECEPTORS IN HEALTH AND DISEASE 5.1. Estrogen Receptors in Bone, Neural, Cardiovascular, and Reproductive Health and in Malignancy As with other steroid hormones, biologic activities of estrogen in breast, uterus, and other target tissues have long been considered to be fully accounted for through activation of a specific high-affinity receptor in cell nuclei. However, it is well established that estrogen can trigger in target cells rapid surges in the levels of intracellular messengers, including calcium and cAMP, as well as activation of MAPK and phospholipase (Table 1). These data have led to a growing consensus that the conventional genomic model does not explain the rapid effects of estrogen and must be expanded to include plasma membrane receptors as essential com- ponents of cellular responsiveness to this and other ste- roid hormones. The first unequivocal evidence for specific mem- brane binding sites for E 2 was reported about 25 yr ago. Intact uterine endometrial cells equipped with ER, but not ER-deficient, control cells became bound to an inert support with covalently linked E 2 . In addition, target cells so bound could be eluted selectively by free hor- mone, in active form, but not by the relatively inactive estradiol-17α; and cells so selected exhibited a greater proliferative response to estrogens than cells that did not bind. Further investigations have continued to provide compelling evidence for the occurrence of a plasma membrane form of ER and support for its role in medi- ating hormone actions (Table 1). Selye first demonstrated that steroids at pharmaco- logic concentrations elicit acute sedative and anes- thetic actions in the brain. However, electrical responses to physiologic levels of E 2 with rapid onset have since been reported in nerve cells from various brain regions. Estrogen has diverse effects on brain functions, including those regulating complex activi- ties, such as hypothalamic-pituitary circuits, cognition, and memory. Some of these estrogenic actions may be attributable to regulation of cAMP signaling by G pro- tein–coupled plasma membrane receptors for the hor- mone. New caveats from randomized controlled clinical tri- als on the increased risk of cardiovascular disease among healthy postmenopausal women prescribed estrogen plus progestin conflict with the long-held belief that hormone therapy might reduce a woman’s risk of coro- nary heart disease. The results of basic research and animal models had suggested the hypothesis that estro- gens were beneficial for cardiovascular health. It is 80 Part II / Hormone Secretion and Action likely that variations in the dose, type or timing of estro- gens or the coadministration of progestogens modifies the final physiologic and clinical responses to estrogen, and these clinical variables may account for differ- ences from the preceding laboratory and observational research studies. However, these clinical trial results also suggest that further understanding of the molecular and cellular determinants of estrogen action are required. Traditional genomic models of estrogen action in the vasculature are incomplete, but, with knowledge of the full spectrum of steroid hormone action in target cells, researchers may yet find ways to manipulate estrogenic actions that promote cardiovascular health. One starting point is to recognize that estrogen has both rapid and long-term effects on the blood vessel wall. Certain vasoprotective effects of estrogen are mediated by mem- brane-associated receptors. Estrogen-induced release of uterine histamine in situ has long been associated with rapid enhancement of the microcirculation by a process that excludes gene activation. Reinforcing these obser- vations are data detailing the role of nitric oxide (NO) in vascular regulation by estrogen. Normal endothelium secretes NO, which relaxes vascular smooth muscle and inhibits platelet aggregation. Estrogens elicit abrupt lib- eration of NO by acute activation of eNOS without al- tering gene expression, a response that is fully inhibited by concomitant treatment with specific ER antagonists. This estrogenic effect is mediated by a receptor local- ized in caveolae of endothelial cell membranes. Mani- pulation of rapid estrogen signaling events may provide new approaches in the medical management of cardio- vascular health. Direct effects of estrogen on the vascu- lature promote acute vasodilation and may contribute to late effects leading to inhibition of the development and progression of atherosclerosis. E strogen deficiency is associated with significant bone loss and is the main cause of postmenopausal osteo - porosis, a disorder that affects about one-third of the postmenopausal female population. When estrogen is diminished, bone turnover increases, and bone resorp- tion increases more than bone formation, leading to net bone loss. In randomized clinical trials, administration of estrogen plus progestin in healthy postmenopausal women increases bone mineral density and reduces the risk of fracture. However, in considering the effects of combined hormonal therapy on other important disease outcomes, such as the risk of ovarian and breast cancer, caution is recommended in the use of continuous com- bined hormonal therapy in the clinic. Hence, the role of membrane ER in regulating bone mass has had increas- ing research emphasis and could promote development of alternative treatments. Evidence for membrane-bind- ing sites and acute effects of estrogen with an onset within 5 s has been observed in both osteoblasts and osteoclasts. The effects of estrogens on bone homeosta- sis also appear to involve rapid activation of MAPK, as has been demonstrated in certain other target cells. Indeed, the “classic” genotropic effects of estrogens may be dispensable for their bone-protective effects. A novel synthetic ligand, 4-estren-3α,17β-diol, stimulates transcription-independent signaling of estrogens and increases bone mass and tensile strength in ovariecto- mized mice. Such therapeutic agents targeted to mem- brane-associated receptor forms may play a role in future treatment and prevention of osteoporosis and may offer an alternative to hormone replacement therapy for this indication. Similar considerations may apply in the case of poor patient tolerance of compounds related to etidronate and calcitonin. Estrogen stimulates the proliferation of breast epi- thelial cells, and endogenous and exogenous estrogens, as well as related synthetic compounds, have been implicated in the pathogenesis of breast cancer. Human breast cancer cells exhibit specific plasma membrane reactivity with antibodies directed to the nuclear form of ERα. In addition, breast cancer cells with these membrane-associated ERs show rapid responses to estradiol, including significant increments in MAPK and phosphatidylinositol 3-kinase (PI3K)/Akt kinase, enzymic molecules that are crucial in the regulation of cell proliferation and survival. There are current indi- cations that these membrane receptors may associate with HER-2/neu growth factor receptors in lipid raft subdomains of plasma membrane and promote tumor growth. Such signaling complexes may offer a new strategy for therapeutic intervention in patients afflicted with breast cancer. 5.2. Progestogen and Androgen Receptors in Reproduction and Malignancy As documented for estrogens, several physiologic effects of progestogens and androgens appear to be regulated, in part, by membrane-associated receptors. Progesterone controls several components of reproduc- tive function and behavior. Some of these activities are mediated by interaction with neurons in specific brain regions, and membrane effects appear to be important in this process. Meiosis in amphibian oocytes is initi- ated by gonadotropins, which stimulate follicle cells to secrete progesterone. The progesterone-induced G 2 /M transition in oocytes was among the first convincing examples of a steroid effect at plasma membrane, since it could be shown that exogenous, but not intracellu- larly injected, progesterone elicited meiosis and that many progesterone-stimulated changes occurred even in enucleated oocytes. Moreover, this process may be Chapter 5 / Plasma Membrane Receptors for Steroids 81 related to progesterone-induced increments in intracel- lular Ca 2+ and release of diacylglycerol (DAG) species that elicit a cascade of further lipid messengers. Progesterone elicits rapid effects on the activity of second messengers and the acrosome reaction in human sperm. Assay of acute sperm responses to progesterone in subfertile patients is highly predictive of fertilization capacity. Effects of the steroid, present in the cumulus matrix surrounding the oocyte, are mediated by elevated intracellular Ca 2+ , tyrosine phosphorylation, chloride efflux, and stimulation of phospholipases, effects at- tributed to activation of a membrane-initiated pathway. Indeed, two different receptors for progesterone, appar- ently distinct from genomic ones, have been identified at the surface of human spermatozoa; nevertheless, a monoclonal antibody against the steroid-binding domain of human intracellular progesterone receptor inhibits progesterone-induced calcium influx and the acrosome reaction in sperm. As with estrogens and progestogens, androgens pro- mote rapid increase in cytosolic Ca 2+ in their cellular targets. Other effects of androgens that are not attribut- able to genomic activation include acute stimulation of MAPK in prostate cancer cells, an action that may be important for promoting their growth. It has been dem- onstrated in fibroblasts that androgens can stimulate membrane-initiated signaling and the onset of DNA synthesis without activation of “classic” androgen receptor (AR)-dependent gene transcription pathways. Rather, other signaling pathways, such as those modu- lated by MAPK, may be operative in androgen-induced stimulation of DNA synthesis. Such observations have important implications for understanding the regulation of cell proliferation in steroid target tissues. The androgen 5β-dihydrotestosterone induces vaso- dilation of aorta, which may be owing to direct action of the steroid on membranes of smooth muscle cells lead- ing to modulation of calcium channels. In osteoblasts, membrane receptors for androgen appear to be coupled to phospholipase C (PLC) via a pertussis toxin–sensi- tive G protein that, after binding testosterone, mediates rapid increments in intracellular calcium and inositol triphosphate (IP 3 ). Testosterone also elicits Ca 2+ mobili- zation in macrophages that lack a “classic” intracellular AR. These cells express an apparent G protein–coupled AR at the cell surface that undergoes agonist-induced internalization and may represent an alternative path- way of steroid hormone action. 5.3. Thyroid Hormone Receptors in Metabolic Regulation Thyroid hormones are well known to regulate energy expenditure and development, and membrane-initiated effects may contribute to these responses. Triiodothyro- nine (T 3 ) rapidly stimulates oxygen consumption and gluconeogenesis in liver. T 3 also promotes an abrupt increase in uptake of the glucose analog 2-deoxyglucose in responsive tissues by augmenting activity of the plasma membrane transport system for glucose. In rat heart, T 3 elicits a positive inotropic effect, increasing left ventricular peak systolic pressure, as early as 15 s after hormone injection. In each tissue investigated, al- terations in intracellular Ca 2+ induced by thyroid hor- mone appear to modulate signal transduction to the cell interior. Membrane-initiated effects of T 3 have been docu- mented in bone cells by means of inositol phosphate signaling, and in brain through calcium channel activa- tion. T 3 can also influence other cell processes, includ- ing the exocytosis of hormones and neurotransmitters, rapid effects that may be attributable to mediation by membrane receptors. Although uptake of T 3 can occur concomitantly with receptor-mediated endocytosis of LDL, and likely is accompanied by carrier proteins, direct uptake of T 3 itself is demonstrable in numerous tissues by means of a high-affinity, stereospecific, and saturable process, such as found for steroid hormones. 5.4. Glucocorticoid Receptors in Metabolic, Immune, and Neural Function In addition to their long-established effects on mo- bilization of energy sources by promoting catabolism and the induction of enzymes involved in gluconeo- genesis, glucocorticoids have profound effects on neu- ron signaling and on induction of apoptosis in lymphocytes, phenomena that appear to be membrane- initiated events. Glucocorticoids rapidly alter neuron- firing patterns. These molecular events lead to glucocorticoid modulation of specific brain functions, such as the rapid response of hypothalamic somatosta- tin neurons to stress. Such abrupt changes in neuron polarization are reinforced by findings of specific, saturable binding of the biologically active radioligand [ 3 H]corticosterone to neuron membranes. Glucocorticoids play an important role in the regu- lation of immune function and in inflammation, espe- cially in severe forms of hematologic, rheumatologic, and neurologic diseases. These steroids have profound anti-inflammatory and immunosuppressive actions when used at therapeutic doses. In lymphoproliferative diseases, glucocorticoids are in wide use for disease management, but the cellular mechanism leading to the therapeutic effect remains unclear. In several stud- ies using both cell lines and freshly prepared leukemia or lymphoma cells, the presence of a membrane-asso- ciated receptor for glucocorticoids has been implicated 82 Part II / Hormone Secretion and Action in modulating cell lysis and death. Moreover, in lym- phocytes, the membrane-binding site is antigenically related to the intracellular glucocorticoid receptor (GR) and may be a natural splice variant of this form. Some glucocorticoids have been shown to inhibit cation transport across the plasma membrane without con- comitant alterations in protein synthesis through tran- scription. It is postulated that the steroid may thus diminish the acute immune response by interfering with immune regulatory events such as the rise in intra- cellular Ca 2+ . An important pharmacologic goal is the development of a steroid compound capable of sepa- rating detrimental side effects of glucocorticoids, such as bone loss, from their beneficial antiinflammatory activity. Future approaches aimed at discrimination of the differential activities of membrane-associated and intranuclear GRs may facilitate this prospect. 5.5. Aldosterone and Digitalis-Like Steroid Receptors in Cardiovascular Health Beyond its classic functions of promoting renal reab- sorption of sodium and excretion of excess potassium, aldosterone enhances sodium absorption from the colon and urinary bladder. In each tissue, the mineralocorti- coid effect is owing to enhanced activity of amiloride- sensitive sodium channels, with aldosterone rapidly augmenting Na + /H + exchange. This function is Ca 2+ and PKC–dependent but independent of nuclear receptor activation. Similarly, nontranscriptional action of aldos- terone has also been reported to underlie its acute effects on cardiac function, such as increased blood pressure and reduced cardiac output, and on sodium transport in vascular smooth muscle cells. Digitalis-like compounds are often overlooked members of the steroid superfamily. These plant- derived agents elicit inotropic and chronotropic effects on the heart but also affect many other tissues. Endo- genous steroidal ligands, termed digitalis-like or oua- bain-like factors, have been found in sera of humans and other animals with blood volume expansion and hypertension and may be released from the adrenal cortex. These ligands elicit inhibition of membrane- associated Na + ,K + -adenosine triphosphatase (ATPase), likely the principal receptor for these agonists. It is notable that the steroid-binding domain of Na + ,K + - ATPase and that of nuclear hormone receptors share significant amino acid sequence homology. In addition to membrane actions of these compounds on Na + ,K + - ATPase, ouabain-induced hypertrophy in myocytes is accompanied by promotion of Ca 2+ flux and initiation of protein kinase–dependent pathways leading, in turn, to specific changes in transcription and altered expres- sion of early and late response genes. Thus, the biologic effects of digitalis-like compounds, long considered the exception to the concept of exclusive genomic influ- ence, may render them more closely integrated with the steroid hormone superfamily than was previously rec- ognized. 5.6. Vitamin D Metabolite Receptors in Bone Health and Disease Membrane-initiated effects of the secosteroid hor- mone 1,25(OH) 2 D 3 are well documented in bone and cartilage. In osteoblasts, interactions have been pro- posed between rapid effects of 1,25(OH) 2 D 3 , requiring milliseconds to minutes, and longer-term effects owing to gene expression. Rapid activation of calcium chan- nels by 1,25(OH) 2 D 3 occurs in these cells. Calcium flux, which can influence gene expression through multiple pathways, promotes key phosphorylation events in certain bone proteins. Osteoblasts exhibit rapid changes in inositol 1,4,5-triphosphate and DAG in response to vitamin D metabolites via activation of PLC. Other bone cells with rapid responses to vitamin D metabolites include osteosarcoma cells and chondrocytes. The latter system is particularly intrigu- ing because chondrocytes elaborate matrix vesicles that appear critical in bone mineralization. Matrix vesicles, which lack nuclei, exhibit specific, saturable binding of 1,25(OH) 2 D 3 , especially when derived from growth- zone chondrocytes. Other rapid effects of vitamin D occur in a variety of cell types. Muscle cells respond within seconds to 1,25(OH) 2 D 3 via several mediators that alter cardiac output in some instances, and acute activation of cal- cium channels in skeletal muscle promotes contraction. Of note, in lymphoproliferative disease, 1,25(OH) 2 D 3 appears to prime monocytic leukemia cells for differen- tiation through acute activation or redistribution of PKC, Ca 2+ , and MAPK. In pancreas and intestine, activation of membrane-associated signaling pathways results in vesicular exocytosis. Pancreatic β-cells respond to 1,25(OH) 2 D 3 with enhanced intracellular Ca 2+ that is coupled to increased insulin release. In intestine, 1,25(OH) 2 D 3 stimulates exocytosis of vesicular calcium and phosphate. These cellular events may be related to vitamin D–promoted alterations in the levels of α-tubu- lin, thereby influencing assembly of microtubules and possibly providing a means for vectorial transport of absorbed ions. Several signal transduction pathways have been found to respond rapidly to exogenous 1,25(OH) 2 D 3 , including activation of protein kinases and promotion of abrupt increments in Ca 2+ , but inte- gration of these signaling cascades with the physiologic response of enhanced ion absorption remains to be estab- lished. Chapter 5 / Plasma Membrane Receptors for Steroids 83 Investigations with vitamin D congeners have recently indicated the potential hormonal nature of 24,25-dihydroxyvitamin D 3 (24,25[OH] 2 D 3 ), once thought to represent merely the inactivation product of precursor 25(OH) 2 D 3 . Acute effects of 24,25(OH) 2 D 3 have been observed in bone cells and in intestine; 24,25(OH) 2 D 3 also inhibits rapid actions of 1,25- (OH) 2 D 3 . This may explain why abrupt effects of 1,25(OH) 2 D 3 often fail to be observed in vivo: normal, vitamin D–replete subjects have endogenous levels of 24,25-(OH) 2 D 3 sufficient to inhibit acute stimulation of calcium transport by 1,25(OH) 2 D 3 , thus providing a feedback regulation system. 5.7. Retinoid Receptors in Development and Malignancy Retinoic acid exerts diverse effects in the control of cell growth during embryonic development and in onco- genesis. Effects of retinoids are widely considered to be mediated exclusively through nuclear receptors, includ- ing those for retinoic acid, as well as retinoid X recep- tors. However, retinoid response pathways independent of nuclear receptors appear to exist. Cellular uptake of retinol (vitamin A) may involve interaction of serum retinol-binding protein with specific surface membrane receptors followed by ligand transfer to cytoplasmic retinol-binding protein. In this regard, targeted disrup- tion of the gene for synthesis of the major endocytotic receptor of renal proximal tubules, megalin, appears to block transepithelial transport of retinol. It is notewor- thy that megalin may also be implicated in receptor- mediated endocytosis of 25-hydroxyvitamin D 3 in complex with its plasma carrier. In addition, retinoic acid binds M-6-P/IGF-2R receptor with moderate affin- ity and enhances its receptor function. M-6-P/IGF-2R is a membrane glycoprotein that functions in binding and trafficking of lysosomal enzymes, in activation of TGF-β, and in degradation of IGF-2, leading to sup- pression of cell proliferation. The concept of multiple ligands binding to and regulating the function of a single receptor is relatively novel but has important implica- tions for modulating and integrating the activities of seemingly independent biologic pathways. 5.8. Steroidal Congeners in Regulation of Angiogenesis Steroidal compounds are also implicated in the regu- lation of angiogenesis. Several steroids, including glu- cocorticoids, have low levels of angiostatic activity. Squalamine, a naturally occurring aminosterol, has highly potent anti-angiogenic activity. The steroidal substance does not bind to any known steroid hormone receptor, but it interacts specifically with caveolar domains at the surface membrane of vascular endothelial cells and disrupts growth factor–induced signaling for the regulation of cell proliferation. In early clinical tri- als, the compound showed considerable promise as a therapeutic agent to block the growth and metastatic spread of lung and ovarian cancers by interfering with tumor-associated angiogenesis. In addition, squalamine steroids may be useful for medical management of macular degeneration, a form of visual loss that is highly correlated with uncontrolled angiogenesis. At this time, there is no known cure for macular degeneration, which afflicts about 30 million people worldwide, and is the leading cause of legal blindness in adults older than 60. 6. CONCLUSION Since the discovery of chromosomal puff induction by the insect steroid hormone ecdysone, cell regulation by steroid hormones has focused primarily on a nuclear mechanism of action. However, even ecdysone is now known to elicit rapid plasma membrane effects that may facilitate later nuclear alterations. Indeed, numerous reports of acute steroid hormone effects in diverse cell types cannot be explained by the generally prevailing theory that centers on the activity of hormone receptors located exclusively in the nucleus. Plasma membrane forms of steroid hormone receptors occur in target cells and are coupled to intracellular signaling pathways that mediate hormone action. Membrane-initiated signals appear to be the primary response of the target cell to steroid hormones and may be a prerequisite for subse- quent genomic activation. Coupling of plasma mem- brane, cytoplasmic and nuclear responses, constitutes a progressive, ordered expansion of initial signaling events. Recent dramatic advances in this area have led to intensified efforts to delineate the nature and bio- logic roles of all classes of receptor molecules that func- tion in steroid hormone–signaling pathways. Molecular details of cross-communication between steroid and peptide receptors are also beginning to emerge, and steroid receptors associated with plasma-membrane signaling platforms may be in a pivotal location to pro- mote convergence among diverse cellular response pathways. This new synthesis has profound implica- tions for integration of the physiology and pathophysi- ology of hormone action in responsive cells and may lead to development of novel approaches for the treat- ment of many cell proliferative, metabolic, inflamma- tory, reproductive, cardiovascular, and neurologic diseases. SELECTED READING Davis PJ, Davis FB. Nongenomic actions of thyroid hormone on the heart. Thyroid 2002;12:459–466. 84 Part II / Hormone Secretion and Action Gruber C, Tschugguel W, Schneeberger C, Huber J. Production and actions of estrogens. N Engl J Med 2002;346:340–352. Hoessli D, Llangumaran S, Soltermann A, Robinson P, Borisch B, Din N. Signaling through sphingolipid microdomians of the plasma membrane: the concept of signaling platform. Glycoconj J 2000;17:191–197. Lösel R, Wehling M. Nongenomic actions of steroid hormones. Nat Rev Mol Biol 2003;4:46–56. Mendelsohn ME, Karas RH. The protective effects of estrogen on the cardiovascular system. N Engl J Med 1999;340:1801– 1811. Migliaccio A, Castoria G, Di Domenico M, de Falco A, Bilancio A, Lombardi M, Bottero D, Varicchio L, Nanayakkara M, Rotondi A, Auricchio F. Sex steroid hormones act as growth factors. J Steroid Biochem Mol Biol 2003;17:1–5. Milner TA, McEwen BS, Hayashi S, Li CJ, Reagan LP, Alves SE. Ultrastructural evidence that hippocampal α-estrogen recep- tors are located at extranuclear sites. J Comp Neurol 2001;429: 355–371. Moss RL, Gu Q, Wong M. Estrogen: nontranscriptional signaling pathway. Recent Prog Hormone Res 1997;52:33–70. Nemere I, Farach-Carson M. Membrane receptors for steroid hor- mones: a case for specific cell surface binding sites for vitamin D metabolites and estrogen. Biochem Biophys Res Commun 1998;248:443–449. Szego CM. Cytostructural correlates of hormone action: new com- mon ground in receptor-mediated signal propagation for steroid and peptide agonists. Endocrine 1994;2:1079–1093. Szego CM, Pietras R. Membrane recognition and effector sites in steroid hormone action. In: Litwack G, ed. Biochemical Actions of Hormones, vol. VIII. New York, NY: Academic 1981:307– 463. Watson CS, Gametchu B. Membrane-initiated steroid actions and the proteins that mediate them. Proc Soc Exp Biol Med 1999; 220:93–19. Weihua Z, Andersson S, Cheng G, Simpson E, Warner M, Gustafsson J-A. Update on estrogen signaling. FEBS Lett 2003; 546:17–24. REFERENCES Chambliss KL, Yuhanna IS, Mineo C, Liu P, German Z, Sherman TS, Mendelsohn ME, Anderson RG, Shaul PW. Estrogen recep- tor alpha and endothelial nitric oxide synthase are organized into a functional signaling module in caveolae. Circ Res 2000;87: E44–E52. Li L, Haynes MP, Bender JR. Plasma membrane localization and function of the estrogen receptor alpha variant (ER46) in human endothelial cells. Proc Natl Acad Sci USA 2003;100:4807–4812. Márquez DC, Pietras RJ. Membrane-associated binding sites for estrogen contribute to growth regulation of human breast cancer cells. Oncogene 2001; 20: 5420–5430. Moats RK II, Ramirez VD. Electron microscopic visualization of membrane-mediated uptake and translocation of estrogen-BSA: colloidal gold by Hep G2 cells. J Endocrinol 2000;166: 631– 647. Navarro CE, Saeed SA, Murdick C, Martinez-Fuentes AJ, Arora K, Krsmanovic LZ, Catt KJ. Regulation of cyclic adenosine 3´,5´- monophosphate signaling and pulsatile neurosecretion by G- coupled plasma membrane estrogen receptors in immortalized gonadotropin-releasing hormone neurons. Mol Endocrinol 2003; 17:1792–1804. Pietras RJ, Nemere I, Szego CM. Steroid hormone receptors in tar- get cell membranes. Endocrine 2001;14:417–427. Pietras RJ, Szego CM. Metabolic and proliferative responses to estro- gen by hepatocytes selected for plasma membrane binding-sites specific for estradiol-17β. J Cell Physiol 1979;98:145–159. Simoncini T, Hafezi-Moghadam A, Brazil DP, Ley K, Chin W, Liao JK. Interaction of oestrogen receptor with the regulatory subunit of phosphatidylinositol-3-OH kinase. Nature 2000;407:538–541. Szego CM, Pietras RJ. Lysosomal functions in cellular activation: propagation of the actions of hormones and other effectors. Int Rev Cytol 1984;88:1–302. Szego CM, Pietras RJ, Nemere I. Encyclopedia of Hormones. In: Henry H, Norman A, eds. Plasma Membrane Receptors for Ste- roid Hormones: Initiation Site of the Cellular Response. San Diego, CA: Academic 2003;657–671. Chapter 6 / Growth Factors 85 85 From: Endocrinology: Basic and Clinical Principles, Second Edition (S. Melmed and P. M. Conn, eds.) © Humana Press Inc., Totowa, NJ 6 Growth Factors Derek LeRoith, MD, PhD and William L. Lowe Jr., MD CONTENTS INTRODUCTION INSULIN-LIKE GROWTH FACTORS OTHER GROWTH FACTORS systems: endocrine and autocrine/paracrine. The IGF family consists of three hormones (insulin, IGF-1 and IGF-2), three receptors (the insulin, IGF-1, and IGF-2 [mannose-6-phosphate, or M-6-P] receptors), and six well-characterized binding proteins (IGFBPs 1–6). The IGFs have structures that resemble insulin, hence their names, and were discovered as circulating hor- mones with insulin-like properties. Following the advent of molecular endocrinology, it was found that IGFs, particularly IGF-1, are produced by all tissues of the body and therefore function as both hormones and growth factors with autocrine/paracrine actions. Insulin interacts with the insulin receptor with high affinity and with the IGF-1 receptor (IGF-1R) with much lower affinity, explaining the metabolic effects mediated by insulin at low circulating levels, whereas insulin’s effect as a mitogen occurs at higher concentra- tions, probably via the IGF-1R. The IGFs, on the other hand, bind to and activate the IGF-1R with high affin- ity, whereas they stimulate metabolic effects through the insulin receptor at high concentrations owing to their low binding affinity for this receptor. The IGF-2R has no apparent signaling capacity and is not discussed fur- ther in this chapter. Both the insulin receptor and the IGF-1R belong to a subgroup of the family of cell-sur- face receptors with endogenous tyrosine kinase activity and are very similar in structure. They are oligomers of αβ−subunits that form an α2β2 heterotetramer. Ligands bind to the extracellular domain of the α−subunit, which 1. INTRODUCTION In this chapter, we discuss various aspects of classic growth factors and their relevance to endocrinology. Although “growth factors” have traditionally been con- sidered to be represented by the family of peptide growth factors, this definition is too restricted given that nonpeptide hormones, e.g., steroid hormones such as estrogen, also stimulate cell growth. Similarly, growth factors have traditionally been considered as tissue fac- tors, functioning locally as autocrine or paracrine fac- tors, as compared to hormones that function in a classic endocrine fashion. We focus here on insulin-like growth factors (IGFs), which represent a paradigm that has both endocrine and autocrine/paracrine modalities. We then discuss other members of classic growth factor families, allowing the reader to compare and contrast them to the IGFs. We also briefly address the numerous cell-surface receptors and the cross talk between receptors. Because we cannot describe here all aspects of the growth fac- tors, their receptors, and interacting proteins, we refer the reader to various other excellent reviews in the Selected Reading section. 2. INSULIN-LIKE GROWTH FACTORS The IGF family of growth factors represents one of the best examples of the overlap of the two classic 86 Part II / Hormone Secretion and Action induces a conformational alteration that results in autophosphorylation on tyrosine residues in the cyto- plasmic domain of the β−subunit. Tyrosine phosphory- lation of the receptor results in binding of cellular substrates that mediate intracellular signaling. It is now evident that the separation of receptors into insulin or IGF-1Rs, does not represent the full spec- trum of receptor expression. Hybrid receptors may rep- resent a significant proportion of the receptors expressed on the cell surface. These hybrids comprise an αβ−subunit from the insulin receptor and an αβ− subunit from the IGF-1R. Hybrid receptors can form in tissues that express both receptors, because their αβ− subunits are similar and are processed by identical path- ways. Generally, these hybrid receptors bind IGF-1 better than insulin, which could explain how IGF-1 induces metabolic effects in certain tissues (such as muscle) even at physiologic concentrations. On the other hand, there are two isoforms of the insulin recep- tor, the A- and B-isoforms, each being differentially expressed by different cells and tissues. Interestingly, the A-isoform, which has an additional 11 amino acids owing to a splicing variation that includes exon 11, has greater mitogenic activity when compared to the meta- bolic activity of the B-isoform. IGF-2 binds the A- isoform with high affinity. Thus, the effect of the different ligands in the IGF family depends, to some extent, on the receptors expressed on the various tis- sues as well as the concentration and composition of the receptors. For example, liver and fat cells express mostly, if not only, insulin receptors and are primary metabolic tissues, whereas most other tissues express a mixture of receptors and may respond to these ligands either with a metabolic response or, more commonly, with mitogenic or differentiated functions. The biologic action of the the IGFs is also dependent on the IGFBPs that bind the IGFs with high affinity but do not bind insulin. In the circulation, the IGFBPs func- tion as classic hormone-binding proteins (e.g., the ste- roid hormone– binding proteins), that bind, neutralize, and protect the IGFs and form a reservoir, making the IGFs available for distribution to the tissues. Although this aspect has been well characterized, it fails to address the growing body of evidence that the IGFBPs represent a complex system of locally produced pro- teins that affect cellular function, thereby representing an autocrine/paracrine system in their own right. Most, if not all, cells express some complement of the six IGFBPs, which they secrete into the local environment. Cell culture experiments have shown that IGFBPs present in the local cellular milieu bind the IGFs with higher affinity than cell-surface IGF-1Rs and are capable of inhibiting their interactions with the cell. On the other hand, posttranslational modifications includ- ing phosphorylation, proteolytic cleavage, or binding of the IGFBPs to the cell surface, as opposed to the extra- cellular matrix, decrease their affinity for the IGFs which releases the bound ligand, thereby allowing delivery to cell-surface receptors. Finally, the IGFBPs can interact with cells and activate cellular events independent of the IGF-1R, via mechanisms presently unknown. Both the ligands (IGF-1 and IGF-2) and the IGFBPs should therefore be viewed as endocrine and autocrine/ paracrine systems that form an interesting paradigm against which to compare other growth factor families. 2.1. IGFs in Health and Disease The IGF system plays a critical role in normal growth and development. There are examples of human disor- ders resulting from genetic mutations in various com- ponents of the system. An IGF-1 gene mutation was described in a severely retarded child who demonstrated growth delay, no response to growth hormone (GH) injections, but a significant response to rhIGF-1. A mutation in the IGF-1R was identified in an infant who was small for gestational age, and a mutation has recently been described in the gene encoding the acid labile-subunit (ALS) of IGFBP-3 resulting in a growth- retarded child. The impact of loss of function of distinct genes in the IGF system has also been examined in mice with null mutations of specific genes. Mice homozygous for deletion of the IGF-1 gene show reduced birth weights with high mortality and severe postnatal growth retardation in the surviving animals. By con- trast, deletion of the IGF-2 gene causes severe growth retardation from embryonic d 13 onward, although postnatal growth continues in parallel with control mice. These findings suggest that IGF-2 plays a role in prenatal growth, whereas IGF-1 plays a role prenatally and a critical role postnatally, especially during the pubertal growth spurt. Mice with deletions of the insu- lin receptor have relatively normal birth weights but die soon after birth secondary to ketoacidosis and severe diabetes. Mice with deletions of the IGF-1R die at birth apparently unable to breathe owing to severe muscle hypoplasia. By contrast, mice heterozygous for deletions of the IGF-1 gene exhibit an increased life- span compared to controls. Similar findings have been observed in Caenorhabditis elegans, in which an insu- lin/IGF-1R deletion is associated with longer survival. IGF-2/M-6-P receptor deletions result in increased birth weight, suggesting that in its absence clearance of IGF-2 protein is reduced, resulting in excess growth via IGF-1R activation. Deletion of the individual genes encoding the IGFBPs has no resultant phenotype, and Chapter 6 / Growth Factors 87 only double or triple crosses lead to some mild pheno- types, suggesting redundancy in the system. Tissue-specific gene deletions have provided further insight into the endocrine and autocrine/paracrine func- tion of the IGF system. Liver-specific deletion of the IGF-1 gene using the cre/loxP system leads to a mouse with a 75% reduction in circulating IGF-1 and a marked increase in circulating GH. The major phenotype in this model is severe insulin resistance owing primarily to the excess GH, but there is also a reduction in spleen weight and bone mineralization, suggesting that circulating IGF-1 is not redundant with tissue IGF-1. This was fur- ther emphasized when a double knockout mouse was created by crossing a mouse with liver-specific deletion of the IGF-1 gene with an ALS knockout mouse. These mice exhibited a more severe reduction in growth and bone mineralization associated with a further reduction in circulating IGF-1. Tissue-specific knockouts of the IGF-1R have been created in bone and the pancreas. In bone, deletion of the IGF-1R results in changes in the growth plate, whereas in pancreatic β-cells, absence of the IGF-1R causes a defect in glucose-stimulated insu- lin secretion associated with reduced expression of the GLUT-2 and glucokinase genes, two proteins critical for glucose uptake and metabolism in β-cells. Essentially all tissues in the body express one or more components of the IGF system. Not surprisingly, every system in the body is controlled, to some degree, by the IGF system during normal growth and development. A few examples of the role of the IGFs in pathophysiology and potential therapeutic applications of IGF-1 are dis- cussed next. 2.2. Cancer The IGF system and its role in cancer cell growth has been the subject of intensive research during the past decade. Components of the system are expressed by vir- tually all cancers and have been shown to affect the growth and function of cancer cells. Most cancers express either IGF-1 or, more commonly, IGF-2, and if they fail to do so, the surrounding stromal tissue releases these ligands. In both circumstances, these ligands stimu- late cell proliferation and are even more active as inhibi- tors of apoptosis, which supports growth of the cancer. IGF-2 is of particular interest because its gene is imprinted, and alterations in imprinting contribute to IGF-2 expression in many tumors. Interestingly, over- expression of a “big IGF-2” by some tumors leads to tumor-induced hypoglycemia, owing to the inability of IGFBP-3 and ALS to totally neutralize this unprocessed form of IGF-2 in the circulation. This leads to high cir- culating levels of unbound big IGF-2 which is then free to interact with tissue receptors (particularly the insulin receptor), resulting in hypoglycemia. Recent epidemiologic studies have demonstrated a correlation between a relative risk of developing pros- tate, breast, colon, lung, and bladder cancer and the level of circulating IGF-1. The greatest correlation was evident in those individuals with IGF-1 levels in the upper quartile of the normal range. The relationship between these two events remains to be determined, but the results have stimulated interest in the connec- tion between the IGF system and cancer growth. Almost all cancers overexpress the IGF-1R, which may explain their more rapid proliferation or protec- tion from apoptosis. One explanation for the overexpression has been found by studies focused on the promoter region of the IGF-1R gene, which is GC rich and normally inhibited by tumor suppressor gene products such as p53, WT1, and BRAC-1. Mutations in these proteins lead to a paradoxical increase in pro- moter activity in colon cancer cells (p53), Wilms tumor (WT1), and breast cancers (BRAC-1). IGFBPs are also expressed by the cancer cells and, in some studies, have been shown to stimulate proliferation (mostly by enhancing IGF-1 function) and, in other cases, to inhibit cell proliferation (in both an IGF-1-dependent and -independent manner). The potential importance of the IGF system, and particularly the IGF-1R, in cancer has led to an intense effort to find blockers of IGF-1R function as potential adjuncts to chemotherapy. These include IGF-1R blocking peptides, antibodies, small molecules, as well as small molecule antagonists to the IGF-1R tyrosine kinase domain. 2.3. Diabetes There has been considerable interest in the possible use of rhIGF-1 in cases of severe insulin resistance. Conceptually, this arose from the knowledge that the IGF-1R is similar to the insulin receptor and can enhance glucose uptake in muscle. As proof of prin- ciple, rhIGF-1 was able to overcome insulin resistance in patients with severe insulin resistance secondary to mutations in the insulin receptor. When administered to patients with type 1 or type 2 diabetes, rhIGF-1 simi- larly reduced the insulin resistance and reduced the requirements for insulin injections. More recently, it has been administered together with IGFBP-3, and, apparently, this mode of administration has fewer side effects. Outstanding questions remain regarding the long-term benefits and potential side effects of IGF-1 on the vasculature and, potentially, cancer cell growth. OTHER GROWTH FACTORS Table 1 lists families of growth factors. In this chap- ter, we only describe briefly some essential elements of [...]... of the author and his group that are included in this chapter were supported by National Institutes of Health grants HD-08129 and HD-20290, and US Department of Agriculture grants 9 8 -3 52 036 635 and 200 4 -3 520 3- 1 4176 REFERENCES Banu S, Arosh JA, Chapeldaine P, Fortier MA Molecular cloning and spatio-temporal expression of the prostaglandin transporter: a basis for the action of prostaglandins in the bovine...88 Part II / Hormone Secretion and Action Table 1 Growth Factor Families IGFs Insulin, IGF-1, IGF-2 VEGFs VEGF, VEGFB, VEGFC, VEGFD, placental growth factor EGF, TGF-α, heparin-binding EGF, amphiregulin, betacellulin PDGF-AA, -BB, -AB TGF-β1–6; inhibin A and B; activin A, B, and C; Müllerian-inhibiting substance; bone morphogenetic proteins NGF, neurotropins (NT -3 , -4 , and -5 ) BDNF 22 family... including brain-derived neurotrophic factor (BDNF), neurotrophin -3 (NT -3 ) , NT-4, and NT-5, also have conserved regions and similar predicted tertiary structures The receptors responsible for mediating their effects are complexes of a low-affinity 75-kDa intrinsic membrane protein (p75) that complexes with either TRK (TRK-A) or TRK-B; TRK-A and TRK-B contain tyrosine kinase activity and when complexed... this receptor include phospholipase C-γ (PLCγ), mitogen-activated protein kinase (MAPK), phosphatidylinositol -3 -kinase (PI3K), and ras guanosine-5´-triphosphatase (GTPase) activating proteins (Fyn and Yes) Recently, a novel VEGF, human endocrine gland– derived VEGF (EG-VEGF) was identified during a screen for endothelial cell mitogens Mature EG-VEGF is an 86-amino-acid peptide that is not structurally... highaffinity functional receptor NGF binds the p75-TRK-A receptor complex, and the signaling pathways that are activated on NGF binding include PLCγ, PI-3K, RAS GTPase-activating protein, SHC, and the MAPK TRKB complex binds BDNF and NT -3 , whereas TRK-C binds NT -3 with high affinity NGFs play important roles in differentiation and survival of neurons, and the specific effects are determined by the expression... then processed to release the mature 5 3- amino-acid molecule in the case of EGF and a 50-amino-acid molecule in the case of TGF-α EGF family members signal through the ErbB family of receptors, which includes ErbB-1 (the EGFR), ErbB-2 (HER2 or Neu), ErbB -3 , and ErbB-4 EGF family members have differing abilities to bind to various homo- Chapter 6 / Growth Factors and heterodimer complexes composed of... hand, TGF-β has antiproliferative effects on T- and B-lymphocytes, and the potential anti-inflammatory and immunosuppressive effects of TGF−β in systemic disorders such as rheumatoid arthritis await further investigation Chapter 6 / Growth Factors 3. 6 Fibroblast Growth Factors There are 22 members of the FGF family, including acidic FGF, basic FGF, and keratinocyte growth factor FGFs bind to low-affinity,... increasing β-cell proliferation and mass It increases DNA synthesis in human fetal pancreatic epithelial cells and enhances βcell development in fetal murine pancreatic explant cultures Finally, betacellulin is expressed in islets and ducts of adult human pancreas and primitive duct cells in the fetal pancreas 3. 3 Platelet-Derived Growth Factors Platelet-derived growth factor-A (PDGF-A) and PDGF-B are encoded... J Lipid Mediators 19 93; 6:275–286 Yokomizo T, et al A second leukotriene B4 receptor, BLT2: a new therapeutic target in inflammation and immunological disorders J Exp Med 2000;192:421– 431 Zurier RB Prostaglandins: then and now and next Semin Arthritis Rheum 20 03; 33: 137 – 139 Chapter 8 / Neuroendocrine–Immune Interface 1 13 8 The Neuroendocrine–Immune Interface Michael S Harbuz, PhD and Stafford L Lightman,... A, and the Xenopus headorganizer, dickkopf EG-VEGF stimulates effects in endothelial cells similar to those of VEGF, including cell proliferation, survival, and chemotaxis This occurs, in part, through increased activity of the MAPKs, extracellular-regulated kinase-1 (ERK-1) and ERK-2, and Akt Interestingly, EG-VEGF is active primarily in endothelial cells of specific origin Indeed, in humans, EG-VEGF . endocrine and autocrine/paracrine. The IGF family consists of three hormones (insulin, IGF-1 and IGF-2), three receptors (the insulin, IGF-1, and IGF-2 [mannose-6-phosphate, or M-6-P] receptors), and. phosphate homeostasis and skeletal mineralization. Am J Physiol Endo- crinol Metab 20 03; 285:E1–E9. Chapter 7 / Prostaglandins and Leukotrienes 93 93 From: Endocrinology: Basic and Clinical Principles, . p75-TRK-A receptor complex, and the signaling pathways that are activated on NGF binding include PLCγ, PI-3K, RAS GTPase-activating protein, SHC, and the MAPK. TRK- B complex binds BDNF and NT -3 ,

Ngày đăng: 10/08/2014, 18:21

Tài liệu cùng người dùng

  • Đang cập nhật ...

Tài liệu liên quan