Genetic strain differences in platelet aggregation and thrombus formation of laboratory rats

2007 ◽  
Vol 97 (04) ◽  
pp. 665-672 ◽  
Author(s):  
Hideki Ito ◽  
Hideki Hayashi ◽  
Yoshie Nagamura ◽  
Kazuyuki Toga ◽  
Yoshihisa Yamada ◽  
...  

SummaryRats are employed to investigate the role of platelets in thrombus formation under flow conditions in vivo and to evaluate the pre-clinical potential of antiplatelet drugs. While Wistar and Sprague-Dawley (SD) strains are commonly used in thrombosis models, a number of rat strains have been established. Each strain possesses genetically unique characteristics such as hypertension, hyperglycemia or hyperlipidemia. The appropriate selection of a strain might have advantages for physiological and pharmacological studies. Comparative investigation of platelet aggregation among laboratory strains of rats is useful for the development of thrombosis models. In the present study, platelet aggregation response in eight laboratory rat strains, ACI, Brown Norway (BN), Donryu, Fischer 344 (F344), LEW, SD, Wistar and WKAH, were compared. Considerable strain differences were observed in ADP-, collagen- and TRAP-induced platelet aggregation. SD and BN are high-platelet-aggregation strains, while F344 and ACI are low-response strains. In the arteriovenous shunt thrombosis model, SD formed larger thrombi than F344 andWistar rats. In the FeCl 3 -induced thrombosis model with the carotid artery, the time to occlusion of SD was significantly shorter than of F344 and ACI rats. F344 and ACI rats had significantly increased bleeding times compared with SD rat. The present study demonstrates that there are considerable strain differences in platelet aggregation among laboratory rats, which reflect thrombus formation.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 4154-4154
Author(s):  
Kazuya Hosokawa ◽  
Tomoko Ohnishi ◽  
Hiroyuki Matsuda ◽  
Kousuke Kashima ◽  
Takehiko Koide

Abstract Thrombosis is a major cause of morbidity and mortality, and thrombin is a major inducer of thrombus formation. Thus several antithrombotic agents targeting thrombin have been developed. We previously reported an anticoagulant and antiplatelet thrombin derivative, ‘M-anhydrothrombin’ prepared by chemical modifications. In this study, we prepared a new thrombin mutant, specificity of which was highly modulated with substantially improved antithrombotic efficacy. The thrombin mutant designated “AAA-Thrombin” in which Lys65, His43 and Ser205 in B-chain have been replaced by Ala revealed higher affinity and specificity for factor VIII with no enzymatic activity. AAA-Thrombin prolonged APTT much more than anhydrothrombin in a dose dependent manner without affecting PT and TT. Platelet aggregation induced by activation of PAR-1 was also effectively suppressed by AAA-Thrombin. “M-AAA-Thrombin” prepared by further chemical modification of carboxyl groups in AAA-Thrombin enhanced its antithrombotic efficacy. M-AAA-Thrombin (250nM) prolonged APTT approx. two times, and suppressed platelet aggregation by PAR-1 activation, while AAA-Thrombin did not at the same concentration. M-AAA-Thrombin also suppressed ristocetin-induced platelet aggregation. In vivo experiments, M-AAA-Thrombin demonstrated significant antithrombotic property in the arterio-venous shunt thrombosis model and the FeCl3-induced carotid artery thrombosis model in guinea pigs. These results indicate that M-AAA-Thrombin would be a candidate for quite an innovative anticoagulant and antiplatelet agent for both arterial and venous thromboses. Further optimization of mutagenesis and modification, in terms of efficacy and safety is in progress in our laboratory.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3361-3361
Author(s):  
Riitta Lassila ◽  
Annukka Jouppila ◽  
Ulla M Marzec ◽  
Stephen R Hanson

Abstract Abstract 3361 We have developed a semi-synthetic antithrombotic heparin complex, APL001, to mimic mast cell-derived natural heparin proteoglycans (HepPG). HepPG attenuate platelet-collagen interactions under blood flow by inhibiting VWF- and GPIIb/IIIa -mediated platelet aggregation. In addition, rat-derived HepPG arrest platelet thrombus growth on collagen surfaces or at vascular injury sites, both in vitro and in vivo (Lassila et al.ATVB 1997, Kauhanen et al. ATVB 2000, Olsson et al. Thromb Haemost 2002). Our objective was to study the inhibitory capacity of APL001 for preventing human platelet aggregation in vitro and acute thrombosis in a baboon model in vivo. The effects of unfractionated heparin (UFH) and APL001 were compared in relevant coagulation assays (APTT, PT, thrombin time, anti-FXa activity, fibrinogen, FVIII:C and VWF activity (VWF:RCo) and antigen). Additionally, agonist-induced (collagen, ristocetin and ADP) platelet aggregation in citrate or hirudin-anticoagulated whole blood (Multiplate®) (n=10 healthy subjects), and platelet function analysis (PFA100®) in citrated platelet rich plasma (PRP) were assessed. In a well-established baboon thrombosis model a collagen-coated PTFE graft (length 2 cm, lumen 4 mm) was placed in an arterio-venous shunt. Prior to blood contact the thrombogenic surface was treated for 10 min with UFH or APL001 (both at 4 mg/mL). Thrombus formation was initiated by exposing the surface to blood flow (100 mL/min, shear rate 265−1), and the deposition of 111-In-labeled platelets and of fibrin was quantified continuously over 1h. Fibrin thrombus accumulation was assessed from the incorporation of circulating 125-I-fibrinogen. In the heparin-relevant coagulation tests APL001 was comparable or 20–30% more potent than UFH while FVIII, fibrinogen and VWF variables remained unaltered. In contrast to UFH, APL001 (300 μg/mL) consistently inhibited collagen- and ristocetin-induced platelet aggregation, whereas UFH had only a modest effect in comparison with PBS control (Table). ADP-induced aggregation was unaffected. Comparable results were observed in the PRP aggregation assay. PFA100 testing also demonstrated inhibitory effects. In the in vivo thrombosis model (n=4) APL001 reduced platelet deposition on collagen (vs. the results with UFH) by 34% (p=0.01), while platelet accumulation in distal propagated thrombus was reduced by 61% (p=0.16). APL001-treated surfaces accumulated 45% less fibrin than the UFH-treated surfaces (p=0.008). In conclusion, when compared with UFH APL001 inhibited both collagen- and ristocetin-induced platelet aggregation in human blood, while anticoagulant properties were comparable. In the absence of systemic antithrombotic drugs, exposure of APL001 to a highly thrombogenic collagen surface arrested thrombus formation in an in vivo baboon model. This finding suggests that locally administered APL001 alone, due to its dual antiplatelet and anticoagulant effects, may limit the growth and size of thrombus and thereby prevent subsequent thrombo-occlusion.TableAnticoagulantInhibition-% of platelet aggregation ± SDConc. 300 μg/mLnColl (3.2 μg/mL)Ristocetin (0.77 mg/mL)ADP (6.4 μM)CitrateAPL0011033 ± 1543 ± 166 ± 24UFH1011 ± 1323 ± 153 ± 7p value0.0030.0100.700HirudinAPL0011032 ± 1043 ± 178 ± 10UFH108 ± 1116 ± 166 ± 9p value0.0000.0020.600 Disclosures: Lassila: Aplagon: Chief Scientific Advisor.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1060-1060
Author(s):  
Soochong Kim ◽  
Carol Dangelmaier ◽  
Dheeraj Bhavanasi ◽  
Shu Meng ◽  
Hong Wang ◽  
...  

Abstract We investigated the mechanism of activation and functional role of a hitherto uncharacterized signaling molecule, RhoG, in platelets. RhoG is a ubiquitously expressed member of the Rho Family of GTPases. We demonstrated for the first time the expression [Fig 1A] and activation of RhoG [Fig 1B] in platelets. Platelet aggregation and dense-granule secretion in response to glycoprotein VI (GPVI) agonists, collagen-related peptide (CRP) and convulxin were significantly inhibited in RhoG-deficient platelets compared to wild type murine platelets [Fig 1C]. Integrin αIIbβ3 activation and α-granule secretion as measured by flow cytometry were also significantly inhibited in RhoG-deficient murine platelets downstream of GPVI agonists. In contrast, 2-MeSADP- and AYPGKF-induced platelet aggregation and secretion [Fig 1D] were minimally affected in RhoG deficient platelets, indicating that the function of RhoG in platelets is GPVI-specific.Figure 1(A): Increasing amounts of human platelet lysate (in μg) were separated by SDS-PAGE, Western blotted, and probed with anti-RhoG antibody. (B) RhoG activation was measured upon stimulation of washed human platelets with 5μg/ml CRP for various times. Washed platelets were lysed and active GTP-bound RhoG was determined by pull-down analysis using bacterially expressed GST-ELMO. (C) Washed platelets from RhoG -/- mice and RhoG +/+ littermates were stimulated with GPVI agonists, 2.5 μg/ml CRP and 100 ng/ml convulxin and (D) G protein coupled receptor agonists, 30 nM 2MeSADP and 100 μM AYPGKF for 3.5 min under stirring conditions. Platelet aggregation and ATP secretion were measured by aggregometry.Figure 1. (A): Increasing amounts of human platelet lysate (in μg) were separated by SDS-PAGE, Western blotted, and probed with anti-RhoG antibody. (B) RhoG activation was measured upon stimulation of washed human platelets with 5μg/ml CRP for various times. Washed platelets were lysed and active GTP-bound RhoG was determined by pull-down analysis using bacterially expressed GST-ELMO. (C) Washed platelets from RhoG -/- mice and RhoG +/+ littermates were stimulated with GPVI agonists, 2.5 μg/ml CRP and 100 ng/ml convulxin and (D) G protein coupled receptor agonists, 30 nM 2MeSADP and 100 μM AYPGKF for 3.5 min under stirring conditions. Platelet aggregation and ATP secretion were measured by aggregometry. CRP-induced phosphorylations of Syk, Akt and ERK, but not Src family kinases (SFKs), were significantly reduced in RhoG-deficient platelets compared to those of wild type [Fig 2A]. Consistently, CRP-induced RhoG activation was abolished by pan-SFK inhibitor but not by Syk or PI 3-kinase inhibitors [Fig 2B]. Interestingly, unlike CRP, platelet aggregation and Syk phosphorylation induced by fucoidan, a CLEC-2 agonist, were unaffected in RhoG deficient platelets [Fig 2C].Figure 2(A): Washed platelets from RhoG -/- mice and RhoG +/+ littermates were stimulated with 2.5 μg/ml CRP and at 37 °C for 2 min and probed with anti-phospho-Syk (Tyr525/526), anti-phospho-Src (Tyr416), anti-phospho-Akt (Ser473), anti-phospho-ERK, or anti-β-actin (lane loading control) antibodies by western blotting. (B): RhoG activation induced by 5μg/ml CRP for 60 sec was evaluated in the presence and absence of 10 μM PP2, 2 μM OXSI-2, or 100nM wortmannin. (C): Wild type and RhoG-deficient platelets were stimulated with 100 μg/ml fucoidan and probed with anti-phospho-Syk (Tyr525/526), anti-phospho-Akt (Ser473), or anti-β-actin (lane loading control) antibodies by western blotting.Figure 2. (A): Washed platelets from RhoG -/- mice and RhoG +/+ littermates were stimulated with 2.5 μg/ml CRP and at 37 °C for 2 min and probed with anti-phospho-Syk (Tyr525/526), anti-phospho-Src (Tyr416), anti-phospho-Akt (Ser473), anti-phospho-ERK, or anti-β-actin (lane loading control) antibodies by western blotting. (B): RhoG activation induced by 5μg/ml CRP for 60 sec was evaluated in the presence and absence of 10 μM PP2, 2 μM OXSI-2, or 100nM wortmannin. (C): Wild type and RhoG-deficient platelets were stimulated with 100 μg/ml fucoidan and probed with anti-phospho-Syk (Tyr525/526), anti-phospho-Akt (Ser473), or anti-β-actin (lane loading control) antibodies by western blotting. Finally, RhoG -/- mice had a significant delay in time to thrombotic occlusion in cremaster arterioles compared to wild type littermates [Fig 3A and 3B], indicating the important in vivo functional role of RhoG in platelets.Figure 3(A): Time required for occlusion of cremaster arterioles in RhoG +/+ and RhoG -/- mice was measured using microvascular thrombosis model with light/dye-induced injury. 5 mice of each genotype were used, and statistical analysis revealed a significant difference between the 2 genotypes of mice (*, P < .01). (B) Representative images of cremaster arterioles were taken from RhoG +/+ and RhoG -/- mice 30 min after the injury. As seen with the outline (arrows) of the thrombus formed, thrombus formation was inhibited in RhoG -/- mice.Figure 3. (A): Time required for occlusion of cremaster arterioles in RhoG +/+ and RhoG -/- mice was measured using microvascular thrombosis model with light/dye-induced injury. 5 mice of each genotype were used, and statistical analysis revealed a significant difference between the 2 genotypes of mice (*, P < .01). (B) Representative images of cremaster arterioles were taken from RhoG +/+ and RhoG -/- mice 30 min after the injury. As seen with the outline (arrows) of the thrombus formed, thrombus formation was inhibited in RhoG -/- mice. In conclusion, we show for the first time that RhoG is expressed and activated in platelets, plays an important role in GPVI/FcRγ-mediated platelet activation and is critical for thrombus formation in vivo. Disclosures: No relevant conflicts of interest to declare.


2016 ◽  
Vol 2016 ◽  
pp. 1-8 ◽  
Author(s):  
Xue Ding ◽  
Tong-dan Liu ◽  
Zhou-ling Xie ◽  
Qi Zhao ◽  
Yuan Cao ◽  
...  

Integrin αIIbβ3 plays a crucial role in the process of platelet aggregation. Three integrin αIIbβ3 antagonists (abciximab, eptifibatide, and tirofiban) have been approved by FDA for clinical use. Unfortunately, they all showed severe side effects such as thrombocytopenia and bleeding risk. Thus, researches on the development of more effective and safer antiplatelet agents are needed. In this manuscript we reported a novel naphthalenic derivative compound ND-1 with potent antithrombotic effect and lower bleeding risk. ND-1 inhibited ADP-, collagen-, thrombin-, and U46619-induced platelet aggregation with IC50 values of 1.29, 14.46, 12.84, and 40.24 μM, respectively. Mechanism studies indicated that ND-1 inhibited the binding of fibrinogen to integrin αIIbβ3 in a dose-dependent manner with an IC50 value of 3.12 μM. ND-1 inhibited P-selectin expression induced by ADP, collagen, thrombin, and U46619 on the surface of platelets. Additionally, this compound reduced platelets spreading to the immobilized fibrinogen. In vivo, ND-1 potently decreased thrombus formation in an arteriovenous shunt thrombosis model in rats and slightly prolonged bleeding time in a tail cutting model in mice. Taken together, our results reveal that ND-1 is a novel antagonist of αIIbβ3 with strong antithrombotic effect and lower bleeding risk.


2011 ◽  
Vol 43 (15) ◽  
pp. 930-941 ◽  
Author(s):  
Toshihiro Konno ◽  
Lea A. Rempel ◽  
M. A. Karim Rumi ◽  
Amanda R. Graham ◽  
Kazuo Asanoma ◽  
...  

The rat possesses a hemochorial form of placentation. Pronounced intrauterine trophoblast cell invasion and vascular remodeling characterize this type of placentation. Strain-specific patterns of placentation are evident in the rat. Some rat strains exhibit deep intrauterine trophoblast invasion and an expanded junctional zone [Holtzman Sprague-Dawley (HSD), Dahl salt sensitive (DSS)], whereas placentation sites of other rat strains are characterized by shallow invasion and a restricted junctional zone [Brown Norway (BN)]. In this report, we identified a quantitative trait that was used to distinguish strain-specific features of rat placentation. Junctional zone prolactin family 5, subfamily a, member 1 ( Prl5a1) transcript levels were significantly greater in BN rats than in HSD or DSS rats. Prl5a1 transcript levels were used as a quantitative trait to screen placentation sites from chromosome-substituted rat strains (BN chromosomes introgressed into the DSS inbred strain; DSS-BN panel). Litter size, placental weights, and fetal weights were not significantly different among the chromosome-substituted strains. Regulation of the junctional zone Prl5a1 transcript-level quantitative trait was multifactoral. Chromosome-substituted strains possessing BN chromosomes 14 or 17 introgressed into the DSS inbred rat strain displayed Prl5a1 transcript levels that were significantly different from the DSS pattern and more closely resembled the BN pattern. The in situ placental distribution of Prl5a1 mRNA and the structure of the junctional zone of DSS-BN17 rats mimicked that observed for the BN rat. Prl5a1 gene expression was also assessed in BN vs. HSD trophoblast stem cells and following reciprocal BN and HSD embryo transfer. Strain differences intrinsic to trophoblast and maternal environment were identified. In summary, we have identified chromosomes 14 and 17 as possessing regulatory information controlling a quantitative trait associated with rat placentation.


Blood ◽  
1992 ◽  
Vol 80 (5) ◽  
pp. 1247-1253
Author(s):  
Y Imura ◽  
JM Stassen ◽  
S Bunting ◽  
F Stockmans ◽  
D Collen

Platelet aggregation plays an important role in the pathogenesis in arterial thrombotic disorders. The binding of fibrinogen via the Arg- Gly-Asp (RGD) recognition sequence to the platelet glycoprotein IIb/IIIa (GPIIb/IIIa) receptor is an essential step of platelet aggregation induced by various physiologic agonists, and RGD-containing peptides that bind to the GPIIb/IIIa receptor inhibit thrombus formation in vivo. L-cysteine, N-(mercaptoacetyl)D-tyrosyl-L- arginylglycyl-L alpha-aspartyl-cyclic (1----5)-sulfide, 5-oxide (G4120), a cyclic RGD-containing synthetic pentapeptide, inhibits adenosine diphosphate (ADP)-induced platelet aggregation with 50% inhibition (IC50) at a concentration of 0.05 microgram/mL in human plasma, 0.12 microgram/mL in hamster plasma, and 11 micrograms/mL in rat plasma. Corresponding values for the linear tetrapeptide Arg-Gly- Asp-Phe (RGDF) were 7 and 100 micrograms/mL in human and hamster plasma. The antithrombotic effects of G4120 and RGDF were evaluated in a hamster model consisting of a mural platelet-rich femoral vein thrombus induced by standardized endothelial cell damage. Bolus intravenous injection of G4120 was followed by a biphasic disappearance of G4120 from plasma with t1/2 alpha of 3.7 minutes and t1/2 beta of 63 minutes, corresponding to a plasma clearance of 5.2 +/- 0.68 mL/min. Bolus intravenous injection of G4120 inhibited ex vivo platelet aggregation with 0.5 mumol/L ADP and in vivo thrombus formation in a dose-dependent manner, with ID50 of 11 and 11 micrograms/kg, respectively. Bolus injection of RGDF inhibited in vivo thrombus formation; 43% inhibition was obtained at a dose of 30 mg/kg. Thus, this hamster platelet-rich femoral vein thrombosis model may be useful for the investigation of the antithrombotic properties of platelet GPIIb/IIIa antagonistic peptides. The cyclic synthetic peptide G4120 appears to have a very potent antithrombotic activity in vivo.


VASA ◽  
2015 ◽  
Vol 44 (4) ◽  
pp. 285-288 ◽  
Author(s):  
Ye Sun ◽  
Ping Mao ◽  
Jingwei Lu ◽  
Lan Li ◽  
Wanli Lu ◽  
...  

Abstract. Background: Ischemic preconditioning (IPC) has many beneficial effects on the cardiovascular system. However, whether localized lower extremity IPC could be protective against the thrombogenic activity generated by lower extremity ischemia is unclear. Material and methods: 41 male Sprague-Dawley rats were randomly assigned to either a IPC group or a sham group. The lower extremity blood inflow was previously treated with 4 cycles of 5 min ischemia followed by 5 min of reperfusion by clamping the abdominal aortic just before ligature of the left iliac vein(LIV) in the IPC group. Rats in the sham group had a 40-minute blank before left iliac vein ligation. The rats were euthanized at day 2 after ligation and the thrombosed LIV was carefully dissected out, while thrombi harvested from the LIV were measured with weight (g), length (mm) and weight/length (mg/mm). Influence of IPC on coagulation function was also tested. Results: 21 and 20 rats were randomly assigned to einter the IPC group or the control group. Left iliac vein thrombosis was successfully generated in all 41 rats. IPC significantly protects the rats from experimental lower extremity thrombosis. Compared to control group, generated thrombus in rats in the IPC group showed significantly lower weight (2.73 ± 0.16 mg vs 1.82 ± 0.13 mg, P < 0.001), length (2.99 ± 0.17 mm vs 2.44 ± 0.08 mm, P < 0.009) and density (0.95 ± 0.05 mg/mm vs 0.75 ± 0.05 mg/mm, P = 0.01). Influence on coagulation function by IPC itself was not significant (P > 0.05). Conclusions: Our study demonstrated that localized lower extremity IPC could reduce DVT formation in rats in an in vivo experimental thrombosis model.


Blood ◽  
1992 ◽  
Vol 80 (5) ◽  
pp. 1247-1253 ◽  
Author(s):  
Y Imura ◽  
JM Stassen ◽  
S Bunting ◽  
F Stockmans ◽  
D Collen

Abstract Platelet aggregation plays an important role in the pathogenesis in arterial thrombotic disorders. The binding of fibrinogen via the Arg- Gly-Asp (RGD) recognition sequence to the platelet glycoprotein IIb/IIIa (GPIIb/IIIa) receptor is an essential step of platelet aggregation induced by various physiologic agonists, and RGD-containing peptides that bind to the GPIIb/IIIa receptor inhibit thrombus formation in vivo. L-cysteine, N-(mercaptoacetyl)D-tyrosyl-L- arginylglycyl-L alpha-aspartyl-cyclic (1----5)-sulfide, 5-oxide (G4120), a cyclic RGD-containing synthetic pentapeptide, inhibits adenosine diphosphate (ADP)-induced platelet aggregation with 50% inhibition (IC50) at a concentration of 0.05 microgram/mL in human plasma, 0.12 microgram/mL in hamster plasma, and 11 micrograms/mL in rat plasma. Corresponding values for the linear tetrapeptide Arg-Gly- Asp-Phe (RGDF) were 7 and 100 micrograms/mL in human and hamster plasma. The antithrombotic effects of G4120 and RGDF were evaluated in a hamster model consisting of a mural platelet-rich femoral vein thrombus induced by standardized endothelial cell damage. Bolus intravenous injection of G4120 was followed by a biphasic disappearance of G4120 from plasma with t1/2 alpha of 3.7 minutes and t1/2 beta of 63 minutes, corresponding to a plasma clearance of 5.2 +/- 0.68 mL/min. Bolus intravenous injection of G4120 inhibited ex vivo platelet aggregation with 0.5 mumol/L ADP and in vivo thrombus formation in a dose-dependent manner, with ID50 of 11 and 11 micrograms/kg, respectively. Bolus injection of RGDF inhibited in vivo thrombus formation; 43% inhibition was obtained at a dose of 30 mg/kg. Thus, this hamster platelet-rich femoral vein thrombosis model may be useful for the investigation of the antithrombotic properties of platelet GPIIb/IIIa antagonistic peptides. The cyclic synthetic peptide G4120 appears to have a very potent antithrombotic activity in vivo.


1979 ◽  
Author(s):  
J. Lansen ◽  
G. Biagi ◽  
P. Niebes ◽  
J. Gordon ◽  
R. Roncucci

Recent findings have suggested that the in vivo balance between the biosynthesis of proaggregating substances by blood platelets (e.g. thromboxane A2, endoperoxides) and antiaggregating substances produced by the vessel wall (PGI2) might be critical for thrombus formation. We therefore investigated the effect of suloctidil (S), indomethacin (I), acetylsalicylic acid (ASA) and tranylcypromine (T) on these parameters. Male Sprague-Dawley rats (200-300 g) fasted for 12 h were given a single i.v. dose (0.5 and 1 mg/kg) of S (glucuronate salt) or 200 mg/kg of the other compounds. Ten min after the injection, rats were killed and segments of the abdominal aorta and inferior vena cava were excised. PGI2 production by these segments vascular tissue was assessed by platelet aggregation inhibitory activity. PGI2 production was almost completely inhibited by ASA, I and T whereas S enhanced the production (or possibly the effect) of PGI2-like activity. The effect of S was dose dependent and was statistically significant at 1 mg/kg. In vitro studies showed that 100 μM S potentiated the inhibitory effect of synthetic PGI2 on platelet aggregation.


Reproduction ◽  
2004 ◽  
Vol 128 (6) ◽  
pp. 857-862 ◽  
Author(s):  
Nicole R Francisco ◽  
Christen M Raymond ◽  
Paul D Heideman

Laboratory rats have been generally considered non-photoresponsive, but strains of laboratory rats have been found to be variable for this trait. Young males of both the Fischer (F344) and Brown Norway strains (BN) suppress reproductive development, food intake and body mass in short winter photoperiod (short days (SD); 8 h light:16 h darkness), and food restriction interacts with SD to enhance the effect of SD alone. Conversely, young male Harlan Sprague Dawley outbred rats, along with other outbred laboratory rats tested, have little or no response to SD except when unmasked by food restriction or other treatments, and have generally been considered nonphotoperiodic. In order to assess how widespread this trait might be among rat strains, and to test for uncoupling of reproductive and nonreproductive responses, we tested 3 additional inbred strains, including ACI, PVG and BUF rats, for photoresponsiveness and for unmasking of photoperiodic responses by food restriction. Young males of all three inbred strains exhibited photoresponsiveness in testis mass (5–20% lower in SD), seminal vesicle mass (20–50% lower in SD), and body mass (5–10% lower in SD). Food restriction also suppressed reproduction, but there was little or no interaction with the effects of photoperiod. The results are consistent with the hypothesis that laboratory rats are genetically variable for photoperiodism, and that photoresponsiveness may be widespread among inbred rat strains, as all five inbred strains tested have shown photoperiodic responses. The results are particularly important because standard research protocols may unknowingly manipulate this pathway in rats, causing unsuspected variability among or within studies.


Sign in / Sign up

Export Citation Format

Share Document