428
Views
7
CrossRef citations to date
0
Altmetric
Reviews

Significance of molecular chaperones and micro RNAs in acquisition of thermo-tolerance in dairy cattle

References

  • Bharati J, Dangi SS, Mishra SR, et al. Expression analysis of Toll like receptors and interleukins in Tharparkar cattle during acclimation to heat stress exposure. J Therm Biol. 2017;65:48–56.
  • Mishra SR, Kundu AK, Mahapatra APK. Effect of ambient temperature on membrane integrity of spermatozoa in different breeds of bulls. The Bioscan. 2013;8:181–183.
  • Nagayach R, Prakash A. Absolute quantification of heat shock protein 70 gene in Jamunapari goat breed. IISUniv J Sc Tech. 2015;4:1–5.
  • Dangi SS, Gupta M, Maurya D, et al. Expression profile of HSP genes during different seasons in goats (Capra hircus). Trop Anim Health Prod. 2012;44(8):1905–1912.
  • Mohanarao GJ, Mukherjee A, Banerjee D, et al. Hsp70 family gene and Hsp27 expression in response to heat and cold stress in vitro in peripheral blood mononuclear cells of goat (Capra hircus). Small Ruminant Res. 2014;116(2-3):94–99.
  • Mehla K, Magotra A, Choudhary J, et al. Genome-wide analysis of the heat stress response in zebu (Sahiwal) cattle. Gene. 2014;533(2):500–507.
  • Abdelnour SA, Abd El-Hack ME, Khafaga AF, et al. Stress biomarkers and proteomics alteration to thermal stress in ruminants: a review. J Therm Biol. 2019;79:120–134.
  • Sahu S, Mishra SR, Kundu AK. Impact of thermal stress on expression dynamics of HSP60 in cardiac fibroblast cells of goat. Anim Biotechnol. 2019;28:1–7.
  • Joo M, Chi JG, Lee H. Expressions of HSP70 and HSP27 in hepatocellular carcinoma. J Korean Med Sci. 2005;20(5):829–834.
  • Sun PM, Liu YT, Wang QH, et al. Localizations of HSP70 and HSP70 mRNA in the tissues of heat stressed broilers. Chin J Agric Biotechnol. 2007;15:404–408.
  • Zulkifli I, Norbaiyah B, Cheah YW, et al. A note on heat shock protein 70 expression in goats subjected to road transportation hot, humid tropical conditions. Animal. 2010;4(6):973–976.
  • Sharma S, Ramesh K, Hyder I, et al. Effect of melatonin administration on thyroid hormones, cortisol and expression profile of heat shock proteins in goats (Capra hircus) exposed to heat stress. Small Ruminant Res. 2013;112(1-3):216–223.
  • Qu H, Donkin SS, Ajuwon KM. Heat stress enhances adipogenic differentiation of subcutaneous fat depot-derived porcine stromovascular cells. J Anim Sci. 2015;93(8):3832–3842.
  • Sodhi M, Mukesh M, Kishore A, et al. Novel polymorphisms in UTR and coding region of inducible heat shock protein 70.1 gene in tropically adapted Indian zebu cattle (Bos indicus) and riverine buffalo (Bubalus bubalis). Gene. 2013;527(2):606–615.
  • Dangi SS, Gupta M, Nagar V, et al. Impact of short-term heat stress on physiological responses and expression profile of HSPs in Barbari goats. Int J Biometeorol. 2014;58(10):2085–2093.
  • Belhadj Slimen I, Najar T, Ghram A, Abdrrabba M. Heat stress effects on livestock: molecular, cellular and metabolic aspects, a review. J Anim Physiol Anim Nutr. 2016;100(3):401–412.
  • Somal A, Aggarwal A, Upadhyay RC. Effect of thermal stress on expression profile of apoptosis related genes in peripheral blood mononuclear cells of transition Sahiwal cow. Iran J Vet Res. 2015;16(2):137–143.
  • Sonna LA, Gaffin SL, Pratt RE, et al. Effect of acute heat shock on gene expression by human peripheral blood mononuclear cells. J Appl Physiol. 2002;92(5):2208–2220.
  • Deb R, Sajjanar B, Pavani KC. Bovine heat shock protein 70 and its application in cellular thermo tolerance. J Vet Sci Technol. 2015;6:1000–1121.
  • Kumar A, Ashraf S, Goud TS, et al. Expression profiling of major heat shock protein genes during different seasons in cattle (Bos indicus) and buffalo (Bubalus bubalis) under tropical climatic condition. J Therm Biol. 2015;51:55–64.
  • Kishore A, Sodhi M, Sharma A, et al. Transcriptional stability of heat shock protein genes and cell proliferation rate provides an evidence of superior cellular tolerance of Sahiwal (Bos indicus) cow PBMCs to summer stress. J Vet Sci. 2016;2:34–40.
  • Rodenhiser D, Jung JHJ, Atkinson BG. Mammalian lymphocytes: stress-induced synthesis of heat-shock proteins in vitro and in vivo. Can J Biochem Cell Biol. 1985;63(7):711–722.
  • Hunter-Lavin C, Davies EL, Bacelar MM, et al. Hsp70 release from peripheral blood mononuclear cells. Biochem Biophys Res Comm. 2004;324(2):511–517.
  • Yu J, Bao E, Yan J, Lei L. Expression and localization of Hsps in the heart and blood vessel of heat-stressed broilers. Cell Stress Chaperones. 2008;13(3):327–335.
  • Das S, Palai TK, Mishra SR, et al. Nutrition in relation to diseases and heat stress in poultry. Vet World. 2011;4:429–432.
  • Romero RD, Montero Pardo A, Montaldo HH, et al. Differences in body temperature, cell viability, and HSP-70 concentrations between Pelibuey and Suffolk sheep under heat stress. Trop Anim Health Prod. 2013;45(8):1691–1696.
  • Kishore A, Sodhi M, Kumari P, et al. Peripheral blood mononuclear cells: a potential cellular system to understand differential heat shock response across native cattle (Bos indicus), exotic cattle (Bos taurus), and riverine buffaloes (Bubalus bubalis) of India. Cell Stress Chaperones. 2014;19(5):613–621.
  • Bharati J, Dangi SS, Chouhan VS, et al. Expression dynamics of HSP70 during chronic heat stress in Tharparkar cattle. Int J Biometeorol. 2017;61(6):1017–1027.
  • Gutierrez JA, Guerriero V. Jr. Quantitation of Hsp70 in tissues using a competitive enzyme-linked immunosorbent assay. J Immunol Methods. 1991;143(1):81–88.
  • Malayer JR, Hansen PJ. Differences between Brahman and Holstein cows in heat-shock induced alterations of protein synthesis and secretion by oviducts and uterine endometrium. J Anim Sci. 1990;68(1):266–280.
  • Zheng Y, Chen K, Zheng X, et al. Identification and bioinformatics analysis of microRNAs associated with stress and immune response in serum of heat-stressed and normal Holstein cows. Cell Stress Chaperones. 2014;19(6):973–981.
  • Sengar GS, Deb R, Singh U, et al. Differential expression of microRNAs associated with thermal stress in Frieswal (Bos taurus x Bos indicus) crossbred dairy cattle. Cell Stress Chaperones. 2018;23(1):155–170.
  • Shandilya UK, Sharma A, Sodhi M, Mukesh M. Heat stress modulates differential response in skin fibroblast cells of native cattle (Bos indicus) and riverine buffaloes (Bubalus bubalis). Biosci Rep. 2020;40:BSR20191544.
  • Hansen PJ. Physiological and cellular adaptations of zebu cattle to thermal stress. Anim Reprod Sci. 2004;82–83:349–360.
  • Gill JK, Arora JS, Kumar BS, et al. Cellular thermotolerance is independent of HSF 1 expression in zebu and crossbred non-lactating cattle. Int J Biometeorol. 2017;61(9):1687–1693.
  • Archana PR, Aleena J, Pragna P, et al. Role of heat shock proteins in livestock adaptation to heat stress. J Dairy Vet Anim Res. 2017;5:00127.
  • Sejian V, Bhatta R, Gaughan JB, et al. Review: adaptation of animals to heat stress. Animal. 2018;12(s2):S431–S444.
  • Berman A. Estimates of heat stress relief needs for Holstein dairy cows. J Anim Sci. 2005;83(6):1377–1384.
  • Collier R, Collier J, Rhoads R, Baumgard LH. Invited review: genes involved in the bovine heat stress response. J Dairy Sci. 2008;91(2):445–454.
  • Li QL, Ju ZH, Huang JM, et al. Two novel SNPs in HSF1 gene are associated with thermal tolerance traits in Chinese Holstein cattle. DNA Cell Biol. 2011;30(4):247–254.
  • Kolli V, Upadhyay RC, Singh D. Peripheral blood leukocytes transcriptomic signature highlights the altered metabolic pathways by heat stress in zebu cattle. Res Vet Sci. 2014;96(1):102–110.
  • Khan R, Sahu AR, Malla WA, et al. HSPs, ubiquitins and antioxidants aid in heat tolerance in Tharparkar indicine cattle. bioRxiv. 2020.DOI: https://doi.org/10.1101/2020.04.09.031153.
  • Bakthisaran R, Tangirala R, Rao CM. Small heat shock proteins: Role in cellular functions and pathology. Biochim Biophys Acta. 2015;1854(4):291–319.
  • Hu H, Zhang Y, Zheng N, et al. The effect of heat stress on gene expression and synthesis of heat-shock and milk proteins in bovine mammary epithelial cells. Anim Sci J. 2016;87(1):84–91.
  • Min L, Cheng J, Shi B, et al. Effects of heat stress on serum insulin, adipokines, AMP-activated protein kinase, heat shock signal molecules in dairy cows. J Zhejiang Univ Sci B. 2015;16(6):541–548.
  • Baek YC, Kim M, Jeong JY, et al. Effects of short-term acute heat stress on physiological responses and heat shock proteins of Hanwoo steer (Korean cattle). J Anim Reprod Biotechnol. 2019;34(3):173–182.
  • Kim WS, Nejad JG, Roh SG, Lee HG. Heat-shock proteins gene expression in peripheral blood mononuclear cells as an indicator of heat stress in beef calves. Animals (Basel. 2020;10(5):895.
  • Pires BV, Stafuzza NB, Lima S, et al. Differential expression of heat shock protein genes associated with heat stress in Nelore and Caracu beef cattle. Livest Sci. 2019;230:103839.
  • Mishra SR, Palai TK. Importance of HSP70 in Livestock – at cellular level. J Mol Pathophysiol. 2014;3(2):30–32.
  • Beere HM, Wolf BB, Cain K, et al. Heat-shock protein 70 inhibits apoptosis by preventing recruitment of procaspase-9 to the Apaf-1 apoptosome. Nat Cell Biol. 2000;2(8):469–475.
  • Palai TP, Mishra SR. Caspases: an apoptosis mediator. J Adv Vet Anim Res. 2015;2(1):18–22.
  • Guerriero V, Jr, Raynes DA. Synthesis of heat stress proteins in lymphocytes from livestock. J Anim Sci. 1990;68(9):2779–2783.
  • Kamwanja LA, Chase CC, Gutierrez JA, et al. Responses of bovine lymphocytes to heat shock as modified by breed and antioxidant status. J Anim Sci. 1994;72(2):438–444.
  • Flanagan SW, Ryan AJ, Gisolfi CV, Moseley PL. Tissue-specific HSP70 response in animals undergoing heat stress. Am J Physiol. 1995;268(1 Pt 2):R28–R32.
  • Edwards JL, Hansen PJ. Elevated temperature increases heat shock protein 70 synthesis in bovine two-cell embryos and compromises function of maturing oocytes. Biol Reprod. 1996;55(2):341–346.
  • Edwards JL, Hansen PJ. Differential responses of bovine oocytes and preimplantation embryos to heat shock. Mol Reprod Dev. 1997;46(2):138–145.
  • Wang S, Diller KR, Aggarwal SJ. Kinetics study of endogenous heat shock protein 70 expression. J Biomech Eng. 2003;125(6):794–797.
  • Schwerin M, Sanftleben H, Grupe S. Genetic predisposition for productive life is associated with functional inactivation of a AP2-binding site in the promoter of the stress protein 70.1-encoding gene in cattle. Arch Tierzucht. 2003;46:177–185.
  • Kristensen TN, Lovendahl P, Berg P, Loeschcke V. Hsp72 is present in plasma from Holstein-Friesian dairy cattle, and the concentration level is repeatable across days and age classes. Cell Stress Chaperones. 2004;9(2):143–149.
  • Kristensen TN, Lovendahl P. Physiological responses to heat stress and their potential use as indicators of reduced animal welfare in jersey calves. Acta Zool Sinica. 2006;52:681–689.
  • Lacetera N, Bernabucci U, Scalia D, et al. Heat stress elicits different responses in peripheral blood mononuclear cells from Brown Swiss and Holstein cows. J Dairy Sci. 2006;89(12):4606–4612.
  • Collier RJ, Dahl GE, VanBaale MJ. Major advances associated with environmental effects on dairy cattle. J Dairy Sci. 2006;89(4):1244–1253.
  • Wang S, Xie W, Rylander MN, et al. HSP70 kinetics study by continuous observation of HSP-GFP fusion protein expression on a perfusion heating stage. Biotechnol Bioeng. 2008;99(1):146–154.
  • Agnew LL, Colditz IG. Development of a method of measuring cellular stress in cattle and sheep. Vet Immunol Immunopathol. 2008;123(3–4):197–204.
  • Liu YX, Li DQ, Cui QW, et al. Analysis of HSP70mRNA level and association between linked microsatellite loci and heat tolerance traits in dairy cows. Yi Chuan. 2010;3:935–941.
  • Roy KS, Collier RJ. Regulation of acclimation to environmental stress. In: Collier RJ, eds. Environmental Physiology of Livestock. 1st ed. Chichester, UK: John Wiley & Sons; 2012:49–54.
  • Gaughan J, Bonner S, Loxton I, Mader T. Effects of chronic heat stress on plasma concentration of secreted heat shock protein 70 in growing feedlot cattle. J Anim Sci. 2013;91(1):120–129.
  • Singh AK, Upadhyay RC, Malakar D, et al. Effect of thermal stress on HSP70 expression in dermal fibroblast of zebu (Tharparkar) and crossbred (Karan-Fries) cattle. J Therm Biol. 2014;43:46–53.
  • Zhang FJ, Weng G, Wang JF, et al. Temperature-humidity index and chromium supplementation on antioxidant activity, heat shock protein 72, and cytokine response of lactating cows. J Anim Sci. 2014;92(7):3026–3034.
  • Mayengbam P, Tolenkhomba TC, Upadhyay RC. Expression of heat-shock protein 72 mRNA in relation to heart rate variability of Sahiwal and Karan-Fries in different temperature-humidity indices. Vet World. 2016;9(10):1051–1055.
  • Parmar MS, Madan AK, Huozha R, et al. Heat Shock Protein70 (HSP70) gene expression pattern in peripheral blood mononuclear cells (PBMCs) during different seasons in Sahiwal cows (Bos indicus). J Anim Res. 2015;5(1):109–113.
  • Bhanuprakash V, Singh U, Sengar G, et al. Differential effect of thermal stress on HSP70 expression, nitric oxide production and cell proliferation among native and crossbred dairy cattle. J Therm Biol. 2016;59:18–25.
  • Maibam U, Hooda O, Sharma P, et al. Expression of HSP70 genes in skin of zebu (Tharparkar) and crossbred (Karan Fries) cattle during different seasons under tropical climatic conditions. J Therm Biol. 2017;63:58–64.
  • Sheikh AA, Aggarwal A, Indu B, Aarif O. Inorganic zinc supplementation modulates heat shock and immune response in heat stressed peripheral blood mononuclear cells of periparturient dairy cows. Theriogenology. 2017;95:75–82.
  • Sengar GS, Deb R, Singh U, et al. Identification of differentially expressed microRNAs in Sahiwal (Bos indicus) breed of cattle during thermal stress. Cell Stress Chaperones. 2018;23(5):1019–1032.
  • Kumar J, Madan AK, Kumar M, et al. Impact of season on antioxidants, nutritional metabolic status, cortisol and heat shock proteins in Hariana and Sahiwal cattle. Biol Rhythm Res. 2018;49(1):29–38.
  • Kumar J, Yadav B, Madan AK, et al. Dynamics of heat-shock proteins, metabolic and endocrine responses during increasing temperature humidity index (THI) in lactating Hariana (Zebu) cattle. Biol Rhythm Res. 2020;51(6):934–950.
  • Saadeldin IM, Swelum AA, Zakri AM, et al. Effects of acute hyperthermia on the thermotolerance of cow and sheep skin-derived fibroblasts. Animals. 2020;10(4):545.
  • Kim WS, Nejad JG, Peng DQ, et al. Identification of heat shock protein gene expression in hair follicles as a novel indicator of heat stress in beef calves. Animal. 2020;14(7):1502–1508.
  • Khan A, Dou J, Wang Y, et al. Evaluation of heat stress effects on cellular and transcriptional adaptation of bovine granulosa cells. J Animal Sci Biotechnol. 2020;11:25.
  • Paula-Lopes FF, Chase CC, Jr, Al-Katanani YM, et al. Genetic divergence in cellular resistance to heat shock in cattle: differences between breeds developed in temperate versus hot climates in responses of preimplantation embryos, reproductive tract tissues and lymphocytes to increased culture temperatures. Reproduction. 2003;125(2):285–294.
  • Hernandez‐Ceron J, Chase CC, Hansen PJ. Differences in heat tolerance between preimplantation embryos from Brahman, Romosinuano, and Angus breeds. J. Dairy Sci. 2004;87(1):53–58.
  • Lamb M, Okimoto R, Broun M, Rosenkranes C. Jr. Associations between cattle breed and heat shock protein 70 gene. Res Ser. 2007;545:205–206.
  • Rosenkrans C, Jr, Banks A, Reiter S, Looper M. Calving traits of crossbred Brahman cows are associated with Heat Shock Protein 70 genetic polymorphisms. Anim Reprod Sci. 2010;119(3–4):178–182.
  • Charoensook R, Gatphayak K, Sharifi AR, et al. Polymorphisms in the bovine HSP90AB1 gene are associated with heat tolerance in Thai indigenous cattle. Trop Anim Health Prod. 2012;44(4):921–928.
  • Deb R, Sajjanar B, Singh U, et al. Promoter variants at AP2 box region of HSP70.1 affect thermal stress response and milk production traits in Frieswal cross bred cattle. Gene. 2013;532(2):230–235.
  • Hassan FU, Nawaz A, Rehman MS, et al. Prospects of HSP70 as a genetic marker for thermo-tolerance and immuno-modulation in animals under climate change scenario. Anim Nutr. 2019;5(4):340–350.
  • Cai Y, Liu Q, Xing G, et al. Polymorphism of the promoter region of HSP70 gene and its relationship with the expression of HSP70mRNA, HSF1mRNA, Bcl-2mrna and Bax-AMrna in lymphocytes in pheripheral blood of heat shocked dairy cows. Asian Australas J Anim Sci. 2005;18(5):734–740.
  • Li Q, Han J, Du F, et al. Novel SNPs in HSP70A1A gene and the association of polymorphisms with thermo tolerance traits and tissue specific expression in Chinese Holstein cattle. Mol Biol Rep. 2011;38(4):2657–2663.
  • Xiong Q, Chai J, Xiong H, et al. Association analysis of HSP70A1A haplotypes with heat tolerance in Chinese Holstein cattle. Cell Stress Chaperones. 2013;18(6):711–718.
  • Bhat S, Kumar P, Kashyap N, et al. Effect of heat shock protein 70 polymorphism on thermotolerance in Tharparkar cattle. Vet World. 2016;9(2):113–117.
  • Basirico L, Morera P, Primi V, et al. Cellular thermotolerance is associated with heat shock protein 70.1 genetic polymorphisms in Holstein lactating cows. Cell Stress Chaperones. 2011;16(4):441–448.
  • Said S, Putra WPB. Novel single nucleotide polymorphisms (SNPs) in the 5′UTR of bovine heat shock protein 70 (bHSP70) gene and its association with service per conception (S/C) of pasundan cattle. Biodiversitas. 2018;19(5):1622–1625.
  • Deb R, Sajjanar B, Singh U, et al. Effect of heat stress on the expression profile of HSP90 among Sahiwal (Bos indicus) and Frieswal (Bos indicus x Bos taurus) breed of cattle: a comparative study. Gene. 2014;536(2):435–440.
  • Katiyatiya CLF, Bradley G, Muchenje V. Thermotolerance, health profile and cellular expression of HSP90AB1 in Nguni and Boran cows raised on natural pastures under tropical conditions. J Therm Biol. 2017;69:85–94.
  • Bharati J, Dangi SS, Bag S, et al. Expression dynamics of HSP90 and nitric oxide synthase (NOS) isoforms during heat stress acclimation in Tharparkar cattle. Int J Biometeorol. 2017b;61(8):1461–1469.
  • Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116(2):281–297.
  • Sun XY, Li J, Fu L, et al. The Epigenetic Modification in Mammals under Heat Stress. World J Vet Sci. 2019;1:1005.
  • Baskerville S, Bartel DP. Microarray profiling of microRNAs reveals frequent coexpression with neighboring miRNAs and host genes. RNA. 2005;11(3):241–247.
  • Harfe BD. MicroRNAs in vertebrate development. Curr Opin Genet Dev. 2005;15(4):410–415.
  • McKenna LB, Schug J, Vourekas A, et al. MicroRNAs control intestinal epithelial differentiation, architecture, and barrier function. Gastroenterology. 2010;139(5):1654–1664.
  • Li Q, Yang C, Du J, et al. Characterization of miRNA profiles in the mammary tissue of dairy cattle in response to heat stress. BMC Genomics. 2018;19(1):975.
  • Ogorevc J, Kunej T, Dovc A, et al. Database of cattle candidate genes and genetic markers for milk production and mastitis. Anim Genet. 2009;40(6):832–851.
  • Fatima A, Waters S, O’Boyle P, Seoighe C, Morris DG. Alterations in hepatic miRNA expression during negative energy balance in postpartum dairy cattle. BMC Genom. 2014;15(1):28.
  • Kropp J, Salih SM, Khatib H. Expression of microRNAs in bovine and human pre-implantation embryo culture media. Front Genet. 2014;5:91.
  • Romao J, Jin W, He M, McAllister T, Guan L. MicroRNAs in bovine adipogenesis: genomic context, expression and function. BMC Genom. 2014;15(1):137.
  • Li R, Beaudoin F, Ammah AA, et al. Deep sequencing shows microRNA involvement in bovine mammary gland adaptation to diets supplemented with linseed oil or safflower oil. BMC Genom. 2015; 16:884.
  • Wang X, Song C, Zhou X, et al. Mitochondria associated microRNA expression profiling of heart failure. Biomed Res Int. 2017; 2017:4042509.
  • Dilda F, Gioia G, Pisani L, et al. Escherichia coli lipopolysaccharides and Staphylococcus aureus enterotoxin B differentially modulate inflammatory microRNAs in bovine monocytes. Vet J. 2012;192(3):514–516.
  • Cai M, Hu Y, Zheng T, et al. MicroRNA-216b inhibits heat stress-induced cell apoptosis by targeting Fas in bovine mammary epithelial cells. Cell Stress Chaperones. 2018;23(5):921–931.
  • Li HX, Wang Z, Zhang Z, et al. Effects of microRNA-24 on bovine mammary epithelial cells proliferation and apoptosis at high temperature. Scientia Agricultura Sinica. 2010;43:4732–4738.
  • Chen KL, Fu YY, Shi MY, et al. Down-regulation of mir-181a can reduce heat stress damage in PBMCs of Holstein cows. In Vitro Cell Dev Biol Anim. 2016;52(8):864–871.
  • Vanselow J, Vernunft A, Koczan D, et al. Exposure of lactating dairy cows to acute pre-ovulatory heat stress affects granulosa cell-specific gene expression profiles in dominant follicles. PLoS One. 2016;11(8):e0160600
  • Morelli FF, Mediani L, Heldens L, et al. An interaction study in mammalian cells demonstrates weak binding of HSPB2 to BAG3, which is regulated by HSPB3 and abrogated by HSPB8. Cell Stress Chaperones. 2017;22(4):531–540.
  • Sharma P, Sharma A, Sodhi M, et al. Characterizing binding sites of heat responsive microRNAs and their expression pattern in heat stressed PBMCs of native cattle, exotic cattle and riverine buffaloes. Mol Biol Rep. 2019;46(6):6513–6524.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.