Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
1.
Science ; 284(5422): 1994-8, 1999 Jun 18.
Article in English | MEDLINE | ID: mdl-10373119

ABSTRACT

In contrast with the prevailing view that most tumors and metastases begin as avascular masses, evidence is presented here that a subset of tumors instead initially grows by coopting existing host vessels. This coopted host vasculature does not immediately undergo angiogenesis to support the tumor but instead regresses, leading to a secondarily avascular tumor and massive tumor cell loss. Ultimately, however, the remaining tumor is rescued by robust angiogenesis at the tumor margin. The expression patterns of the angiogenic antagonist angiopoietin-2 and of pro-angiogenic vascular endothelial growth factor (VEGF) suggest that these proteins may be critical regulators of this balance between vascular regression and growth.


Subject(s)
Endothelial Growth Factors/physiology , Lymphokines/physiology , Membrane Glycoproteins/physiology , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/pathology , Neovascularization, Pathologic , Proteins/physiology , Adenocarcinoma/blood supply , Adenocarcinoma/pathology , Angiopoietin-1 , Angiopoietin-2 , Animals , Apoptosis , Blood Vessels/pathology , Endothelial Growth Factors/genetics , Endothelium, Vascular/pathology , Endothelium, Vascular/physiology , Glioblastoma/blood supply , Glioblastoma/pathology , Glioma/blood supply , Glioma/pathology , In Situ Hybridization , Lymphokines/genetics , Male , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiology , Neoplasm Transplantation , Proteins/genetics , Rats , Rats, Sprague-Dawley , Up-Regulation , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
2.
Science ; 277(5322): 55-60, 1997 Jul 04.
Article in English | MEDLINE | ID: mdl-9204896

ABSTRACT

Angiogenesis is thought to depend on a precise balance of positive and negative regulation. Angiopoietin-1 (Ang1) is an angiogenic factor that signals through the endothelial cell-specific Tie2 receptor tyrosine kinase. Like vascular endothelial growth factor, Ang1 is essential for normal vascular development in the mouse. An Ang1 relative, termed angiopoietin-2 (Ang2), was identified by homology screening and shown to be a naturally occurring antagonist for Ang1 and Tie2. Transgenic overexpression of Ang2 disrupts blood vessel formation in the mouse embryo. In adult mice and humans, Ang2 is expressed only at sites of vascular remodeling. Natural antagonists for vertebrate receptor tyrosine kinases are atypical; thus, the discovery of a negative regulator acting on Tie2 emphasizes the need for exquisite regulation of this angiogenic receptor system.


Subject(s)
Blood Vessels/metabolism , Endothelium, Vascular/cytology , Neovascularization, Physiologic , Proteins/metabolism , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Amino Acid Sequence , Angiopoietin-1 , Angiopoietin-2 , Animals , Blood Vessels/embryology , Cells, Cultured , Cloning, Molecular , Embryo, Mammalian/metabolism , Endothelial Growth Factors/genetics , Endothelial Growth Factors/metabolism , Endothelium, Vascular/metabolism , Female , Humans , Ligands , Lymphokines/genetics , Lymphokines/metabolism , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/metabolism , Mice , Mice, Transgenic , Molecular Sequence Data , Phosphorylation , Proteins/chemistry , Rats , Rats, Sprague-Dawley , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, TIE-2 , Recombinant Fusion Proteins/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
3.
Neuron ; 8(5): 983-93, 1992 May.
Article in English | MEDLINE | ID: mdl-1375039

ABSTRACT

The pattern of retrograde axonal transport of the target-derived neurotrophic molecule, nerve growth factor (NGF), correlates with its trophic actions in adult neurons. We have determined that the NGF-related neurotrophins, brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3), are also retrogradely transported by distinct populations of peripheral and central nervous system neurons in the adult. All three 125I-labeled neurotrophins are retrogradely transported to sites previously shown to contain neurotrophin-responsive neurons as assessed in vitro, such as dorsal root ganglion and basal forebrain neurons. The patterns of transport also indicate the existence of neuronal populations that selectively transport NT-3 and/or BDNF, but not NGF, such as spinal cord motor neurons, neurons in the entorhinal cortex, thalamus, and neurons within the hippocampus itself. Our observations suggest that neurotrophins are transported by overlapping as well as distinct populations of neurons when injected into a given target field. Retrograde transport may thus be predictive of neuronal types selectively responsive to either BDNF or NT-3 in the adult, as first demonstrated for NGF.


Subject(s)
Axonal Transport , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Animals , Brain-Derived Neurotrophic Factor , CHO Cells , Cricetinae , Ganglia, Spinal/metabolism , Ganglia, Sympathetic/metabolism , Hippocampus/metabolism , Iodine Radioisotopes , Male , Mice , Motor Neurons/metabolism , Neurotrophin 3 , Prosencephalon/metabolism , Rats , Rats, Inbred Strains , Spinal Cord/metabolism , Thalamus/metabolism
4.
Neuron ; 5(4): 501-9, 1990 Oct.
Article in English | MEDLINE | ID: mdl-1688327

ABSTRACT

To obtain insight into the site and stage specificity of nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), and neurotrophin-3 (NT-3) action in vivo, we compared the expression patterns of the genes for these three related neurotrophic factors as well as for the NGF receptor in developing and adult rats. Initial embryonic expression of these related neurotrophic factors approximately coincides with the onset of neurogenesis. However, the levels at which the three factors are expressed at this time and throughout the developing nervous system are dramatically different. NT-3 is by far the most highly expressed in immature regions of the CNS in which proliferation, migration, and differentiation of neuronal precursors is ongoing. NT-3 expression dramatically decreases with maturation of these regions. By contrast, BDNF expression is low in developing regions of the CNS and increases as these regions mature. NGF expression varies during the development of discrete CNS regions, but not in any consistent manner compared with NT-3 and BDNF. Despite the dramatic variations, NT-3, BDNF, and NGF do share one striking similarity--high level expression in the adult hippocampus. Our observations are consistent with the idea that NT-3, BDNF, and NGF have paralleled as well as reciprocal roles in vivo.


Subject(s)
Nerve Growth Factors/metabolism , Nerve Tissue Proteins/metabolism , Nervous System/metabolism , Aging/metabolism , Animals , Animals, Newborn , Brain-Derived Neurotrophic Factor , Central Nervous System/metabolism , Gene Expression Regulation , Growth Substances/metabolism , Nerve Growth Factors/genetics , Nerve Tissue Proteins/genetics , Nervous System/growth & development , RNA, Messenger/metabolism , Rats , Rats, Inbred Strains , Tissue Distribution
5.
Neuron ; 9(2): 295-305, 1992 Aug.
Article in English | MEDLINE | ID: mdl-1497895

ABSTRACT

Adult rat sciatic nerve is known to express high levels of ciliary neurotrophic factor (CNTF) mRNA and protein. Here we examine the cellular localization of CNTF protein and mRNA in peripheral nerve and the regulation of CNTF expression by peripheral axons. In intact nerve, CNTF immunoreactivity is found predominantly in the cytoplasm of myelin-related Schwann cells. After axotomy, CNTF immunoreactivity and mRNA levels fall dramatically and do not recover unless axons regenerate. This behavior is similar to the pattern of myelin gene expression in these nerves. We conclude that the expression of CNTF in Schwann cells depends on axon-Schwann cell interactions.


Subject(s)
Gene Expression Regulation , Myelin Sheath/physiology , Nerve Tissue Proteins/genetics , Schwann Cells/metabolism , Sciatic Nerve/metabolism , Animals , Axons/physiology , Blotting, Northern , Blotting, Western , Ciliary Neurotrophic Factor , Cytoplasm/metabolism , Denervation , Immunohistochemistry , Nerve Crush , Nerve Regeneration , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/metabolism , Nucleic Acid Hybridization , RNA, Messenger/metabolism , Rats , Rats, Inbred Strains
6.
Neuron ; 10(1): 89-102, 1993 Jan.
Article in English | MEDLINE | ID: mdl-8381290

ABSTRACT

We recently proposed that ciliary neurotrophic factor (CNTF) shares two receptor components with a generally acting cytokine, leukemia inhibitory factor (LIF), but that CNTF also requires a third receptor component (CNTFR alpha) that is mostly restricted to the nervous system in its expression. Here we demonstrate that a transfected CNTFR alpha gene is sufficient to confer CNTF responsiveness upon hemopoietic cells normally responsive only to LIF, providing evidence that CNTFR alpha is a required receptor component that uniquely characterizes CNTF-responding cells. Consistent with this notion, CNTFR alpha expression could be localized to neurons within all known peripheral targets of CNTF. CNTFR alpha was also widely expressed within neurons of the CNS, suggesting that CNTF has broader CNS actions than previously appreciated. However, in vivo localization of CNTFR alpha, as well as of CNTF itself, is consistent with a particularly important role for CNTF in motor function as well as during neuropoiesis.


Subject(s)
Nervous System/growth & development , Receptors, Cell Surface/physiology , Signal Transduction/physiology , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Brain/embryology , Brain/metabolism , Brain Chemistry , Cloning, Molecular , Ganglia/chemistry , Gene Expression , In Situ Hybridization , Molecular Sequence Data , Muscles/embryology , Muscles/innervation , Muscles/metabolism , Nervous System/embryology , Nervous System Physiological Phenomena , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Receptor, Ciliary Neurotrophic Factor , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Spinal Cord/chemistry , Spinal Cord/embryology , Spinal Cord/metabolism , Structure-Activity Relationship , Transfection
7.
Trends Neurosci ; 17(5): 182-90, 1994 May.
Article in English | MEDLINE | ID: mdl-7520198

ABSTRACT

Recent advances in the understanding of the physiological role of nerve growth factor (NGF) have raised the question of whether neurotrophic factors might have clinical potential in the treatment of neurodegenerative disease or nerve trauma. Although NGF was first characterized as a target-derived survival factor for developing sympathetic and sensory neurons, it is now clear that it plays an important role in the maintenance and regeneration of mature peripheral neurons. However, the highly restricted specificity of NGF for sympathetic neurons, subpopulations of neural-crest-derived sensory neurons, and striatal and basal forebrain cholinergic neurons has, for almost two decades, stimulated the search for other neurotrophic factors that might act on the many classes of neurons that do not respond to NGF. In this article, the biology of the recently discovered NGF-related family of neurotrophic factors and ciliary neurotrophic factor and their receptors are reviewed, especially in the context of the therapeutic potential of these factors in the treatment of neurological disorders of the CNS.


Subject(s)
Nerve Tissue Proteins , Animals , Central Nervous System/metabolism , Central Nervous System Diseases/metabolism , Ciliary Neurotrophic Factor , Humans , Nerve Degeneration , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/therapeutic use , Nervous System Diseases/drug therapy , Nervous System Diseases/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Ciliary Neurotrophic Factor , Receptors, Growth Factor/metabolism
8.
Oncogene ; 18(38): 5356-62, 1999 Sep 20.
Article in English | MEDLINE | ID: mdl-10498889

ABSTRACT

Our analyses in several different tumor settings challenge the prevailing view that malignancies and metastases generally initiate as avascular masses that only belatedly induce vascular support. Instead, we find that malignant cells rapidly co-opt existing host vessels to form an initially well-vascularized tumor mass. Paradoxically, the co-opted vasculature does not undergo angiogenesis to support the growing tumor, but instead regresses (perhaps as part of a normal host defense mechanism) via a process that involves disruption of endothelial cell/smooth muscle cell interactions and endothelial cell apoptosis. This vessel regression in turn results in necrosis within the central part of the tumor. However, robust angiogenesis is initiated at the tumor margin, rescuing the surviving tumor and supporting further growth. The expression patterns of Angiopoietin-2 (the natural antagonist for the angiogenic Tie2 receptor) and vascular endothelial growth factor (VEGF) strongly implicate these factors in the above processes. Angiopoietin-2 is highly induced in co-opted vessels, prior to VEGF induction in the adjacent tumor cells, providing perhaps the earliest marker of tumor vasculature and apparently marking the co-opted vessels for regression. Subsequently, VEGF upregulation coincident with Angiopoietin-2 expression at the tumor periphery is associated with robust angiogenesis. Thus, in tumors, Angiopoietin-2 and VEGF seem to reprise the roles they play during vascular remodeling in normal tissues, acting to regulate the previously underappreciated balance between vascular regression and growth.


Subject(s)
Models, Biological , Neoplasms/blood supply , Neovascularization, Pathologic/physiopathology , Angiopoietin-1 , Angiopoietin-2 , Animals , Blood Vessels/pathology , Blood Vessels/physiopathology , Endothelial Growth Factors/physiology , Humans , Lymphokines/physiology , Membrane Glycoproteins/physiology , Neoplasms/pathology , Proteins/physiology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
9.
J Neurosci ; 21(17): 6706-17, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11517260

ABSTRACT

The findings that brain-derived neurotrophic factor (BDNF) promotes in vitro the survival and/or differentiation of postnatal subventricular zone (SVZ) progenitor cells and increases in vivo the number of the newly generated neurons in the adult rostral migratory stream and olfactory bulb prompted us to investigate whether the infusion of BDNF influences the proliferation and/or differentiation of cells in other regions of the adult forebrain. We examined the distribution and phenotype of newly generated cells in the adult rat forebrain 16 d after intraventricular administration of BDNF in conjunction with the cell proliferation marker bromodeoxyuridine (BrdU) for 12 d. BDNF infusion resulted in numerous BrdU(+) cells, not only in the SVZ lining the infused lateral ventricle, but moreover, in specific parenchymal structures lining the lateral and third ventricles, including the striatum and septum, as well as the thalamus and hypothalamus, in which neurogenesis had never been demonstrated previously during adulthood. In each region, newly generated cells expressed the neuronal marker microtubule-associated protein-2, or neuron-specific tubulin, identified by the antibody TuJ1. The percentage of the newly generated cells expressing TuJ1 ranged from 27 to 42%, suggesting that the adult forebrain has a more profound capacity to produce neurons than recognized previously. The extent of cell proliferation after BDNF infusion was correlated with the level of expression of full-length TrkB, the high-affinity receptor for BDNF, despite the fact that the BrdU(+) cells were not themselves TrkB(+). Collectively, our results demonstrate that the adult brain parenchyma may recruit and/or generate new neurons, which could replace those lost as a result of injury or disease.


Subject(s)
Brain-Derived Neurotrophic Factor/administration & dosage , Lateral Ventricles/drug effects , Neurons/drug effects , Prosencephalon/drug effects , Animals , Antigens, Differentiation/biosynthesis , Bromodeoxyuridine , Cell Count , Cell Division/drug effects , Corpus Striatum/cytology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Hypothalamus/cytology , Hypothalamus/drug effects , Hypothalamus/metabolism , Injections, Intraventricular , Lateral Ventricles/cytology , Lateral Ventricles/metabolism , Microtubule-Associated Proteins/biosynthesis , Neurons/cytology , Neurons/metabolism , Phenotype , Prosencephalon/cytology , Prosencephalon/metabolism , Rats , Rats, Sprague-Dawley , Receptor, trkB/biosynthesis , Septum of Brain/cytology , Septum of Brain/drug effects , Septum of Brain/metabolism , Thalamus/cytology , Thalamus/drug effects , Thalamus/metabolism , Tissue Distribution
10.
J Neurosci ; 20(11): 4081-90, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10818143

ABSTRACT

Ciliary neurotrophic factor (CNTF) is pleiotrophic for central, peripheral, and sensory neurons. In the mature retina, CNTF treatment enhances survival of retinal ganglion and photoreceptor cells exposed to otherwise lethal perturbation. To understand its mechanism of action in vivo, the adult rat retina was used as a model to investigate CNTF-mediated activation of Janus kinase/signal transducer and activator of transcription (Jak-STAT) and ras-mitogen activated protein kinase (ras-MAPK). Intravitreal injection of Axokine, an analog of CNTF, phosphorylates STAT3 and MAPK and produces delayed upregulation of total STAT3 and STAT1 protein in rat retina. Activated STAT3 is predominantly localized in nuclei of retinal Müller (glial) cells, ganglion cells, and astrocytes, but not in photoreceptors. Although CNTF alpha-receptor (CNTFRalpha) mRNA and protein are localized predominantly if not exclusively in retinal neurons, coincident CNTF-mediated STAT3 signaling was observed in both glia and neurons. CNTF-induced activation of Jak-STAT signaling prompted us to investigate STAT3 phosphorylation after a variety of stress-mediated, conditioning stimuli. We show that STAT3 is activated in the retina after exposure to subtoxic bright light, mechanical trauma, and systemic administration of the alpha(2)-adrenergic agonist xylazine, all of which have been shown previously to condition photoreceptors to resist light-induced degeneration. These results demonstrate that CNTF directly stimulates Jak-STAT and ras-MAPK cascades in vivo and strongly suggest that STAT3 signaling is an underlying component of neural responsiveness to stress stimuli. The observation that CNTF activates STAT3 in ganglion cells, but not in photoreceptors, suggests that Jak-STAT signaling influences neuronal survival via both direct and indirect modes of action.


Subject(s)
Ciliary Neurotrophic Factor/pharmacology , Extracellular Signal-Regulated MAP Kinases , Neuroglia/metabolism , Neurons/metabolism , Retina/metabolism , Signal Transduction/drug effects , Stress, Psychological/metabolism , Transcription, Genetic/physiology , Animals , Conditioning, Psychological/physiology , DNA-Binding Proteins/biosynthesis , Glial Fibrillary Acidic Protein/biosynthesis , Humans , Immunohistochemistry , MAP Kinase Kinase Kinases/biosynthesis , Male , Nerve Tissue Proteins/pharmacology , Neuroglia/drug effects , Neuroglia/enzymology , Neurons/drug effects , Neurons/enzymology , Phosphorylation , Photic Stimulation , Rats , Rats, Sprague-Dawley , Retina/cytology , Retina/drug effects , STAT3 Transcription Factor , Trans-Activators/biosynthesis
11.
Diabetes ; 48(3): 588-94, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10078561

ABSTRACT

Systemic administration of brain-derived neurotrophic factor (BDNF) decreases nonfasted blood glucose in obese, non-insulin-dependent diabetic C57BLKS-Lepr(db)/lepr(db) (db/db) mice, with a concomitant decrease in body weight. By measuring percent HbA1c in BDNF-treated and pair-fed animals, we show that the effects of BDNF on nonfasted blood glucose levels are not caused by decreased food intake but reflect a significant improvement in blood glucose control. Furthermore, once established, this effect can persist for weeks after cessation of BDNF treatment. Oral glucose tolerance tests were performed to examine the effects of BDNF on blood glucose control in the fasted state and after an oral glucose challenge. BDNF treatment normalized fasting blood glucose from initially hyperglycemic levels and also showed evidence for beneficial, although less marked, effects on the ability to remove exogenous glucose from blood. One means to lower fasting blood glucose is to reduce the glucose output of peripheral tissues that normally play a part in the maintenance of fasting hyperglycemia. Because the liver is the major endogenous source of glucose in blood during fasting, and because hepatic weight and glucose output are increased in type 2 diabetes, we evaluated the effects of BDNF on liver tissue. BDNF reduced the hepatomegaly present in db/db mice, in association with reduced liver glycogen and reduced liver enzyme activity in serum, supporting the possible involvement of liver tissue in the mechanism of action for BDNF.


Subject(s)
Blood Glucose/metabolism , Brain-Derived Neurotrophic Factor/pharmacology , Brain-Derived Neurotrophic Factor/therapeutic use , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus/metabolism , Hyperglycemia/prevention & control , Obesity , Animals , Blood Glucose/drug effects , Diabetes Mellitus/blood , Diabetes Mellitus/drug therapy , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/drug therapy , Fasting , Glucose Tolerance Test , Glycated Hemoglobin/analysis , Heterozygote , Liver/drug effects , Liver/metabolism , Liver Glycogen/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Time Factors
12.
Endocrinology ; 102(5): 1645-8, 1978 May.
Article in English | MEDLINE | ID: mdl-570487

ABSTRACT

Very small electrolytic lesions were made over the anterior or posterior portion of the optic chiasm in mature female rats showing normal estrous cycles. Lesions over the posterior portion of chiasm destroyed the suprachiasmatic nucleus of the hypothalamus (SCN) while the anterior lesions destroyed a small neural structure, here designated as the medial preoptic nucleus (MPN). Both lesions were effective in inducing persistent vaginal estrus, but when animals were ovariectomized and treated with exogenous and progesterone it was found that lesions including the MPN alone, but not the SCN alone, eliminated the positive feedback effects of this steroid regimen on LH release.


Subject(s)
Estrus , Hypothalamus/physiology , Luteinizing Hormone/metabolism , Progesterone/pharmacology , Animals , Castration , Female , Hypothalamus/injuries , Pregnancy , Preoptic Area/injuries , Preoptic Area/physiology , Rats
13.
Endocrinology ; 127(5): 2408-17, 1990 Nov.
Article in English | MEDLINE | ID: mdl-2171915

ABSTRACT

There exists considerable evidence to suggest that CRH and arginine vasopressin (AVP)-secreting parvocellular neurosecretory neurons of the hypothalamic paraventricular nucleus (PVN) are central integrators of negative feedback effects evoked by circulating glucocorticoid hormones. Most evidence suggests that these neurons may be receptive to circulating glucocorticoid levels, either via glucocorticoid receptors indigenous to these cells and/or via extrahypothalamic glucocorticoid-receptive neurons interacting with the PVN secretory cell. In an effort to address this issue, we performed anterior (ANT), posterior (POST) and total (TOT) deafferentations of the PVN region in male Sprague-Dawley rats using microknives fashioned from narrow-gauge spinal needles. Effective knife cuts were verified immunohistochemically, and deemed acceptable only if they avoided damage to the PVN proper and fibers of CRH and AVP-containing neurons coursing through the hypothalamus en route to the median eminence, while effectively eliminating neuronal input into the PVN region. Subsequent to surgery, levels of mRNA encoding for CRH and AVP in the parvocellular and magnocellular PVN were assayed via semiquantitative in situ hybridization histochemistry. Results indicate that TOT deafferentations resulted in significant increases in CRH mRNA expression in the PVN, and a slight but noticeable induction of AVP mRNA in the medial parvocellular but not posterior magnocellular divisions of the PVN. ANT lesions also produced an up-regulation of CRH and AVP mRNA relative to operated control rats. POST lesions did not produce a clear induction in either CRH or AVP mRNA. The data indicate that in the absence of neuronal input coming from anterior structures, CRH mRNA expression is up-regulated, suggesting that local effects of glucocorticoids on the PVN neuron are ineffective in maintaining normal CRH mRNA expression. These results support a role for neuronal feedback in regulation of the CRH neuron. The limited up-regulation (compared with adrenalectomized rats) of AVP mRNA in the TOT group suggests that while neuronal input may have some control of AVP mRNA expression, local glucocorticoid feedback is clearly able to restrict AVP message to levels considerably less than those seen in steroid-deficient animals. Analysis of knife-cut effects on plasma corticosterone and ACTH levels reveals that POST and TOT, but not ANT, deafferentations prohibit the secretory activity of the hypothalamo-pituitary-adrenocortical (HPA) axis seen pursuant to the anesthesia/thoracotomy in lesion and operated control groups.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Arginine Vasopressin/genetics , Corticotropin-Releasing Hormone/genetics , Hypothalamus/physiology , Paraventricular Hypothalamic Nucleus/metabolism , RNA, Messenger/metabolism , Adrenocorticotropic Hormone/blood , Afferent Pathways/physiology , Animals , Corticosterone/blood , Denervation , Immunohistochemistry , Male , Nucleic Acid Hybridization , Rats , Rats, Inbred Strains
14.
Endocrinology ; 142(7): 3244-54, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11416048

ABSTRACT

The aims of this study were to 1) quantify changes in angiogenesis during follicular growth in a primate model; 2) investigate the molecular regulation using in situ hybridization of vascular endothelial growth factor (VEGF), its receptor, Flt-1, the angiopoietins (Ang-1 and Ang-2), and their receptor, Tie-2; 3) elucidate the role of VEGF in follicular angiogenesis by blocking its action by treatment with a soluble truncated form of the Flt-1 receptor, (VEGF Trap(A40)). Changes in angiogenesis were quantified using bromodeoxyuridine to obtain a proliferation index, and CD31 immunocytochemistry to visualize endothelial cell area. Percentage of proliferating endothelial cells was calculated by double labeling for bromodeoxyuridine and CD31. Vascularization was first observed in follicles containing four granulosa cell layers. A significant increase in proliferation in the thecal layer was observed from the early to late secondary stage, and dual staining showed that 25% of proliferating cells were of endothelial cell origin. VEGF messenger RNA (mRNA) was expressed in granulosa cells with an increase of grain density from late secondary to tertiary follicles. Ang-1 was weakly expressed in the theca of tertiary follicles. Ang-2 mRNA was not detected in any follicles. The mRNA for the Flt-1 and Tie-2 receptors was localized in endothelial cells of the theca. Unexpectedly, Tie-2 mRNA was also found in granulosa cells of early follicular stages and its translation was confirmed by immunocytochemistry. VEGF trap treatment for 3 days resulted in an 87% decrease of proliferation in the theca of secondary and tertiary follicles, a reduction in endothelial cell area and a marked decline in Flt-1 mRNA expression. Granulosa cell proliferation also decreased. These results show that onset and establishment of the follicle vasculature takes place early during follicular development. The ability of VEGF trap treatment to severely restrict follicular angiogenesis establishes that VEGF is the major regulator of this process in the primate ovary.


Subject(s)
Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Ovarian Follicle/blood supply , Ovarian Follicle/physiology , Proto-Oncogene Proteins/pharmacology , Receptor Protein-Tyrosine Kinases/pharmacology , Angiotensin II/genetics , Animals , Bromodeoxyuridine/metabolism , Callithrix , Endothelial Growth Factors/genetics , Female , Immunohistochemistry , In Situ Hybridization , Lymphokines/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , RNA, Messenger/metabolism , Receptor, TIE-2 , Reference Values , Tissue Distribution , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1 , Vascular Endothelial Growth Factors
15.
J Clin Endocrinol Metab ; 86(7): 3377-86, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11443214

ABSTRACT

The possibility of stimulating or inhibiting paracrine factors regulating angiogenesis may lead to new approaches for the treatment of pathological conditions of the female reproductive tract. We examined the effects of a clinical candidate, a soluble truncated form of the Flt-1 receptor, vascular endothelial growth factor trap(A40) (VEGF trap), in a primate model to determine its ability to prevent the onset of luteal angiogenesis or intervene with the on-going process. Marmosets were treated from the day of ovulation until luteal day 3 (prevention regimen) or on luteal day 3 for 1 day (intervention regimen). Effects of VEGF inhibition were studied by obtaining a proliferation index using bromodeoxyuridine incorporation, quantifying endothelial cell area using CD31, and assessing luteal function by plasma progesterone. After both treatments, intense luteal endothelial proliferation was suppressed, a concomitant decrease in endothelial cell area confirmed the inhibition of vascular development, and a marked fall in plasma progesterone levels showed that luteal function was compromised. In situ hybridization was used to localize and quantify compensatory effects on the expression of angiogenic genes. VEGF messenger ribonucleic acid (mRNA) expression in luteal cells was increased, whereas expression of its receptor, Flt, was decreased. Inhibition of VEGF resulted in localized increased expression of angiopoietin-2 mRNA and its receptor, Tie-2. The results show that the VEGF trap can prevent luteal angiogenesis and inhibit the established process with resultant suppression of luteal function. Luteal Flt mRNA expression is dependent upon VEGF, and VEGF inhibition results in abortive increases in expression of VEGF, angiopoietin-2, and Tie-2.


Subject(s)
Corpus Luteum/blood supply , Neovascularization, Physiologic/drug effects , Proto-Oncogene Proteins/pharmacology , Receptor Protein-Tyrosine Kinases/pharmacology , Angiopoietin-2 , Animals , Bromodeoxyuridine/analysis , Bromodeoxyuridine/metabolism , Callithrix , Cell Division , Corpus Luteum/physiology , Endothelial Growth Factors/antagonists & inhibitors , Endothelial Growth Factors/genetics , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Female , Immunoglobulin Fc Fragments/genetics , In Situ Hybridization , Luteal Phase/drug effects , Lymphokines/antagonists & inhibitors , Lymphokines/genetics , Organ Size , Ovary/anatomy & histology , Ovulation , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Progesterone/blood , Proteins/genetics , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , RNA, Messenger/analysis , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor , Recombinant Fusion Proteins/pharmacology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1 , Vascular Endothelial Growth Factors
16.
Mol Neurobiol ; 23(2-3): 121-35, 2001.
Article in English | MEDLINE | ID: mdl-11817215

ABSTRACT

During the past decade, there has been a surge of interest in growth factors (GFs) that act selectively on vascular endothelium and perivascular cells. Studies employing mutant mice or the administration of recombinant proteins have suggested that these factors not only mediate the proliferation of endothelial cells, but also regulate vascular differentiation, regression, and permeability. During and after cerebral ischemia, brain vasculature becomes leaky and unstable, and the normally impermeable blood-brain barrier breaks down. Several days after the ischemic insult, endothelial cells begin to proliferate, and angiogenesis occurs. Expression studies have shown that key vascular GFs are regulated, during these processes, in a complex and coordinated manner. The distinct pattern of regulation exhibited by each vascular GF suggests a unique role for each factor during the initial vascular destabilization and subsequent angiogenesis that occurs after cerebral ischemia. Data from studies in other biological systems support these suggested roles. Thus, manipulation of vascular GFs may prove to be an effective means of stabilizing or enriching brain vasculature after ischemia, and ameliorating the detrimental effects of blood-brain barrier breakdown and vessel regression after stroke.


Subject(s)
Brain Ischemia/metabolism , Endothelial Growth Factors/metabolism , Lymphokines/metabolism , Animals , Brain Ischemia/drug therapy , Brain Ischemia/pathology , Endothelial Growth Factors/genetics , Endothelial Growth Factors/therapeutic use , Humans , Lymphokines/genetics , Lymphokines/therapeutic use , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
17.
J Comp Neurol ; 267(4): 562-79, 1988 Jan 22.
Article in English | MEDLINE | ID: mdl-3346377

ABSTRACT

The hypothalamic suprachiasmatic nucleus (SCh) is the principal brain structure involved in the generation of circadian rhythms. In the present study, we have employed immunohistochemical techniques to evaluate the development of the fetal SCh following its transplantation to the brain of adult host animals. Donor hypothalami were obtained from normal Long-Evans fetuses and transplanted to the lateral, third, or fourth ventricle of Brattleboro rats. Neuronal aggregations exhibiting the organotypic features of the SCh were present in over 90% of the grafts recovered at each transplantation site. Like the normal endogenous SCh, SCh-like cell groups identified within the transplants contained a prominent population of parvicellular (9-13 micron), neurophysin-containing neurons that were immunopositive for vasopressin (VP) but not oxytocin. These SCh-like cell groups also invariably contained similar small neurons that were immunoreactive for vasoactive intestinal polypeptide (VIP). Typically, VP and VIP immunoreactive perikarya were concentrated in contiguous, complementary parts of the grafted SCh, but fibers immunoreactive for either peptide were distributed throughout the extent of the nucleus. Because the brain of the Brattleboro rat is deficient in vasopressin, it was possible to evaluate the projection of the vasopressinergic component of the transplanted SCh to the host brain. Although SCh were identified in grafts recovered from each intraventricular transplantation site, an appreciable input to the host brain could be identified only when the fetal tissue was grafted to the third ventricle. Here, grafted SCh established efferent connections with periventricular diencephalic structures which ordinarily receive a projection from the in situ SCh. Specifically, VP immunoreactive fibers originating from transplanted SCh were identified in the medial preoptic area, the periventricular and dorsomedial hypothalamic nuclei, the paraventricular nuclei of the thalamus and hypothalamus, and in the retrochiasmatic area, arcuate nucleus, and suprachiasmatic nucleus of the host brain. These results demonstrate that the fetal SCh not only survives transplantation but also retains its distinguishing cytological features and the capacity to form an appropriately restricted set of efferent connections with the brain of adult host animals.


Subject(s)
Suprachiasmatic Nucleus/transplantation , Animals , Arginine Vasopressin/metabolism , Efferent Pathways/physiology , Hypothalamus/cytology , Immunohistochemistry , Male , Neurophysins/metabolism , Oxytocin/immunology , Rats , Rats, Brattleboro , Rats, Inbred Strains , Suprachiasmatic Nucleus/cytology , Suprachiasmatic Nucleus/metabolism , Vasoactive Intestinal Peptide/immunology
18.
J Comp Neurol ; 192(1): 1-19, 1980 Jul 01.
Article in English | MEDLINE | ID: mdl-7410605

ABSTRACT

The cell bodies of origin of axons terminating in the median eminence have been identified by retrograde axonal transport of horseradish peroxidase (HRP) or 125I-wheat germ agglutinin (WGA). The tracers were injected into the median eminence by pressure and under direct visual control, using a ventral surgical approach. The retrogradely labeled cells are exclusively located within the hypothalamus. The most heavily labeled cells are parvocellular neurons in the arcuate nucleus, the periventricular area, the medial part of the paraventricular nucleus, and the rostral paraventricular nucleus; a few cells are also located in the rostral part of the preoptic area immediately lateral and dorsal to the organum vasculosum of the lamina terminalis (OVLT). Less heavily located cells are found in the magnocellular neurosecretory nuclei, including the lateral parts of the paraventricular and rostral paraventricular nuclei, the supraoptic nucleus, and the accessory magnocellular nuclei. Retrogradely labeled cells are not found in the ventromedial hypothalamic nucleus, except for a few lightly labeled cells in the posterior division of the nucleus. However, if the injected tracer spreads into the arcuate nucleus, labeled cells are present throughout the ventromedial nucleus. Labeled cells are not found in other parts of the hypothalamus, including lateral and posterior portions of the preoptic area, the anterior hypothalamic area, and the suprachiasmatic nucleus, or in catecholaminergic cell groups of the midbrain, pons, and medulla. Control injections of HRP into the posterior pituitary and the ventromedial nucleus produce patterns of cell labeling which are very distinct from that seen with injections into the median eminence. Following injections into the posterior pituitary, the cells of the magnocellular neurosecretory nuclei are all heavily labeled, but small cells in the parvocellular neuronal groups are not labeled. Direct injections into the ventromedial nucleus resulted in labeled cells in widespread parts of the hypothalamus, as well as in the bed nucleus of the stria terminalis and the lateral septum, in parts of the amygdaloid complex and the subiculum, and in several cell groups in the midbrain, pons, and medulla.


Subject(s)
Hypothalamo-Hypophyseal System/anatomy & histology , Median Eminence/anatomy & histology , Afferent Pathways/anatomy & histology , Animals , Axons/ultrastructure , Female , Horseradish Peroxidase , Hypothalamus, Middle/anatomy & histology , Male , Neurons, Afferent/ultrastructure , Paraventricular Hypothalamic Nucleus/anatomy & histology , Pituitary Gland, Posterior/innervation , Preoptic Area/anatomy & histology , Rats , Supraoptic Nucleus/anatomy & histology
19.
J Comp Neurol ; 206(1): 28-48, 1982 Mar 20.
Article in English | MEDLINE | ID: mdl-6284805

ABSTRACT

The cortical projection of the nucleus submedius (Sm) was studied in the cat with the autoradiographic and horseradish peroxidase (HRP) methods. The results indicate that Sm projects topographically on to layer 3 of a distinct agranular cortical field that occupies the posterolateral gyrus proreus, the adjacent fundus of the rhinal sulcus, and the postero-ventral portion of the medial wall of the presylvian sulcus. This cortical field is denoted the ventrolateral orbital cortex (VLO), consonant with previous nomenclature in the rat (Krettek and Price, '77a). The more ventral part (VLO beta) is cytoarchitectonically distinct from the dorsal part (VLO alpha); the former receives input from the anterior part of Sm (Sma), while the latter receives input from the dorsal and ventral parts of Sm (Smd and Smv). A light input to superficial layer 1 of VLO probably also arises from Sm, and there may be an input to layers 5 and 6. The corticothalamic projection from VLO to Sm reciprocates the ipsilateral thalamocortical projection and also has a moderate contralateral component. A dense, subpial layer 1 input to VLO arises from cells of the ventromedial nucleus (VM) subjacent to Sm. The present experiments also indicate that clusters of cells in VM probably provide input to layer 3 of the cortex in the fundus of the presylvian sulcus, as well as area 6a beta in the lateral wall of the presylvian sulcus and the ventral bank of the cruciate sulcus. Results from the HRP experiments additionally indicate that VLO beta and the anteroventral (Smv) portion of VLO alpha are reciprocally connected with the ventral agranular insular cortex and the cingulate cortex, ipsilaterally, while the posterodorsal (Smd) portion of VLO alpha is instead connected wih specific portions of the somatosensory cortical areas bilaterally. All portions of VLO alpha appear to project to the ventrolateral periaqueductal gray. In light of the recent suggestion that Smd is involved with nociception (Craig and Burton, '81), the present results suggest that the related portion of VLO alpha may serve as a cortical representation for noxious stimuli.


Subject(s)
Cerebral Cortex/anatomy & histology , Thalamic Nuclei/anatomy & histology , Afferent Pathways/anatomy & histology , Animals , Autoradiography , Cats , Dominance, Cerebral/physiology , Efferent Pathways/anatomy & histology , Frontal Lobe/anatomy & histology , Horseradish Peroxidase , Medulla Oblongata/anatomy & histology , Neurons/ultrastructure , Nociceptors/ultrastructure , Spinal Cord/anatomy & histology , Synaptic Transmission
20.
J Comp Neurol ; 357(2): 296-317, 1995 Jun 26.
Article in English | MEDLINE | ID: mdl-7665731

ABSTRACT

To evaluate effective means for delivering exogenous neurotrophins to neuron populations in the brain, we compared the distribution and transport of brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), and neurotrophin-3 (NT-3) following intracerebral delivery. Rats received an injection of radioiodinated or unlabeled neurotrophin into the lateral ventricle and were killed humanely after 1.5-24 hours. Other rats received continuous infusion of unlabeled neurotrophin into the lateral ventricle, the striatum, or the hippocampus for 3-14 days. The neurotrophins were detected by autoradiography or immunohistochemistry. There were striking differences between BDNF, NGF, and NT-3 in their penetration through brain tissue. These differences occurred regardless of the site or method of delivery, but were most pronounced following a bolus intracerebroventricular (ICV) injection. After ICV injection, NGF was widely distributed in tissues around the ventricles and at the surface of the brain, whereas the penetration of BDNF into brain tissue was distinctly less than that of NGF, and the penetration of NT-3 was intermediate. An ICV injection of NGF produced prominent but transient labeling of cells that contain the low-affinity NGF receptor, whereas an injection of BDNF prominently labeled the ventricular ependyma. During ICV infusion (12 micrograms/day), the distribution of BDNF was no greater than that observed after a 12-micrograms bolus injection. A sixfold increase in the amount of BDNF infused (72 micrograms/day) produced a more widespread distribution in the brain and doubled the depth of penetration into periventricular tissues near the cannula. Corresponding to their differences in penetration, NGF was retrogradely transported by basal forebrain cholinergic neurons after ICV or intrastriatal delivery, whereas NT-3 was transported by a few basal forebrain neurons after ICV delivery, and BDNF was rarely detected in neurons after ICV delivery. Delivery of BDNF directly to the striatum or the hippocampus labeled numerous neurons in nuclei afferent to these structures. In situ hybridization studies confirmed that the high-affinity BDNF receptor (TrkB) was much more widely expressed in neurons than was the high-affinity NGF receptor (TrkA). Moreover, mRNA for truncated forms of TrkB was expressed at high levels in the ependyma, the choroid epithelium, and the gray matter. It is likely that binding of BDNF to TrkB, which appears to be more abundant and ubiquitous than TrkA, restricts the diffusion of BDNF relative to that of NGF.


Subject(s)
Brain Chemistry/physiology , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Nerve Growth Factor/metabolism , Animals , Autoradiography , Basal Ganglia/metabolism , Brain/cytology , Brain-Derived Neurotrophic Factor , Cerebral Ventricles/anatomy & histology , Cerebral Ventricles/cytology , Cerebral Ventricles/metabolism , Diffusion , Immunohistochemistry , In Situ Hybridization , Injections, Intraventricular , Iodine Radioisotopes , Neuroglia/metabolism , Neurons/metabolism , Neurotrophin 3 , Rats , Rats, Sprague-Dawley , Receptor Protein-Tyrosine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL