Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Mol Biol ; 425(7): 1111-8, 2013 Apr 12.
Article in English | MEDLINE | ID: mdl-23318952

ABSTRACT

The tumor suppressor gene p53 induces growth arrest and/or apoptosis in response to DNA damage/hypoxia. Inactivation of p53 confers a selective advantage to tumor cells under a hypoxic microenvironment during tumor progression. The subterranean blind mole rat, Spalax, spends its life underground at low-oxygen tensions, hence developing a wide range of respiratory/molecular adaptations to hypoxic stress, including critical changes in p53 structure and signaling pathway. The highly conserved p53 Arg(R)-172 is substituted by lysine (K) in Spalax, identical with a tumor-associated mutation. Functionality assays revealed that Spalax p53 is unable to activate apoptotic target genes but is still capable of activating cell cycle arrest genes. Furthermore, we have shown that the transcription patterns of representative p53-induced genes (Apaf1 and Mdm2) in Spalax are influenced by hypoxia. Cell cycle arrest allows the cells to repair DNA damage via different DNA repair genes. We tested the transcription pattern of three p53-related DNA repair genes (p53R2, Mlh1, and Msh2) under normoxia and short-acute hypoxia in Spalax, C57BL/6 wild-type mice, and two strains of mutant C57BL/6 mice, each carrying a different mutation at the R172 position. Our results show that while wild-type/mutant mice exhibit strong hypoxia-induced reductions of repair gene transcript levels, no such inhibition is found in Spalax under hypoxia. Moreover, unlike mouse p53R2, Spalax p53R2 transcript levels are strongly elevated under hypoxia. These results suggest that critical repair functions, which are known to be inhibited under hypoxia in mice, remain active in Spalax, as part of its unique hypoxia tolerance mechanisms.


Subject(s)
DNA Repair/genetics , Gene Expression Profiling , Spalax/genetics , Tumor Suppressor Protein p53/genetics , Adaptation, Physiological/genetics , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Substitution , Animals , Apoptosis/genetics , Cell Cycle Checkpoints/genetics , Cell Cycle Proteins/genetics , Hypoxia , Mice , Mice, Inbred C57BL , MutS Homolog 2 Protein/genetics , Mutation , Nuclear Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleotide Reductases/genetics , Transcription, Genetic
2.
J Comp Physiol B ; 182(7): 961-9, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22576753

ABSTRACT

Vertebrate brains are sensitive to oxygen depletion, which may lead to cell death. Hypoxia sensitivity originates from the high intrinsic rate of ATP consumption of brain tissue, accompanied by the release of glutamate, leading to the opening of ionotropic glutamate receptors, such as N-methyl-D-aspartate (NMDA) receptors (NMDARs). The relative expression levels of the four NMDAR-2 (NR2) subunits change during mammalian development with higher levels of units NR2B and NR2D observed during early development and correlated with hypoxic tolerance during embryonic and neonatal stages of development. Higher levels of NR2D are also abundant in brains of hypoxia tolerant species such as the crucian carp. The subterranean mole-rat, Spalax spends its life underground in sealed burrows and has developed a wide range of adaptations to this special niche including hypoxia-tolerance. In this study, we compared the in vivo mRNA expression of NR2 subunits in the brains of embryonic, neonatal and adult Spalax and rat. Our results demonstrate that under normoxic conditions, mRNA levels of NR2D are higher in Spalax than in rat at all developmental stages studied and are similar to levels in neonatal rat and in other hypoxia/anoxia tolerant species. Furthermore, under hypoxia Spalax NR2D mRNA levels increase while no response was observed in rat. Similarly, hypoxia induces an increase in mRNA levels of Spalax NR2A, claimed to promote neuronal survival. We suggest that indeed the proportional combinations of NMDAR-2 subunits contribute to the ability of the Spalax brain to cope with hypoxic environments.


Subject(s)
Adaptation, Biological/physiology , Brain/metabolism , Gene Expression Regulation/genetics , Hypoxia/metabolism , Protein Subunits/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Spalax/metabolism , Animals , DNA Primers/genetics , Protein Subunits/genetics , RNA, Messenger/metabolism , Rats , Spalax/genetics , Species Specificity
3.
Proc Natl Acad Sci U S A ; 107(50): 21570-5, 2010 Dec 14.
Article in English | MEDLINE | ID: mdl-21115824

ABSTRACT

The subterranean mole rat Spalax is an excellent model for studying adaptation of a mammal toward chronic environmental hypoxia. Neuroglobin (Ngb) and cytoglobin (Cygb) are O(2)-binding respiratory proteins and thus candidates for being involved in molecular hypoxia adaptations of Spalax. Ngb is expressed primarily in vertebrate nerves, whereas Cygb is found in extracellular matrix-producing cells and in some neurons. The physiological functions of both proteins are not fully understood but discussed with regard to O(2) supply, the detoxification of reactive oxygen or nitrogen species, and apoptosis protection. Spalax Ngb and Cygb coding sequences are strongly conserved. However, mRNA and protein levels of Ngb in Spalax brain are 3-fold higher than in Rattus norvegicus under normoxia. Importantly, Spalax expresses Ngb in neurons and additionally in glia, whereas in hypoxia-sensitive rodents Ngb expression is limited to neurons. Hypoxia causes an approximately 2-fold down-regulation of Ngb mRNA in brain of rat and mole rat. A parallel regulatory response was found for myoglobin (Mb) in Spalax and rat muscle, suggesting similar functions of Mb and Ngb. Cygb also revealed an augmented normoxic expression in Spalax vs. rat brain, but not in heart or liver, indicating distinct tissue-specific functions. Hypoxia induced Cygb transcription in heart and liver of both mammals, with the most prominent mRNA up-regulation (12-fold) in Spalax heart. Our data suggest that tissue globins contribute to the remarkable tolerance of Spalax toward environmental hypoxia. This is consistent with the proposed cytoprotective effect of Ngb and Cygb under pathological hypoxic/ischemic conditions in mammals.


Subject(s)
Adaptation, Physiological , Globins/metabolism , Hypoxia/metabolism , Myoglobin/metabolism , Nerve Tissue Proteins/metabolism , Spalax/physiology , Animals , Base Sequence , Cytoglobin , Gene Expression Regulation , Globins/genetics , Humans , Myoglobin/genetics , Nerve Tissue Proteins/genetics , Neuroglobin , Rats , Rats, Sprague-Dawley , Sequence Analysis, DNA
4.
J Mol Evol ; 70(1): 1-12, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19967343

ABSTRACT

The muscle ankyrin repeat proteins (MARPs), also known as muscle stretch proteins, are members of a conserved family of genes known to be induced under stress conditions. The three primary members, cardiac ankyrin repeat protein (CARP), Ankyrin Repeat Domain 2 (ARPP), and diabetes-related ankyrin repeat protein (DARP) are expressed in cardiac and skeletal muscle, binding to the giant protein titin. In addition, both CARP and ARPP are proposed to have regulatory functions, shuttling to the nucleus and serving as a liaison between mechanical stress and the transcriptional response. In mouse and human models, CARP is induced during wound healing, denervation, neurogenesis, and angiogenesis; ARPP during an immobilized stretch; DARP is up-regulated in type 2 diabetes, as well as brown adipose tissue, suggesting a role in energy metabolism. Most animal models have focused on stretch response stress; however, little is known about the response of MARPs to hypoxic stress. The blind subterranean mole rat is a model for hypoxia tolerance with the ability to survive extremely hypoxic and hypercapnic underground conditions. Following observations that CARP is differentially expressed in the Spalax muscle in response to hypoxia, we have sequenced the Spalax orthologs of the MARP proteins and profiled expression patterns under varying levels of hypoxic stress among two Spalax species and Rattus. Results show expression patterns highly correlated to the degree of hypoxic tolerance among the three species. Understanding the differences in MARP expression further elucidates mechanisms of hypoxia tolerance with relevance to human ischemic disease.


Subject(s)
Adaptation, Physiological/genetics , Ankyrin Repeat , Blindness , Gene Expression Regulation , Hypoxia/genetics , Muscle Proteins/genetics , Spalax/genetics , Adaptation, Physiological/drug effects , Amino Acid Sequence , Animals , Cloning, Molecular , Gene Expression Regulation/drug effects , Molecular Sequence Data , Muscle Proteins/chemistry , Muscle Proteins/metabolism , Open Reading Frames/genetics , Oxygen/pharmacology , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Sequence Alignment , Stress, Physiological/drug effects , Stress, Physiological/genetics , Time Factors
5.
FASEB J ; 23(7): 2327-35, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19255257

ABSTRACT

The blind subterranean mole rat of the Spalax ehrenbergi superspecies is an excellent animal model for hypoxic tolerance. Unique physiological, functional, and gene structure changes allow Spalax species to survive lower oxygen levels than most terrestrial animals. BNIP3, an HIF-1 dependent hypoxia-response gene, has a proapoptotic function; however, expression is suppressed in many types of cancers. Under hypoxic conditions, BNIP3 also functions as a mediator of mitochondrial autophagy, a survival adaptation to control ROS production and DNA damage. Using real-time PCR and Western blotting, we investigated the impact of hypoxia on BNIP3 expression and mitophagy, in the skeletal muscle and heart, of the Rattus and two Spalax species. BNIP3 transcript, as well as protein levels, increased to significantly higher levels under hypoxia in Rattus tissues, with smaller changes in Spalax. Mitophagy was correlated with BNIP3 expression in the heart with an inverse correlation to hypoxia tolerance. A dense network of vessels in Spalax muscle may offer protection from physiological hypoxia, while the response in Rattus reflects the increase of hypoxic stress. In Spalax tissues, as in many cancers, BNIP3 expression and mitophagy are significantly less affected by hypoxia. Similar mechanisms, beneficial to organisms adapted to stressful environments, may also confer malignant cells with survival features. Understanding the molecular basis of such adaptations may enhance development of new therapeutic modalities.


Subject(s)
Autophagy/genetics , Hypoxia/genetics , Membrane Proteins/genetics , Proto-Oncogene Proteins/genetics , Animals , Hypoxia/metabolism , Mitochondrial Proteins , Mole Rats , Muscle, Skeletal/metabolism , Myocardium/metabolism , Rats , Species Specificity
6.
Acta Histochem ; 111(5): 415-9, 2009.
Article in English | MEDLINE | ID: mdl-18676007

ABSTRACT

Mole rats of the Spalax ehrenbergi superspecies are blind subterranean rodents that live under fluctuating oxygen supply, reduced to a measured 6% O(2), and mostly probably lower, during the rainy season. Fiber typing of muscles of the neck (trapezius) and leg (gastrocnemius, quadriceps) using standard histochemical techniques (succinic dehydrogenase, myosin ATPase) showed that the muscle fibers of mole rats in natural settings, as well as after extended captivity, were predominantly type IIa. The same muscles in laboratory rats showed the full range of fiber types. In contrast, the hearts of the mole rats and the laboratory rats were very similar. Our results indicate that skeletal muscle in the mole rats appears to have evolved in response to specific environmental demands to permit intensive endurance burrowing activities under conditions of severe or chronic hypoxia.


Subject(s)
Hypoxia/physiopathology , Motor Activity/physiology , Muscle, Skeletal/metabolism , Animals , Mole Rats , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/cytology , Myosins/metabolism , Rats , Rats, Sprague-Dawley , Succinate Dehydrogenase/metabolism
7.
FASEB J ; 22(1): 105-12, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17726089

ABSTRACT

Vascular endothelial growth factor receptor (VEGF) plays a critical role in blood vessel formation and affects nerve growth and survival. VEGF receptor 2 (Flk1) functions as the major signal transducer of angiogenesis, mediating VEGF induction of endothelial tubulogenesis. We have cloned and analyzed expression of Flk1 in the blind subterranean mole rat Spalax ehrenbergi. Spalax experience abrupt and sharp changes in oxygen supply in their sealed underground niche and, hence, are genetically adapted to hypoxia and serve as a unique, natural mammalian model organism for hypoxia tolerance. Spalax Flk1 is relatively conserved at the nucleic acid and amino acid level compared to human, mouse, and rat orthologs. Reverse transcription-quantitative polymerase chain reaction was used to analyze Flk1 expression in muscle and brain of animals exposed to ambient or variant hypoxic oxygen levels at multiple stages of development. Transcript levels were compared with those obtained from Rattus, a primary model for human physiology. Our findings demonstrate that under normoxic conditions Flk1 patterns of expression correlate well with our previous investigations of VEGF expression. Exposure to hypoxic conditions resulted in divergent patterns of Flk1 expression between Spalax and Rattus and between muscle and brain. It appears that the regulatory mechanisms differentiating expression between the species and between tissues are most likely unique, suggesting that Flk1 expression may be regulated by multiple processes, including both angiogenesis and neurogenesis.


Subject(s)
Adaptation, Physiological/genetics , Hypoxia/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Base Sequence , Cloning, Molecular , DNA Primers , DNA, Complementary , Mole Rats , Open Reading Frames , RNA, Messenger/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
8.
Article in English | MEDLINE | ID: mdl-16479405

ABSTRACT

The blind subterranean mole rat Spalax ehrenbergi superspecies has evolved adaptive strategies to cope with underground stress. Hypoxia is known to stimulate reactive oxygen species generation; however, mechanisms by which Spalax counteracts oxidative damage have not been investigated before. We studied in Spalax the oxidative status of the Harderian gland (HG), an organ which is particularly vulnerable to oxidative stress in many rodents. With regard to the sexual dimorphism found in this gland, differences between males and females were determined and compared to the surface-dwelling Syrian hamster. Our results show, for the first time, that Spalax exhibits remarkably low biomolecular damage, which implies the existence of physiological strategies to avoid oxidative damage under fluctuating O(2) and CO(2) levels existing in the mole rat's subterranean niche. Correspondingly, main antioxidant enzymes, such as superoxide dismutase (SOD), catalase, and glutathione reductase (GR), exhibited high activities in both genders; in particular, remarkably high levels were measured in SOD. SOD and GR activities showed statistically significant differences between sexes. Melatonin, an important circadian agent is also a very important antioxidant molecule and is synthesized in the Harderian glands (HGs) of Spalax. Therefore, the possible interaction between antioxidant enzymes and melatonin is suggested.


Subject(s)
Acclimatization/physiology , Antioxidants/metabolism , Harderian Gland/enzymology , Oxidative Stress/physiology , Analysis of Variance , Animals , Catalase/metabolism , Cricetinae , Female , Glutathione Reductase/metabolism , Hypoxia/enzymology , Hypoxia/physiopathology , Lipid Peroxidation/physiology , Male , Mesocricetus , Mole Rats , Protein Carbonylation/physiology , Reactive Oxygen Species/metabolism , Sex Factors , Species Specificity , Superoxide Dismutase/metabolism
9.
FASEB J ; 19(10): 1314-6, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16000366

ABSTRACT

The blind subterranean mole rat superspecies Spalax ehrenbergi has evolved adaptations that allow it to survive and carry out intensive activities in its highly hypoxic underground sealed burrows. A key component of this adaptation is a higher capillary density in some Spalax tissues, primarily in muscles used in digging and in other energetic activities, resulting in a shorter diffusion distance for oxygen. Vascular endothelial growth factor (VEGF) is an angiogenic factor that is critical for angiogenesis during development and is found in response to tissue ischemia. We demonstrate here that due to physiological differences, the Spalax muscle regulatory mechanism for VEGF is different than in Rattus muscle. In vivo, the constitutive level of the VEGF mRNA and the mRNA levels of its transcriptional regulator HIF-1alpha and its mRNA stabilizer HuR are significantly higher in Spalax muscle than in Rattus muscle. Furthermore, as opposed to Rattus, the mRNA levels of HIF-1alpha, HuR, VEGF, as well as that of LDH-A, the enzyme that catalyzes the production of lactate, an accepted marker of anaerobic metabolism, are not increased in Spalax after hypoxia. However, ex vivo, when oxygenation by blood vessels is no longer relevant, the expression pattern of all these genes is similar in the two rodents under both normoxic and hypoxic conditions. Our studies provide evidence that the highly vascularized muscle in Spalax, the most energy consuming tissue during digging, is resistant to the effects of oxygen deprivation. The significance of these results with respect to ischemic vascular disease is abundantly clear.


Subject(s)
Hypoxia/physiopathology , Mole Rats/physiology , Muscles/blood supply , Vascular Endothelial Growth Factor A/genetics , Animals , Antigens, Surface/genetics , DNA, Complementary/isolation & purification , ELAV Proteins , ELAV-Like Protein 1 , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Isoenzymes/genetics , L-Lactate Dehydrogenase/genetics , Lactate Dehydrogenase 5 , Muscles/metabolism , Neovascularization, Physiologic , RNA, Messenger/analysis , RNA-Binding Proteins/genetics , Rats
10.
Proc Natl Acad Sci U S A ; 101(33): 12236-41, 2004 Aug 17.
Article in English | MEDLINE | ID: mdl-15302922

ABSTRACT

The tumor suppressor gene p53 controls cellular response to a variety of stress conditions, including DNA damage and hypoxia, leading to growth arrest and/or apoptosis. Inactivation of p53, found in 40-50% of human cancers, confers selective advantage under hypoxic microenvironment during tumor progression. The mole rat, Spalax, spends its entire life cycle underground at decidedly lower oxygen tensions than any other mammal studied. Because a wide range of respiratory adaptations to hypoxic stress evolved in Spalax, we speculated that it might also have developed hypoxia adaptation mechanisms analogous to the genetic/epigenetic alterations acquired during tumor progression. Comparing Spalax with human and mouse p53 revealed an arginine (R) to lysine (K) substitution in Spalax (Arg-174 in human) in the DNA-binding domain, identical to known tumor associated mutations. Multiple p53 sequence alignments with 41 additional species confirmed that Arg-174 is highly conserved. Reporter assays uncovered that Spalax p53 protein is unable to induce apoptosis-regulating target genes, resulting in no expression of apaf1 and partial expression of puma, pten, and noxa. However, cell cycle arrest and p53 stabilization/homeostasis genes were overactivated by Spalax p53. Lys-174 was found critical for apaf1 expression inactivation. A DNA-free p53 structure model predicts that Arg-174 is important for dimerization, whereas Spalax Lys-174 prevents such interactions. Similar neighboring mutations found in human tumors favor growth arrest rather than apoptosis. We hypothesize that, in an analogy with human tumor progression, Spalax underwent remarkable adaptive p53 evolution during 40 million years of underground hypoxic life.


Subject(s)
Evolution, Molecular , Genes, p53 , Mole Rats/genetics , Mutation , Neoplasms/genetics , Adaptation, Physiological/genetics , Amino Acid Sequence , Amino Acid Substitution , Animals , Apoptosis/genetics , Base Sequence , DNA/genetics , Humans , Hypoxia/genetics , Mice , Models, Genetic , Models, Molecular , Mole Rats/physiology , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Conformation , Sequence Homology, Amino Acid , Species Specificity , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics
11.
J Biol Rhythms ; 19(1): 22-34, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14964701

ABSTRACT

The blind subterranean mole rat superspecies Spalax ehrenbergi is an extreme example of mammalian adaptation to life underground. Though this rodent is totally visually blind, harboring a drastically degenerated subcutaneous rudimentary eye, its daily activity rhythm is entrainable to LD cycles. This indicates that it confers light information to the clock, as has been previously shown by the authors in behavioral studies as well as by molecular analyses of its Clock/MOP3 and its three Per genes. The Cryptochrome (Cry) genes found in animals and plants act both as photoreceptors and as essential components of the negative feedback mechanism of the biological clock. To further understand the circadian system of this unique mammal, the authors cloned and characterized the open reading frame of Spalax Cry1 and Cry2. The Spalax CRY1 protein is significantly closer to the human homolog than to the mice one, in contrast to the evolutionary expectations. They have found two isoforms of Cry2 in Spalax, which differ in their 5' end of the open reading frame and defined their expression in Spalax populations. They found a large and significant excess of heterozygotes of sCry2 (sCry2L/S genotype). Both sCry1 and sCry2 mRNAs were found in the SCN, the eye, the harderian gland, as well as in a wide range of peripheral tissues. Their expression pattern under different LD conditions has also been analyzed. As was already shown for other circadian genes, despite being blind and living in darkness, the Cry genes of Spalax behave in a similar, though not identical, pattern as in sighted animals. Once again, the results indicate that the uniquely hypertrophied harderian gland of Spalax plays a key role in its circadian system.


Subject(s)
Biological Clocks/genetics , Circadian Rhythm/genetics , Flavoproteins/genetics , Gene Expression Regulation , Mole Rats/physiology , Protein Isoforms/genetics , Amino Acid Sequence , Animals , Base Sequence , Biological Clocks/physiology , Circadian Rhythm/physiology , Cloning, Molecular , Cryptochromes , Flavoproteins/metabolism , Harderian Gland/physiology , Humans , Light , Mole Rats/genetics , Molecular Sequence Data , Open Reading Frames , Photoperiod , Protein Isoforms/metabolism , Rats , Sequence Alignment , Tissue Distribution
12.
Proc Natl Acad Sci U S A ; 100(8): 4644-8, 2003 Apr 15.
Article in English | MEDLINE | ID: mdl-12672967

ABSTRACT

The optimal vector, regulatory sequences, and method of delivery of angiogenic gene therapy are of considerable interest. The Spalax ehrenbergi superspecies live in subterranean burrows at low oxygen tensions and its tissues are highly vascularized. We tested whether continuous perimuscular administration of Spalax vascular endothelial growth factor (VEGF) DNA could increase tissue perfusion in a murine hindlimb ischemia model. Placebo or VEGF +/- internal ribosome entry site (IRES) was continuously administrated perimuscularly in the ischemic zone by using an infusion pump. None of the mice in the VEGF-treated group (>50 microg) developed visible necrosis vs. 33% of the placebo group. Microscopic necrosis was observed only in the placebo group. Spalax VEGF muscular infiltration resulted in a faster and more complete restoration of blood flow. The restoration of blood flow by VEGF was dose-dependent and more robust and rapid when using the VEGF-IRES elements. The flow restoration using continuous perimuscular infiltration was faster than single i.m. injections. Vessel density was higher in the VEGF and VEGF-IRES (-) groups compared with the placebo. Continuous perimuscular administration of angiogenic gene therapy offers a new approach to restore blood flow to an ischemic limb. Incorporation of an IRES element may assist in the expression of transgenes delivered to ischemic tissues. Further studies are needed to determine whether VEGF from the subterranean mole rat Spalax VEGF is superior to VEGF from other species. If so, 40 million years of Spalax evolution underground, including adaptive hypoxia tolerance, may prove important to human angiogenic gene therapy.


Subject(s)
Endothelial Growth Factors/genetics , Intercellular Signaling Peptides and Proteins/genetics , Ischemia/therapy , Lymphokines/genetics , Mole Rats/genetics , Neovascularization, Physiologic/genetics , Plasmids/administration & dosage , Plasmids/genetics , Animals , Blood Flow Velocity/drug effects , Endothelial Growth Factors/metabolism , Endothelial Growth Factors/pharmacology , Genetic Therapy , Hindlimb/blood supply , Infusion Pumps, Implantable , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Ischemia/physiopathology , Lymphokines/metabolism , Lymphokines/pharmacology , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
13.
Curr Biol ; 12(22): 1919-22, 2002 Nov 19.
Article in English | MEDLINE | ID: mdl-12445384

ABSTRACT

The subterranean mole rat Spalax ehrenbergi superspecies represents an extreme example of adaptive visual and neuronal reorganization. Despite its total visual blindness, its daily activity rhythm is entrainable to light-dark cycles, indicating that it can confer light information to the clock. Although most individuals are active during the light phase under laboratory conditions (diurnal animals), some individuals switch their activity period to the night (nocturnal animals). Similar to other rodents, the Spalax circadian clock is driven by a set of clock genes, including the period (sPer) genes. In this work, we show that diurnal mole rats express the Per genes sPer1 and sPer2 with a peak during the light period. Light can synchronize sPer gene expression to an altered light-dark cycle and thereby reset the clock. In contrast, nocturnal Spalax express sPer2 in the dark period and sPer1 in a biphasic manner, with a light-dependent maximum during the day and a second light-independent maximum during the night. Although sPer1 expression remains light inducible, this is not sufficient to reset the molecular clockwork. Hence, the strict coupling of light, Per expression, and the circadian clock is lost. This indicates that Spalax can dissociate the light-driven resetting pathway from the central clock oscillator.


Subject(s)
Circadian Rhythm/physiology , Mole Rats/physiology , Animals , Blindness/physiopathology , Darkness , Light , Motor Activity/physiology , Periodicity
14.
Proc Natl Acad Sci U S A ; 99(18): 11718-23, 2002 Sep 03.
Article in English | MEDLINE | ID: mdl-12193657

ABSTRACT

We demonstrated that a subterranean, visually blind mammal has a functional set of three Per genes that are important components of the circadian clockwork in mammals. The mole rat superspecies Spalax ehrenbergi is a blind subterranean animal that lives its entire life underground in darkness. It has degenerated eyes, but the retina and highly hypertrophic harderian gland are involved in photoperiodic perception. All three Per genes oscillate with a periodicity of 24 h in the suprachiasmatic nuclei, eye, and harderian gland and are expressed in peripheral organs. This oscillation is maintained under constant conditions. The light inducibility of sPer1 and sPer2, which are similar in structure to those of other mammals, indicates the role of these genes in clock resetting. However, sPer3 is unique in mammals and has two truncated isoforms, and its expressional analysis leaves its function unresolved. Per's expression analysis in the harderian gland suggests an important participation of this organ in the stabilization and resetting mechanism of the central pacemaker in the suprachiasmatic nuclei and in unique adaptation to life underground.


Subject(s)
Circadian Rhythm/genetics , Mole Rats/genetics , Nuclear Proteins/genetics , Animals , Cell Cycle Proteins , Cloning, Molecular , Evolution, Molecular , In Situ Hybridization , Mole Rats/physiology , Molecular Sequence Data , Period Circadian Proteins , Reverse Transcriptase Polymerase Chain Reaction , Species Specificity , Suprachiasmatic Nucleus/metabolism , Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...