S. A. Kayser, C. A. Vangilder, E. A. Ayello, and C. Lachenbruch, National Pressure Ulcer Advisory Panel, European Pressure Ulcer Advisory Panel, Pan Pacific Pressure Injury Alliance. Prevention and Treatment of Pressure Ulcers: Quick Reference Guide, Adv Skin Wound Care, vol.31, pp.276-285, 2018.

E. J. Benjamin, M. J. Blaha, S. E. Chiuve, M. Cushman, S. R. Das et al., Heart Disease and Stroke Statistics-2017 Update: A Report From the American Heart Association, Circulation, vol.135, pp.146-603, 2017.

D. Berlowitz, C. L. Vandeusen, V. Parker, A. Niederhauser, J. Silver et al., Preventing pressure ulcers in hospitals: A toolkit for improving quality of care

G. Claudia, J. M. Brown, N. E. Andrea, B. Michelle, and S. Lisette, Dealey Carol et al. Impact of Pressure Ulcers on Quality of Life in Older Patients: A Systematic Review, J Am Geriatr Soc, vol.57, pp.1175-1183, 2009.

B. M. Bates-jensen, M. Guihan, S. L. Garber, A. S. Chin, and S. P. Burns, Characteristics of Recurrent Pressure Ulcers in Veterans With Spinal Cord Injury, J Spinal Cord Med, vol.32, pp.34-42, 2009.

P. Wurzer, R. Winter, S. O. Stemmer, J. Ivancic, P. B. Lebo et al., Risk factors for recurrence of pressure ulcers after defect reconstruction, Wound Repair Regen Off Publ Wound Heal Soc Eur Tissue Repair Soc, vol.26, pp.64-68, 2018.

M. Schivo, A. A. Aksenov, A. Pasamontes, R. Cumeras, S. Weisker et al., A Rabbit Model for Assessment of Volatile Metabolite Changes Observed from Skin: a Pressure Ulcer Case Study, J Breath Res, vol.11, p.16007, 2017.

R. K. Daniel, D. L. Priest, and D. C. Wheatley, Etiologic factors in pressure sores: an experimental model, Arch Phys Med Rehabil, vol.62, pp.492-498, 1981.

E. Linder-ganz, S. Engelberg, M. Scheinowitz, and A. Gefen, Pressure-time cell death threshold for albino rat skeletal muscles as related to pressure sore biomechanics, J Biomech, vol.39, pp.2725-2732, 2006.

S. Loerakker, The relative contributions of muscle deformation and ischaemia to pressure ulcer development, 2011.

N. Lenche, D. Katerina, A. Nikolchev, P. Lidija, P. Biljana et al., The influence of comorbidity on the prevalence of pressure ulcers in geriatric patients, Glob Dermatol, vol.3, pp.319-322, 2016.

R. Salcido, A. Popescu, and C. Ahn, Animal models in pressure ulcer research, J Spinal Cord Med, vol.30, pp.107-116, 2007.

A. Stekelenburg, C. Oomens, and D. Bader, Compression-Induced Tissue Damage: Animal Models, Pressure Ulcer Research, pp.187-204, 2005.

T. K. Sin, X. M. Pei, B. T. Teng, E. W. Tam, B. Y. Yung et al., Oxidative stress and DNA damage signalling in skeletal muscle in pressure-induced deep tissue injury, Pflugers Arch, vol.465, pp.295-317, 2013.

P. M. Siu, B. T. Teng, X. M. Pei, and E. W. Tam, Proteasome inhibition alleviates prolonged moderate compressioninduced muscle pathology, BMC Musculoskelet Disord, vol.12, p.58, 2011.

B. T. Teng, E. W. Tam, I. F. Benzie, and P. M. Siu, Protective effect of caspase inhibition on compression-induced muscle damage, J Physiol, vol.589, pp.3349-3369, 2011.

B. T. Teng, X. M. Pei, E. W. Tam, I. F. Benzie, and P. M. Siu, Opposing responses of apoptosis and autophagy to moderate compression in skeletal muscle, Acta Physiol Oxf Engl, vol.201, pp.239-254, 2011.

Y. Wu, D. Van-der-schaft, F. P. Baaijens, and C. Oomens, Cell death induced by mechanical compression on engineered muscle results from a gradual physiological mechanism, J Biomech, vol.49, pp.1071-1077, 2016.

X. Wehrens and A. R. Marks, Ryanodine Receptors: Structure, function and dysfunction in clinical disease, 2006.

M. Kolaj, L. Zhang, and L. P. Renaud, L-type calcium channels and MAP kinase contribute to thyrotropin-releasing hormone-induced depolarization in thalamic paraventricular nucleus neurons, Am J Physiol Regul Integr Comp Physiol, vol.310, pp.1120-1127, 2016.

I. Swain, The Measurement of Interface Pressure, Pressure Ulcer Research, pp.51-71, 2005.

M. Hashimoto, T. Kurose, and S. Kawamata, Comparison between a weight compression and a magnet compression for experimental pressure ulcers in the rat. Histological studies and effects of anesthesia, Arch Histol Cytol, vol.71, pp.303-316, 2008.

A. M. Bellinger, S. Reiken, M. Dura, P. W. Murphy, S. Deng et al., Remodeling of ryanodine receptor complex causes "leaky" channels: A molecular mechanism for decreased exercise capacity, Proc Natl Acad Sci, vol.105, pp.2198-2202, 2008.

, Etude 1 : RyR1, un nouvel acteur d'escarre | 139

T. Kurose, M. Hashimoto, J. Ozawa, and S. Kawamata, Analysis of Gene Expression in Experimental Pressure Ulcers in the Rat with Special Reference to Inflammatory Cytokines, PloS One, vol.10, p.132622, 2015.

Y. Saito, M. Hasegawa, M. Fujimoto, T. Matsushita, M. Horikawa et al., The loss of MCP-1 attenuates cutaneous ischemia-reperfusion injury in a mouse model of pressure ulcer, J Invest Dermatol, vol.128, pp.1838-1851, 2008.

A. Gefen and D. Weihs, Cytoskeleton and plasma-membrane damage resulting from exposure to sustained deformations: A review of the mechanobiology of chronic wounds, Med Eng Phys, vol.38, pp.828-833, 2016.

N. S. Jagannathan and L. Tucker-kellogg, Membrane permeability during pressure ulcer formation: A computational model of dynamic competition between cytoskeletal damage and repair, J Biomech, vol.49, pp.1311-1320, 2016.

L. Jiang, Y. Dai, F. Cui, Y. Pan, H. Zhang et al., Expression of cytokines, growth factors and apoptosisrelated signal molecules in chronic pressure ulcer wounds healing, Spinal Cord, vol.52, pp.145-151, 2014.

D. Allen and J. Seo, ER Stress Activates the TOR Pathway through Atf6, J Mol Signal, vol.13, p.1, 2018.

R. F. Hillary and U. Fitzgerald, A lifetime of stress: ATF6 in development and homeostasis, J Biomed Sci, vol.25, p.48, 2018.

M. J. Peterson, N. Gravenstein, W. K. Schwab, J. H. Van-oostrom, and L. J. Caruso, Patient repositioning and pressure ulcer risk--monitoring interface pressures of at-risk patients, J Rehabil Res Dev, vol.50, pp.477-488, 2013.

S. Hagisawa and M. Ferguson-pell, Evidence supporting the use of two-hourly turning for pressure ulcer prevention, J Tissue Viability, vol.17, pp.76-81, 2008.

M. C. Nowycky, Intracellular calcium signaling, J Cell Sci, vol.115, pp.3715-3716, 2002.

G. Santulli, R. Nakashima, Q. Yuan, and A. R. Marks, Intracellular calcium release channels: an update, J Physiol, vol.595, pp.3041-3051, 2017.

A. M. Bellinger, S. Reiken, C. Carlson, M. Mongillo, X. Liu et al., Hypernitrosylated ryanodine receptor calcium release channels are leaky in dystrophic muscle, Nat Med, vol.15, pp.325-330, 2009.

J. Fauconnier, A. C. Meli, J. Thireau, S. Roberge, J. Shan et al., Ryanodine receptor leak mediated by caspase-8 activation leads to left ventricular injury after myocardial ischemia-reperfusion, Proc Natl Acad Sci U S A, vol.108, pp.13258-13263, 2011.
URL : https://hal.archives-ouvertes.fr/hal-02542114

S. Matecki, H. Dridi, B. Jung, N. Saint, S. R. Reiken et al., Leaky ryanodine receptors contribute to diaphragmatic weakness during mechanical ventilation, Proc Natl Acad Sci U S A, vol.113, pp.9069-9074, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01800610

H. Gissel, The role of Ca2+ in muscle cell damage, Ann N Y Acad Sci, vol.1066, pp.166-180, 2005.

F. Cui, Y. Pan, H. Xie, X. Wang, H. Shi et al., Pressure Combined with Ischemia/Reperfusion Injury Induces Deep Tissue Injury via Endoplasmic Reticulum Stress in a Rat Pressure Ulcer Model, Int J Mol Sci, vol.17, p.284, 2016.

L. Galluzzi, I. Vitale, J. M. Abrams, E. S. Alnemri, E. H. Baehrecke et al., Molecular definitions of cell death subroutines: recommendations of the Nomenclature Committee on Cell Death, Cell Death Differ, vol.19, pp.107-120, 2012.

P. C. Grisotto, A. C. Santos, J. Coutinho-netto, C. J. Piccinato, and C. E. , Indicators of oxidative injury and alterations of the cell membrane in the skeletal muscle of rats submitted to ischemia and reperfusion, J Surg Res, vol.92, pp.1-6, 2000.

M. Kruidering and G. I. Evan, Caspase-8 in Apoptosis: The Beginning of "The End, IUBMB, vol.50, pp.85-90

R. M. Murphy, R. J. Snow, and G. D. Lamb, mu-Calpain and calpain-3 are not autolyzed with exhaustive exercise in humans, Am J Physiol Cell Physiol, vol.290, pp.116-122, 2006.

N. Mizushima and T. Yoshimori, How to Interpret LC3 Immunoblotting, Autophagy, vol.3, pp.542-545, 2007.

S. Wu, C. Sun, D. Tian, Y. Li, X. Gao et al., Expression and clinical significances of Beclin1, LC3 and mTOR in colorectal cancer, Int J Clin Exp Pathol, vol.8, pp.3882-3891, 2015.

D. Bader and C. Oomens, The Potential of Biomarkers in the Early Detection of Pressure Ulcers, pp.1-15, 2018.

S. Loerakker, E. S. Huisman, H. Seelen, J. Glatz, F. Baaijens et al., Plasma variations of biomarkers for muscle damage in male nondisabled and spinal cord injured subjects, J Rehabil Res Dev, vol.49, pp.361-372, 2012.

S. Hagisawa, M. W. Ferguson-pell, V. R. Palmieri, and G. V. Cochran, Pressure sores: a biochemical test for early detection of tissue damage, Arch Phys Med Rehabil, vol.69, pp.668-671, 1988.

B. Hemmes, L. A. De-wert, P. Brink, C. Oomens, D. L. Bader et al., Cytokine IL1? and lactate as markers for tissue damage in spineboard immobilisation. A prospective, randomised open-label crossover trial, J Mech Behav Biomed Mater, vol.75, pp.82-88, 2017.

M. Makhsous, F. Lin, A. Pandya, M. S. Pandya, and C. C. Chadwick, Elevation in the serum and urine concentration of injury-related molecules after the formation of deep tissue injury in a rat spinal cord injury pressure ulcer model, PM R, vol.2, pp.1063-1065, 2010.

O. Stojadinovic, J. Minkiewicz, A. Sawaya, J. W. Bourne, P. Torzilli et al., Deep tissue injury in development of pressure ulcers: a decrease of inflammasome activation and changes in human skin morphology in response to aging and mechanical load, PloS One, vol.8, p.69223, 2013.

T. Minematsu, G. Nakagami, Y. Sari, T. Akase, J. Sugama et al., Candidate biomarkers for deep tissue damage from molecular biological and biochemical aspects, J Tissue Viability, vol.19, pp.77-83, 2010.

L. Jiang, Q. Tu, Y. Wang, and E. Zhang, Ischemia-reperfusion injury-induced histological changes affecting early stage pressure ulcer development in a rat model, Ostomy Wound Manage, vol.57, pp.55-60, 2011.

M. Suman, M. Giacomello, L. Corain, C. Ballarin, S. Montelli et al., Estradiol effects on intracellular Ca(2+) homeostasis in bovine brain-derived endothelial cells, Cell Tissue Res, vol.350, pp.109-118, 2012.

C. Bonne, Potential efficiency of antioxidants to prevent pressure ulcers. A neglected hypothesis, Med Hypotheses, vol.91, pp.28-31, 2016.

D. J. Margolis, J. Knauss, and W. Bilker, Hormone replacement therapy and prevention of pressure ulcers and venous leg ulcers, Lancet Lond Engl, vol.359, pp.675-677, 2002.

X. Liu, L. Harlow, Z. A. Graham, W. A. Bauman, and C. Cardozo, Spinal Cord Injury Leads to Hyperoxidation and Nitrosylation of Skeletal Muscle Ryanodine Receptor-1 Associated with Upregulation of Nicotinamide Adenine Dinucleotide Phosphate Oxidase 4, J. Neurotrauma, vol.34, pp.2069-2074, 2017.

A. Gélis, Pressure ulcer risk factors in persons with spinal cord injury Part 2: the chronic stage, Spinal Cord, vol.47, pp.651-661, 2009.

E. A. Kruger, M. Pires, Y. Ngann, M. Sterling, and S. Rubayi, Comprehensive management of pressure ulcers in spinal cord injury: Current concepts and future trends, J. Spinal Cord Med, vol.36, pp.572-585, 2013.

E. E. Dupont-versteegden, J. D. Houlé, C. M. Gurley, and C. A. Peterson, Early changes in muscle fiber size and gene expression in response to spinal cord transection and exercise, Am. J. Physiol, vol.275, pp.1124-1133, 1998.

R. Scelsi, Skeletal muscle pathology after spinal cord injury: Our 20 year experience and results on skeletal muscle changes in paraplegics, related to functional rehabilitation, Basic Appl Myol, vol.11, pp.75-85, 2001.

M. R. Carry, S. P. Ringel, and J. M. Starcevich, Distribution of capillaries in normal and diseased human skeletal muscle, Muscle Nerve, vol.9, pp.445-454, 1986.

M. A. Petrie, M. Suneja, E. Faidley, and R. K. Shields, Low force contractions induce fatigue consistent with muscle mRNA expression in people with spinal cord injury, Physiol. Rep, vol.2, p.248, 2014.

W. R. Frontera, Single muscle fiber size and contractility after spinal cord injury in rats, Muscle Nerve, vol.34, pp.101-104, 2006.

O. B. Clarke and W. A. Hendrickson, Structures of the Colossal RyR1 Calcium Release Channel, Curr. Opin. Struct. Biol, vol.39, pp.144-152, 2016.

J. W. Witherspoon and K. G. Meilleur, Review of RyR1 pathway and associated pathomechanisms, Acta Neuropathol. Commun, vol.4, 2016.

A. M. Bellinger, Hypernitrosylated ryanodine receptor calcium release channels are leaky in dystrophic muscle, Nat. Med, vol.15, pp.325-330, 2009.

D. C. Andersson, Ryanodine receptor oxidation causes intracellular calcium leak and muscle weakness in aging, Cell Metab, vol.14, pp.196-207, 2011.

A. M. Bellinger, Remodeling of ryanodine receptor complex causes "leaky" channels: A molecular mechanism for decreased exercise capacity, Proc. Natl. Acad. Sci. 105, pp.2198-2202, 2008.

M. C. Nowycky, Intracellular calcium signaling, J. Cell Sci, vol.115, pp.3715-3716, 2002.

G. Santulli, R. Nakashima, Q. Yuan, and A. R. Marks, Intracellular calcium release channels: an update, J. Physiol, vol.595, pp.3041-3051, 2017.

R. Scelsi, L. Scelsi, R. Bocchi, and S. Lotta, Morphological changes in the skin microlymphatics in recently injured paraplegic patients with ilio-femoral venous thrombosis, Paraplegia, vol.33, pp.472-475, 1995.

M. D. Siperstein, P. Raskin, and H. Burns, Electron microscopic quantification of diabetic microangiopathy, Diabetes, vol.22, pp.514-527, 1973.

S. Latimer, W. Chaboyer, and B. M. Gillespie, The repositioning of hospitalized patients with reduced mobility: a prospective study, Nurs. Open, vol.2, pp.85-93, 2015.

M. Zomorodi, D. Topley, and M. Mcanaw, Developing a Mobility Protocol for Early Mobilization of Patients in a Surgical/Trauma ICU, Crit. Care Res. Pract, 2012.

S. Hagisawa and M. Ferguson-pell, Evidence supporting the use of two-hourly turning for pressure ulcer prevention, J. Tissue Viability, vol.17, pp.76-81, 2008.

R. F. Capogrosso, Ryanodine channel complex stabilizer compound S48168/ARM210 as a disease modifier in dystrophin-deficient mdx mice: proof-of-concept study and independent validation of efficacy, FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol, vol.32, pp.1025-1043, 2018.

N. Shabshin, G. Zoizner, A. Herman, V. Ougortsin, and A. Gefen, Use of weight-bearing MRI for evaluating wheelchair cushions based on internal soft-tissue deformations under ischial tuberosities, J. Rehabil. Res. Dev, vol.47, pp.31-42, 2010.

E. Linder-ganz, G. Yarnitzky, Z. Yizhar, I. Siev-ner, and A. Gefen, Real-time finite element monitoring of sub-dermal tissue stresses in individuals with spinal cord injury: toward prevention of pressure ulcers, Ann. Biomed. Eng, vol.37, pp.387-400, 2009.

E. Landis, Micro-injection studies of capillary blood pressure in human skin, Heart, vol.15, pp.209-228, 1930.

E. Linder-ganz, S. Engelberg, M. Scheinowitz, and A. Gefen, Pressure-time cell death threshold for albino rat skeletal muscles as related to pressure sore biomechanics, J. Biomech, vol.39, pp.2725-2732, 2006.

V. Luboz, Personalized modeling for real-time pressure ulcer prevention in sitting posture, J. Tissue Viability, vol.27, pp.54-58, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01930078

S. Loerakker, Aetiology of pressure ulcers, p.31, 2007.

E. Leopold and A. Gefen, A simple stochastic model to explain the sigmoid nature of the strain-time cellular tolerance curve, J. Tissue Viability, vol.21, pp.27-36, 2012.

R. G. Breuls, C. V. Bouten, C. W. Oomens, D. L. Bader, and F. P. Baaijens, Compression induced cell damage in engineered muscle tissue: an in vitro model to study pressure ulcer aetiology, Ann. Biomed. Eng, vol.31, pp.1357-1364, 2003.

D. Bader, Biochemical Status of Soft Tissues Subjected to Sustained Pressure, Pressure Ulcer Research: Current and Future Perspectives, pp.109-127, 2005.

E. Leopold, R. Sopher, and A. Gefen, The effect of compressive deformations on the rate of build-up of oxygen in isolated skeletal muscle cells, Med. Eng. Phys, vol.33, pp.1072-1078, 2011.

S. Arias, Effects on interface pressure and tissue oxygenation under ischial tuberosities during the application of an alternating cushion, J. Tissue Viability, vol.24, pp.91-101, 2015.

D. Bader, C. Oomens, M. Romanelli, M. Clark, A. Gefen et al., The Potential of Biomarkers in the Early Detection of Pressure Ulcers. in Science and Practice of Pressure Ulcer Management, pp.1-15, 2018.

R. Sopher, J. Nixon, C. Gorecki, and A. Gefen, Effects of Intramuscular Fat Infiltration, Scarring, and Spasticity on the Risk for Sitting-Acquired Deep Tissue Injury in Spinal Cord Injury Patients, J. Biomech. Eng, vol.133, pp.21011-021011, 2011.

S. Loerakker, Plasma variations of biomarkers for muscle damage in male nondisabled and spinal cord injured subjects, J. Rehabil. Res. Dev, vol.49, pp.361-372, 2012.

T. W. Du-clos, Function of C-reactive protein, Ann. Med, vol.32, pp.274-278, 2000.

G. P. Ladwig, Ratios of activated matrix metalloproteinase-9 to tissue inhibitor of matrix metalloproteinase-1 in wound fluids are inversely correlated with healing of pressure ulcers, Wound Repair Regen. Off. Publ. Wound Heal. Soc. Eur. Tissue Repair Soc, vol.10, pp.26-37, 2002.

, Etude 3 : Le graphène pour booster la cicatrisation | 173

, Representative photographs of the wound evolution for SHAM and graphene. (b) Representative hematoxylin eosin saffron (HES) at day 0, 4, 10, and 16 after wound excision. Neo-epithelialization was progressing at the same rate at day 21. Scale black bar: 200 µm and scale red bar: 100 µm. (c) Dot plot of the re-epithelialization, Graphene accelerates early phase of healing on a chronic wound model (a)

X. Wang, J. Ge, E. E. Tredget, and Y. Wu, The mouse excisional wound splinting model, including applications for stem cell transplantation, Nat Protoc, vol.8, pp.302-309, 2013.

B. Lu, T. Li, H. Zhao, X. Li, C. Gao et al., Graphene-based composite materials beneficial to wound healing, Nanoscale, vol.4, pp.2978-2982, 2012.

C. Heo, J. Yoo, S. Lee, J. A. Jung, S. Yoo et al., The control of neural cell-to-cell interactions through noncontact electrical field stimulation using graphene electrodes, Biomaterials, vol.32, pp.19-27, 2011.

S. Pattnaik, K. Swain, and Z. Lin, Graphene and -based nanocomposites: biomedical applications and biosafety, J Mater Chem B, vol.4, pp.7813-7831, 2016.

S. Kumar and K. Chatterjee, Comprehensive Review on the Use of Graphene-Based Substrates for Regenerative Medicine and Biomedical Devices, ACS Appl Mater Interfaces, vol.8, pp.26431-26457, 2016.

V. W. Wong, M. Sorkin, J. P. Glotzbach, M. T. Longaker, and G. C. Gurtner, Surgical Approaches to Create Murine Models of Human Wound Healing, BioMed Res. Int, 2011.

H. D. Zomer and A. G. Trentin, Skin wound healing in humans and mice: Challenges in translational research, J Dermatol Sci, vol.90, pp.3-12, 2018.

R. G. Frykberg and J. Banks, Challenges in the Treatment of Chronic Wounds, Adv Wound Care, vol.4, pp.560-582, 2015.

G. Han and R. Ceilley, Chronic Wound Healing: A Review of Current Management and Treatments, Adv Ther, vol.34, pp.599-610, 2017.

H. Shi, H. Xie, Y. Zhao, C. Lin, F. Cui et al., Myoprotective effects of bFGF on skeletal muscle injury in pressure-related deep tissue injury in rats, Burns Trauma, vol.4, p.26, 2016.

R. Sartori, G. Milan, M. Patron, C. Mammucari, B. Blaauw et al., Smad2 and 3 transcription factors control muscle mass in adulthood, Am J Physiol Cell Physiol, vol.296, pp.1248-1257, 2009.

V. Falanga, L. Zhou, and T. Yufit, Low oxygen tension stimulates collagen synthesis and COL1A1 transcription through the action of TGF-beta1, J Cell Physiol, vol.191, pp.42-50, 2002.

G. Dow, A. Browne, and R. G. Sibbald, Infection in chronic wounds: controversies in diagnosis and treatment, Ostomy Wound Manage, vol.45, pp.41-42, 1999.

A. R. Siddiqui and J. M. Bernstein, Chronic wound infection: facts and controversies, Clin Dermatol, vol.28, pp.519-526, 2010.

W. Mcguiness, E. Vella, and D. Harrison, Influence of dressing changes on wound temperature, J Wound Care, vol.13, pp.383-385, 2004.

D. A. Kliot and S. J. Birnbaum, Thermographic studies of wound healing, Am J Obstet Gynecol, vol.93, pp.515-521, 1965.

G. Nakagami, H. Sanada, S. Iizaka, T. Kadono, T. Higashino et al., Predicting delayed pressure ulcer healing using thermography: a prospective cohort study, J Wound Care, vol.19, pp.465-472, 2010.

D. Sigaudo-roussel, C. Demiot, B. Fromy, A. Koïtka, G. Lefthériotis et al., Early Endothelial Dysfunction Severely Impairs Skin Blood Flow Response to Local Pressure Application in Streptozotocin-Induced Diabetic Mice, Diabetes, vol.53, pp.1564-1569, 2004.

C. Yu, K. Leung, K. Fung, F. Lam, E. Ng et al., The characterization of a fullthickness excision open foot wound model in n5-streptozotocin (STZ)-induced type 2 diabetic rats that mimics diabetic foot ulcer in terms of reduced blood circulation, higher C-reactive protein, elevated inflammation, and reduced cell proliferation, Exp Anim, vol.66, pp.259-269, 2017.

K. Cheng, Z. Lin, Y. Cheng, H. Chiu, N. Yeh et al., Wound Healing in Streptozotocin-Induced Diabetic Rats Using Atmospheric-Pressure Argon Plasma, Jet. Sci Rep, vol.8, p.12214, 2018.

K. Kawamoto, H. Miyaji, E. Nishida, S. Miyata, A. Kato et al., Characterization and evaluation of graphene oxide scaffold for periodontal wound healing of class II furcation defects in dog, Int J Nanomedicine, vol.13, pp.2365-2376, 2018.

Y. Li, X. Liu, L. Tan, Z. Cui, X. Yang et al., Rapid Sterilization and Accelerated Wound Healing Using Zn2+ and Graphene Oxide Modified g-C3N4 under Dual Light Irradiation, Adv Funct Mater, vol.28, p.1800299, 2018.

R. Senthil, R. Berly, B. Ram, T. Gobi, and N. , Electrospun poly(vinyl) alcohol/collagen nanofibrous scaffold hybridized by graphene oxide for accelerated wound healing, Int J Artif Organs, vol.41, pp.467-473, 2018.

J. Chu, P. Shi, W. Yan, J. Fu, Z. Yang et al., PEGylated graphene oxide-mediated quercetin-modified collagen hybrid scaffold for enhancement of MSCs differentiation potential and diabetic wound healing, Nanoscale, vol.10, pp.9547-9560, 2018.

V. Bouchiat, J. Coraux, and Z. Han, Process and device for forming a graphene layer, 2018.

Z. Fan, B. Liu, J. Wang, S. Zhang, Q. Lin et al., A Novel Wound Dressing Based on Ag/Graphene Polymer Hydrogel: Effectively Kill Bacteria and Accelerate Wound Healing, Adv Funct Mater, vol.24, pp.3933-3943, 2014.

M. Tang, Q. Song, N. Li, Z. Jiang, R. Huang et al., Enhancement of electrical signaling in neural networks on graphene films, Biomaterials, vol.34, pp.6402-6411, 2013.

N. Li, Q. Zhang, S. Gao, Q. Song, R. Huang et al., Three-dimensional graphene foam as a biocompatible and conductive scaffold for neural stem cells, Sci Rep, vol.3, p.1604, 2013.

S. Barg, F. M. Perez, N. Ni, V. Pereira-p-do, R. C. Maher et al., Mesoscale assembly of chemically modified graphene into complex cellular networks, Nat Commun, vol.5, p.4328, 2014.

M. Jaspers, I. Maltha, J. Klaessens, H. De-vet, R. M. Verdaasdonk et al., Insights into the use of thermography to assess burn wound healing potential: a reliable and valid technique when compared to laser Doppler imaging, J Biomed Opt, vol.21, p.96006, 2016.

N. J. Prindeze, P. Fathi, M. J. Mino, N. A. Mauskar, T. E. Travis et al., Examination of the Early Diagnostic Applicability of Active Dynamic Thermography for Burn Wound Depth Assessment and Concept Analysis, J Burn Care Res Off Publ Am Burn Assoc, vol.36, pp.626-635, 2015.

C. Huang, Y. Wu, C. Hwang, Y. Jong, C. Chao et al., The application of infrared thermography in evaluation of patients at high risk for lower extremity peripheral arterial disease, J Vasc Surg, vol.54, pp.1074-1080, 2011.

A. Peleki and A. Da-silva, Novel Use of Smartphone-based Infrared Imaging in the Detection of Acute Limb Ischaemia, EJVES Short Rep, vol.32, pp.1-3, 2016.

M. A. Garduño-ramón, S. G. Vega-mancilla, L. A. Morales-henández, and R. A. Osornio-rios, Supportive Noninvasive Tool for the Diagnosis of Breast Cancer Using a Thermographic Camera as Sensor, Sensors, vol.17, 2017.

N. Hussain, D. Connah, H. Ugail, P. A. Cooper, R. A. Falconer et al., The use of thermographic imaging to evaluate therapeutic response in human tumour xenograft models, Sci Rep, vol.6, p.31136, 2016.

W. Xie, P. Mccahon, K. Jakobsen, and C. Parish, Evaluation of the ability of digital infrared imaging to detect vascular changes in experimental animal tumours, Int J Cancer, vol.108, pp.790-794, 2004.

H. Zheng, R. Ma, M. Gao, X. Tian, Y. Li et al., Antibacterial applications of graphene oxides: structure-activity relationships, molecular initiating events and biosafety, Sci Bull, vol.63, pp.133-142, 2018.

M. Aslam, M. S. Ahmad, and M. A. Ahmad, Application of Graphene, Graphene oxide and their derivatives as Wound healing: A Brief Review, Adv Environ Biol, vol.11, pp.1-5, 2017.

K. Kawamoto, H. Miyaji, E. Nishida, S. Miyata, A. Kato et al., Characterization and evaluation of graphene oxide scaffold for periodontal wound healing of class II furcation defects in dog, Int J Nanomedicine, vol.13, pp.2365-2376, 2018.

B. O. Adegoke, A. C. Odole, L. O. Akindele, and A. O. Akinpelu, Pressure ulcer prevalence among hospitalised adults in university hospitals in South-west Nigeria, Wound Pract. Res. J. Aust. Wound Manag. Assoc, vol.21, p.128, 2013.

K. Agrawal and N. Chauhan, Pressure ulcers: Back to the basics, Indian J. Plast. Surg. Off. Publ. Assoc. Plast. Surg. India, vol.45, pp.244-254, 2012.

J. T. Alander, I. Kaartinen, A. Laakso, T. Pätilä, T. Spillmann et al., A review of indocyanine green fluorescent imaging in surgery, Int. J. Biomed. Imaging, p.940585, 2012.

J. Alderden, J. D. Whitney, S. M. Taylor, and S. Zaratkiewicz, Risk Profile Characteristics Associated With Outcomes of Hospital-Acquired Pressure Ulcers: A Retrospective Review, Crit. Care Nurse, vol.31, pp.30-43, 2011.

Y. Amir, F. E. Tan, R. Halfens, C. Lohrmann, and J. Schols, Pressure Ulcer Prevalence and Care in Indonesian Hospitals: A Multicenter, Cross-sectional Evaluation Using an Extended Donabedian Model, Ostomy. Wound Manage, vol.63, pp.8-23, 2017.

L. W. Andersen, J. Mackenhauer, J. C. Roberts, K. M. Berg, M. N. Cocchi et al., Etiology and therapeutic approach to elevated lactate, Mayo Clin. Proc, vol.88, pp.1127-1140, 2013.

D. C. Andersson and A. R. Marks, Fixing ryanodine receptor Ca2+ leak -a novel therapeutic strategy for contractile failure in heart and skeletal muscle, Drug Discov. Today Dis. Mech, vol.7, pp.151-157, 2010.

D. C. Andersson, M. J. Betzenhauser, S. Reiken, A. C. Meli, A. Umanskaya et al., Ryanodine receptor oxidation causes intracellular calcium leak and muscle weakness in aging, Cell Metab, vol.14, pp.196-207, 2011.

H. Arao, M. Obata, T. Shimada, and S. Hagisawa, Morphological characteristics of the dermal papillae in the development of pressure sores, J. Tissue Viability, vol.8, pp.17-23, 1998.

D. G. Armstrong, L. A. Lavery, P. J. Liswood, W. F. Todd, and J. A. Tredwell, Infrared dermal thermometry for the high-risk diabetic foot, Phys. Ther, vol.77, pp.176-177, 1997.

R. A. Atkinson and N. A. Cullum, Interventions for pressure ulcers: a summary of evidence for prevention and treatment, Spinal Cord, vol.56, pp.186-198, 2018.

F. Aubert-viard, Conception et évaluation d'un pansement multicouche antibactérien pour le traitement des plaies chroniques, 2014.

P. A. Bacon, A. J. Collins, and J. A. Cosh, Thermographic assessment of the anti-inflammatory effect of flurbiprofen in rheumatoid arthritis, Curr. Med. Res. Opin, vol.3, pp.20-26, 1975.

D. Bader, Y. Wang, S. Knight, A. Polliack, T. James et al., Biochemical Status of Soft Tissues Subjected to Sustained Pressure, Pressure Ulcer Research: Current and Future Perspectives, pp.109-127, 2005.

R. I. Barnett, A. , and J. A. , Skin vascular reaction to standard patient positioning on a hospital mattress, Adv. Wound Care J. Prev. Heal, vol.7, pp.58-65, 1994.

B. Barrois, C. Labalette, P. Rousseau, A. Corbin, D. Colin et al., A national prevalence study of pressure ulcers in French hospital inpatients, J. Wound Care, vol.17, pp.378-379, 2008.

K. Bauer, K. Rock, M. Nazzal, O. Jones, and W. Qu, Pressure Ulcers in the United States' Inpatient Population From 2008 to 2012: Results of a Retrospective Nationwide Study, Ostomy. Wound Manage, vol.62, pp.30-38, 2016.

W. A. Bauman and A. M. Spungen, Disorders of carbohydrate and lipid metabolism in veterans with paraplegia or quadriplegia: a model of premature aging, Metabolism, vol.43, pp.749-756, 1994.

A. M. Bellinger, M. Mongillo, and A. R. Marks, Stressed out: the skeletal muscle ryanodine receptor as a target of stress, J. Clin. Invest, vol.118, pp.445-453, 2008.

A. M. Bellinger, S. Reiken, C. Carlson, M. Mongillo, X. Liu et al., Hypernitrosylated ryanodine receptor calcium release channels are leaky in dystrophic muscle, Nat. Med, vol.15, pp.325-330, 2009.

S. Bergstrand, U. Källman, A. Ek, L. Lindberg, M. Engström et al., Pressureinduced vasodilation and reactive hyperemia at different depths in sacral tissue under clinically relevant conditions, pp.761-771, 2014.

D. M. Bers, Cardiac excitation-contraction coupling, 2002.

S. Bhattacharya and R. K. Mishra, Pressure ulcers: Current understanding and newer modalities of treatment, Indian J. Plast. Surg. Off. Publ. Assoc. Plast. Surg. India, vol.48, pp.4-16, 2015.

H. Bichraoui, Identification de nouveaux déterminants moléculaires de l'interaction du récepteur des Dihydropyridines avec le récepteur à la Ryanodine. phdthesis, 2010.

F. W. Blaisdell, The pathophysiology of skeletal muscle ischemia and the reperfusion syndrome: a review, Cardiovasc. Surg, vol.10, pp.620-630, 2002.

B. Blaylock and C. Gardner, Measuring tissue interface pressures of two support surfaces used in the operating room, Ostomy. Wound Manage, vol.40, p.48, 1994.

K. Bogie and D. Bader, Susceptibility of Spinal Cord-Injured Individuals to Pressure Ulcers, Pressure Ulcer Research: Current and Future Perspectives, pp.73-88, 2005.

M. Bonnefoy, J. Coulon, J. Bienvenu, R. C. Boisson, and L. Rys, Implication of Cytokines in the Aggravation of Malnutritution and Hypercatabolism in Elderly Patients with Severe Pressure Sores, Age Ageing, vol.24, pp.37-42, 1995.

A. B. Borisov, S. Huang, and B. M. Carlson, Remodeling of the vascular bed and progressive loss of capillaries in denervated skeletal muscle, Anat. Rec, vol.258, pp.292-304, 2000.

E. M. Bosboom, C. V. Bouten, C. W. Oomens, F. P. Baaijens, and K. Nicolay, Quantifying pressure sore-related muscle damage using high-resolution MRI, J. Appl. Physiol. Bethesda Md, vol.95, pp.2235-2240, 1985.

C. V. Bouten, Etiology and pathology of pressure sores:a literature review, 1996.

P. Brancaccio, G. Lippi, and N. Maffulli, Biochemical markers of muscular damage, Clin. Chem. Lab. Med, vol.48, pp.757-767, 2010.

M. L. Brandão, J. E. Roselino, C. E. Piccinato, C. , and J. , Mitochondrial alterations in skeletal muscle submitted to total ischemia, J. Surg. Res, vol.110, pp.235-240, 2003.

R. G. Breuls, C. V. Bouten, C. W. Oomens, D. L. Bader, and F. P. Baaijens, Compression induced cell damage in engineered muscle tissue: an in vitro model to study pressure ulcer aetiology, Ann. Biomed. Eng, vol.31, pp.1357-1364, 2003.

D. Brienza, S. Krishnan, P. Karg, G. Sowa, A. et al., Predictors of pressure ulcer incidence following traumatic spinal cord injury: a secondary analysis of a prospective longitudinal study, Spinal Cord, vol.56, pp.28-34, 2018.

A. Brill, How to avoid interface problems in acute noninvasive ventilation, Breathe, vol.10, pp.230-242, 2014.

D. Bronneberg, Biochemical markers for early detection of superficial pressure ulcers, 2007.

D. Bronneberg, C. V. Bouten, C. W. Oomens, P. M. Van-kemenade, and F. P. Baaijens, An in vitro Model System to Study the Damaging Effects of Prolonged Mechanical Loading of the Epidermis, Ann. Biomed. Eng, vol.34, pp.506-514, 2006.

D. Bronneberg, S. W. Spiekstra, L. H. Cornelissen, C. W. Oomens, S. Gibbs et al., Cytokine and chemokine release upon prolonged mechanical loading of the epidermis, Exp. Dermatol, vol.16, pp.567-573, 2007.

T. J. Burkholder, Mechanotransduction in skeletal muscle, Front. Biosci. J. Virtual Libr, vol.12, pp.174-191, 2007.

J. C. Calderón, P. Bolaños, and C. Caputo, The excitation-contraction coupling mechanism in skeletal muscle, Biophys. Rev, vol.6, pp.133-160, 2014.

M. R. Carry, S. P. Ringel, and J. M. Starcevich, Distribution of capillaries in normal and diseased human skeletal muscle, Muscle Nerve, vol.9, pp.445-454, 1986.

N. W. Carter, F. C. Rector, D. S. Campion, and D. W. Seldin, Measurement of Intracellular pH of Skeletal Muscle with pH-sensitive Glass Microelectrodes*, J. Clin. Invest, vol.46, pp.920-933, 1967.

F. B. Cerra, T. Z. Lajos, M. Montes, and J. H. Siegel, Hemorrhagic infarction: A reperfusion injury following prolonged myocardial ischemic anoxia, Surgery, vol.78, pp.95-104, 1975.

A. Chanmugam, D. Langemo, K. Thomason, J. Haan, E. A. Altenburger et al., Relative Temperature Maximum in Wound Infection and Inflammation as Compared with a Control Subject Using Long-Wave Infrared Thermography, Adv. Skin Wound Care, vol.30, p.406, 2017.

M. E. Chaves, F. S. Silva, . Da, V. P. Soares, R. A. Ferreira et al., Evaluation of healing of pressure ulcers through thermography: a preliminary study, Res. Biomed. Eng, vol.31, pp.3-9, 2015.

M. Clark, M. J. Semple, N. Ivins, K. Mahoney, and K. Harding, National audit of pressure ulcers and incontinence-associated dermatitis in hospitals across Wales: a cross-sectional study, BMJ Open, vol.7, 2017.

O. B. Clarke and W. A. Hendrickson, Structures of the Colossal RyR1 Calcium Release Channel, Curr. Opin. Struct. Biol, vol.39, pp.144-152, 2016.

S. Coleman, J. Nixon, J. Keen, L. Wilson, E. Mcginnis et al., A new pressure ulcer conceptual framework, J. Adv. Nurs, vol.70, pp.2222-2234, 2014.

A. Contreras-ferrat, S. Lavandero, E. Jaimovich, and A. Klip, Calcium signaling in insulin action on striated muscle, Cell Calcium, vol.56, pp.390-396, 2014.

L. H. Cornelissen, D. Bronneberg, D. L. Bader, F. P. Baaijens, and C. W. Oomens, The transport profile of cytokines in epidermal equivalents subjected to mechanical loading, Ann. Biomed. Eng, vol.37, pp.1007-1018, 2009.

K. J. Cowan and K. B. Storey, Mitogen-activated protein kinases: new signaling pathways functioning in cellular responses to environmental stress, J. Exp. Biol, vol.206, pp.1107-1115, 2003.

J. Cox and S. Roche, Vasopressors and development of pressure ulcers in adult critical care patients, Am. J. Crit. Care Off. Publ. Am. Assoc. Crit.-Care Nurses, vol.24, pp.501-510, 2015.

J. J. Cragg, V. K. Noonan, M. Dvorak, A. Krassioukov, G. B. Mancini et al., Spinal cord injury and type 2 diabetes, Neurology, vol.81, pp.1864-1868, 2013.

|. Bibliographie,

F. Cui, Y. Pan, H. Xie, X. Wang, H. Shi et al., , 2016.

, Pressure Combined with Ischemia/Reperfusion Injury Induces Deep Tissue Injury via Endoplasmic Reticulum Stress in a Rat Pressure Ulcer Model, Int. J. Mol. Sci, vol.17, p.284

J. Dale, E. Myhre, and K. Rasmussen, Elimination of Hemoglobin and Lactate Dehydrogenase from Plasma in Normals and Patients with Intravascular Hemolysis. Scand, J. Clin. Lab. Invest, vol.29, pp.389-395, 1972.

T. Defloor, The risk of pressure sores: a conceptual scheme, J. Clin. Nurs, vol.8, pp.206-216, 1999.

T. N. Demidova-rice, M. R. Hamblin, and I. M. Herman, Acute and Impaired Wound Healing: Pathophysiology and Current Methods for Drug Delivery, Part 1: Normal and Chronic Wounds: Biology, Causes, and Approaches to Care, Adv. Skin Wound Care, vol.25, pp.304-314, 2012.

N. S. Dhalla, A. B. Elmoselhi, T. Hata, and N. Makino, Status of myocardial antioxidants in ischemiareperfusion injury, Cardiovasc. Res, vol.47, pp.446-456, 2000.

F. Dini, R. Capuano, T. Strand, A. Ek, M. Lindgren et al., , 2013.

K. T. Dodd and D. R. Gross, Three-dimensional tissue deformation in subcutaneous tissues overlying bony prominences may help to explain external load transfer to the interstitium, J. Biomech, vol.24, pp.11-19, 1991.

D. J. Drake, M. Swanson, G. Baker, M. Pokorny, M. A. Rose et al., The association of BMI and Braden total score on the occurrence of pressure ulcers, J. Wound Ostomy Cont. Nurs. Off. Publ. Wound Ostomy Cont. Nurses Soc, vol.37, pp.367-371, 2010.

T. W. Du-clos, Function of C-reactive protein, Ann. Med, vol.32, pp.274-278, 2000.

A. F. Dulhunty and C. Franzini-armstrong, The relative contributions of the folds and caveolae to the surface membrane of frog skeletal muscle fibres at different sarcomere lengths, J. Physiol, vol.250, pp.513-539, 1975.

N. K. Durrani, R. Yavuzer, V. Mittal, M. M. Bradford, C. Lobocki et al., The effect of gradually increased blood flow on ischemia-reperfusion injury in rat kidney, Am. J. Surg, vol.191, pp.334-337, 2006.

S. Ebashi, Excitation-Contraction Coupling and the Mechanism of Muscle Contraction, Annu. Rev. Physiol, vol.53, pp.1-17, 1991.

L. E. Edsberg, J. T. Wyffels, R. Ogrin, B. C. Craven, and P. Houghton, A pilot study evaluating protein abundance in pressure ulcer fluid from people with and without spinal cord injury, J. Spinal Cord Med, vol.38, pp.456-467, 2015.

P. Elias, S. Ahn, B. Brown, D. Crumrine, and K. R. Feingold, Origin of the epidermal calcium gradient: regulation by barrier status and role of active vs passive mechanisms, J. Invest. Dermatol, vol.119, pp.1269-1274, 2002.

H. K. Eltzschig and T. Eckle, Ischemia and reperfusion-from mechanism to translation, Nat. Med, vol.17, 2011.

S. A. Eming, P. Martin, and M. Tomic-canic, Wound repair and regeneration: Mechanisms, signaling, and translation, Sci. Transl. Med, vol.6, pp.265-271, 2014.

A. Espinosa, M. Estrada, J. , and E. , IGF-I and insulin induce different intracellular calcium signals in skeletal muscle cells, J. Endocrinol, vol.182, pp.339-352, 2004.

V. Falanga, L. Zhou, Y. , and T. , Low oxygen tension stimulates collagen synthesis and COL1A1 transcription through the action of TGF-beta1, J. Cell. Physiol, vol.191, pp.42-50, 2002.

S. Farah, M. A. Sabry, A. F. Shubaili, J. T. Anim, J. M. Hussain et al., Hereditary spastic paraparesis with distal muscle wasting, microcephaly, mental retardation, arachnodactyly and tremors: new entity?, Clin. Neurol. Neurosurg, vol.99, pp.66-70, 1997.

J. Fauconnier, A. C. Meli, J. Thireau, S. Roberge, J. Shan et al., Ryanodine receptor leak mediated by caspase-8 activation leads to left ventricular injury after myocardial ischemia-reperfusion, Proc. Natl. Acad. Sci. U. S. A, vol.108, pp.13258-13263, 2011.
URL : https://hal.archives-ouvertes.fr/hal-02542114

Y. Feng, J. Chai, W. Chu, L. Ma, P. Zhang et al., Combination of ketamine and xylazine exacerbates cardiac dysfunction in severely scalded rats during the shock stage, Exp. Ther. Med, vol.6, pp.641-648, 2013.

M. Ferguson-pell and S. Hagisawa, Biochemical changes in sweat following prolonged ischemia, J. Rehabil. Res. Dev, vol.25, pp.57-62, 1988.

Y. Ferraq, Développement d'un modèle de cicatrisation épidermique après une désépidermisation laser. thesis, vol.3, 2007.

M. Fierheller and R. G. Sibbald, A clinical investigation into the relationship between increased periwound skin temperature and local wound infection in patients with chronic leg ulcers, Adv. Skin Wound Care, vol.23, pp.380-381, 2010.

H. Frankel, J. Sperry, and L. Kaplan, Risk factors for pressure ulcer development in a best practice surgical intensive care unit, Am. Surg, vol.73, pp.1215-1217, 2007.

B. Fraysse, J. Desaphy, J. Rolland, S. Pierno, A. Liantonio et al., Fiber type-related changes in rat skeletal muscle calcium homeostasis during aging and restoration by growth hormone, Neurobiol. Dis, vol.21, pp.372-380, 2006.

B. Fromy, E. Lingueglia, D. Sigaudo-roussel, J. L. Saumet, and M. Lazdunski, Asic3 is a neuronal mechanosensor for pressure-induced vasodilation that protects against pressure ulcers, Nat. Med, vol.18, pp.1205-1207, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00731447

W. R. Frontera, H. Choi, G. Krishnan, L. S. Krivickas, S. Sabharwal et al., Single muscle fiber size and contractility after spinal cord injury in rats, Muscle Nerve, vol.34, pp.101-104, 2006.

R. G. Frykberg and J. Banks, Challenges in the Treatment of Chronic Wounds, Adv. Wound Care, vol.4, pp.560-582, 2015.

U. Fuoco, G. Scivoletto, A. Pace, V. U. Vona, and V. Castellano, Anaemia and serum protein alteration in patients with pressure ulcers, Spinal Cord, vol.35, pp.58-60, 1997.

M. D. Gaubert, L'effet du vieillissement sur la microcirculation cutanée. phdthesis, p.ép, 2008.

A. Gefen, Plantar soft tissue loading under the medial metatarsals in the standing diabetic foot, Med. Eng. Phys, vol.25, pp.491-499, 2003.

A. Gefen, The biomechanics of heel ulcers, J. Tissue Viability, vol.19, pp.124-131, 2010.

A. Gélis, A. Dupeyron, P. Legros, C. Benaïm, J. Pelissier et al., Pressure ulcer risk factors in persons with spinal cord injury Part 2: the chronic stage, Spinal Cord, vol.47, pp.651-661, 2009.

M. Gengenbacher, H. B. Stähelin, A. Scholer, and W. O. Sciler, Low biochemical nutritional parameters in acutely ill hospitalized elderly patients with and without stage III to IV pressure ulcers, Aging Clin. Exp. Res, vol.14, pp.420-423, 2002.

|. Bibliographie,

T. Gibson, R. M. Kenedi, and J. E. Craik, The mobile micro-architecture of dermal collagen: A bioengineering study, BJS, vol.52, pp.764-770, 1965.

B. M. Gillespie, W. P. Chaboyer, E. Mcinnes, B. Kent, J. A. Whitty et al., Repositioning for pressure ulcer prevention in adults, Cochrane Database Syst. Rev. CD009958, 2014.

H. Gissel, The role of Ca2+ in muscle cell damage, Ann. N. Y. Acad. Sci, vol.1066, pp.166-180, 2005.

L. Gould, P. Abadir, H. Brem, M. Carter, T. Conner-kerr et al., Chronic Wound Repair and Healing in Older Adults: Current Status and Future Research, Wound Repair Regen. Off. Publ. Wound Heal. Soc. Eur. Tissue Repair Soc, vol.23, pp.1-13, 2015.

R. J. Gray, D. Voegeli, and D. L. Bader, Features of lymphatic dysfunction in compressed skin tissues -Implications in pressure ulcer aetiology, J. Tissue Viability, vol.25, pp.26-31, 2016.

R. J. Gray, P. R. Worsley, D. Voegeli, and D. L. Bader, Monitoring contractile dermal lymphatic activity following uniaxial mechanical loading, Med. Eng. Phys, vol.38, pp.895-903, 2016.

P. C. Grisotto, A. C. Santos, J. Coutinho-netto, J. Cherri, and C. E. Piccinato, Indicators of oxidative injury and alterations of the cell membrane in the skeletal muscle of rats submitted to ischemia and reperfusion, J. Surg. Res, vol.92, pp.1-6, 2000.

A. Guerrero-hernandez and A. Verkhratsky, Calcium signalling in diabetes, Cell Calcium, vol.56, pp.297-301, 2014.

H. Guesdon, Devenir médical, familial et socioprofessionnel d'une cohorte de patients paraplégiques d'origine traumatique: étude rétrospective à plus de 4 ans de l'accident, 2010.

M. Guihan, M. Sohn, W. A. Bauman, A. M. Spungen, G. M. Powell-cope et al., Difficulty in Identifying Factors Responsible for Pressure Ulcer Healing in Veterans With Spinal Cord Injury, Arch. Phys. Med. Rehabil, vol.97, 2016.

L. Gunningberg, Risk, prevalence and prevention of pressure ulcers in three Swedish healthcare settings, J. Wound Care, vol.13, pp.286-290, 2004.

R. A. Gunther, C. , and M. , The effect of a dynamic pressure-redistributing bed support surface upon systemic lymph flow and composition, J. Tissue Viability, vol.10, pp.10-15, 2000.

S. Hagisawa and T. Shimada, Skin Morphology and Its Mechanical Properties Associated with Loading, Pressure Ulcer Research: Current and Future Perspectives, pp.161-185, 2005.

S. Hagisawa, M. Ferguson-pell, M. Cardi, and S. D. Miller, Assessment of skin blood content and oxygenation in spinal cord injured subjects during reactive hyperemia, J. Rehabil. Res. Dev, vol.31, pp.1-14, 1994.

S. Hagisawa, T. Shimada, H. Arao, and Y. Asada, Morphological architecture and distribution of blood capillaries and elastic fibres in the human skin, J. Tissue Viability, vol.11, pp.59-63, 2001.

P. W. Hamer, J. M. Mcgeachie, M. J. Davies, and M. D. Grounds, Evans Blue Dye as an in vivo marker of myofibre damage: optimising parameters for detecting initial myofibre membrane permeability, J. Anat, vol.200, pp.69-79, 2002.

N. Hatanaka, Y. Yamamoto, K. Ichihara, S. Mastuo, Y. Nakamura et al., A new predictive indicator for development of pressure ulcers in bedridden patients based on common laboratory tests results, J. Clin. Pathol, vol.61, pp.514-518, 2008.

B. Hemmes, L. A. De-wert, P. R. Brink, C. W. Oomens, D. L. Bader et al., Cytokine IL1? and lactate as markers for tissue damage in spineboard immobilisation. A prospective, randomised open-label crossover trial, J. Mech. Behav. Biomed. Mater, vol.75, pp.82-88, 2017.

R. Heyningen and J. S. Weiner, The effect of arterial occlusion on sweat composition, J. Physiol, vol.116, pp.404-413, 1952.

M. Hinzmann, S. Jaworski, M. Kutwin, J. Jagie??o, R. Kozi?ski et al., Nanoparticles containing allotropes of carbon have genotoxic effects on glioblastoma multiforme cells, Int. J. Nanomedicine, vol.9, pp.2409-2417, 2014.

G. H. Hirsch, M. R. Menard, A. , and H. A. , Anemia after traumatic spinal cord injury, Arch. Phys. Med. Rehabil, vol.72, pp.195-201, 1991.

H. M. Honda, P. Korge, and J. N. Weiss, Mitochondria and Ischemia/Reperfusion Injury, Ann. N. Y. Acad. Sci, vol.1047, pp.248-258, 2005.

I. Hoogendoorn, J. Reenalda, B. F. Koopman, and J. S. Rietman, The effect of pressure and shear on tissue viability of human skin in relation to the development of pressure ulcers: a systematic review, J. Tissue Viability, vol.26, pp.157-171, 2017.

T. Husain, An experimental study of some pressure effects on tissues, with reference to the bed-sore problem, J. Pathol. Bacteriol, vol.66, pp.347-358, 1953.

S. Hyun, X. Li, B. Vermillion, C. Newton, M. Fall et al., Body Mass Index and Pressure Ulcers: Improved Predictability of Pressure Ulcers in Intensive Care Patients, Am. J. Crit. Care Off. Publ. Am. Assoc. Crit.-Care Nurses, vol.23, pp.494-501, 2014.

K. Ikebe, T. Kato, M. Yamaga, J. Hirose, T. Tsuchida et al., Increased ischemia-reperfusion blood flow impairs the skeletal muscle contractile function, J. Surg. Res, vol.99, pp.1-6, 2001.

L. Jiang, Y. Dai, F. Cui, Y. Pan, H. Zhang et al., Expression of cytokines, growth factors and apoptosis-related signal molecules in chronic pressure ulcer wounds healing, Spinal Cord, vol.52, pp.145-151, 2014.

Q. Jiang, X. Li, X. Qu, Y. Liu, L. Zhang et al., The incidence, risk factors and characteristics of pressure ulcers in hospitalized patients in China, Int. J. Clin. Exp. Pathol, vol.7, pp.2587-2594, 2014.

Z. Kang and X. Zhai, The Association between Pre-existing Diabetes Mellitus and Pressure Ulcers in Patients Following Surgery: A Meta-analysis, Sci. Rep, vol.5, 2015.

Y. Kasuya, H. Umezawa, and M. Hatano, Stress-Activated Protein Kinases in Spinal Cord Injury: Focus on Roles of p38, Int. J. Mol. Sci, vol.19, p.867, 2018.

B. Kim, H. T. Kim, S. H. Park, J. Cha, T. Yufit et al., Fibroblasts from chronic wounds show altered TGF-beta-signaling and decreased TGF-beta Type II receptor expression, J. Cell. Physiol, vol.195, pp.331-336, 2003.

L. C. Kloth, J. E. Berman, S. Dumit-minkel, C. H. Sutton, P. E. Papanek et al., Effects of a normothermic dressing on pressure ulcer healing, Adv. Skin Wound Care, vol.13, pp.69-74, 2000.

S. L. Knight, R. P. Taylor, A. A. Polliack, and D. L. Bader, Establishing predictive indicators for the status of loaded soft tissues, J. Appl. Physiol, vol.90, pp.2231-2237, 2001.

|. Bibliographie,

A. Koïtka, P. Abraham, B. Bouhanick, D. Sigaudo-roussel, C. Demiot et al., Impaired pressure-induced vasodilation at the foot in young adults with type 1 diabetes, Diabetes, vol.53, pp.721-725, 2004.

S. Krishnan, P. E. Karg, M. L. Boninger, Y. Vodovotz, G. Constantine et al., Early Detection of Pressure Ulcer Development Following Traumatic Spinal Cord Injury Using Inflammatory Mediators, Arch. Phys. Med. Rehabil, vol.97, pp.1656-1662, 2016.

T. A. Krouskop, N. P. Reddy, W. A. Spencer, and J. W. Secor, Mechanisms of decubitus ulcer formation -An hypothesis, Med. Hypotheses, vol.4, pp.37-39, 1978.

E. A. Kruger, M. Pires, Y. Ngann, M. Sterling, and S. Rubayi, Comprehensive management of pressure ulcers in spinal cord injury: Current concepts and future trends, J. Spinal Cord Med, vol.36, pp.572-585, 2013.

A. Kurz, D. I. Sessler, and R. Lenhardt, Perioperative normothermia to reduce the incidence of surgicalwound infection and shorten hospitalization. Study of Wound Infection and Temperature Group, N. Engl. J. Med, vol.334, pp.1209-1215, 1996.

A. Kushnir, B. Wajsberg, and A. R. Marks, Ryanodine receptor dysfunction in human disorders, Biochim. Biophys. Acta, 2018.

C. Lachenbruch, Y. Tzen, D. Brienza, P. E. Karg, and P. A. Lachenbruch, Relative contributions of interface pressure, shear stress, and temperature on ischemic-induced, skin-reactive hyperemia in healthy volunteers: a repeated measures laboratory study, Ostomy. Wound Manage, vol.61, pp.16-25, 2015.

G. P. Ladwig, M. C. Robson, R. Liu, M. A. Kuhn, D. F. Muir et al., Wound Repair Regen, vol.10, pp.26-37, 2002.

N. A. Lahmann, R. J. Halfens, and T. Dassen, Pressure ulcers in German nursing homes and acute care hospitals: prevalence, frequency, and ulcer characteristics, Ostomy. Wound Manage, vol.52, pp.20-33, 2006.

Y. Lai, C. Lin, Y. Chang, M. Lin, S. Lee et al., Spinal cord injury increases the risk of Type 2 diabetes: a population-based cohort study, Spine J, vol.14, pp.1957-1964, 2014.

E. Landis, Micro-injection studies of capillary blood pressure in human skin, Heart, vol.15, pp.209-228, 1930.

J. T. Lanner, D. K. Georgiou, A. D. Joshi, and S. L. Hamilton, Ryanodine Receptors: Structure, Expression, Molecular Details, and Function in Calcium Release, Cold Spring Harb. Perspect. Biol, vol.2, 2010.

I. R. Lanza and K. S. Nair, Chapter 20: Functional Assessment of Isolated Mitochondria In Vitro, Methods in Enzymology, pp.349-372, 2009.

S. Latimer, W. Chaboyer, and B. M. Gillespie, The repositioning of hospitalized patients with reduced mobility: a prospective study, Nurs. Open, vol.2, pp.85-93, 2015.

K. M. Le, B. L. Madsen, P. W. Barth, G. A. Ksander, J. B. Angell et al., An in-depth look at pressure sores using monolithic silicon pressure sensors, Plast. Reconstr. Surg, vol.74, pp.745-756, 1984.

L. Gall, M. Matecki, S. Agdeppa, E. Lacampagne, A. Téot et al., Les supports de prévention et de traitement des escarres, Rev. Francoph. Cicatrisation, vol.1, pp.15-19, 2017.

L. V. Leak and J. F. Burke, ULTRASTRUCTURAL STUDIES ON THE LYMPHATIC ANCHORING FILAMENTS, J. Cell Biol, vol.36, pp.129-149, 1968.

B. Lee, S. Benyajati, J. A. Woods, J. , and Y. , Effect of local cooling on pro-inflammatory cytokines and blood flow of the skin under surface pressure in rats: feasibility study, J. Tissue Viability, vol.23, pp.69-77, 2014.

E. Leopold and A. Gefen, A simple stochastic model to explain the sigmoid nature of the strain-time cellular tolerance curve, J. Tissue Viability, vol.21, pp.27-36, 2012.

J. M. Levine, Historical notes on pressure ulcers: the cure of Ambrose Paré, Decubitus, vol.5, pp.23-24, 1992.

A. Levy, K. Kopplin, and A. Gefen, Simulations of skin and subcutaneous tissue loading in the buttocks while regaining weight-bearing after a push-up in wheelchair users, J. Mech. Behav. Biomed. Mater, vol.28, pp.436-447, 2013.

Y. Li, Y. Liu, Y. Fu, T. Wei, L. Le-guyader et al., The triggering of apoptosis in macrophages by pristine graphene through the MAPK and TGF-beta signaling pathways, Biomaterials, vol.33, pp.402-411, 2012.

E. Linder-ganz, S. Engelberg, M. Scheinowitz, and A. Gefen, Pressure-time cell death threshold for albino rat skeletal muscles as related to pressure sore biomechanics, J. Biomech, vol.39, pp.2725-2732, 2006.

E. Linder-ganz, N. Shabshin, and A. Gefen, Patient-specific modeling of deep tissue injury biomechanics in an unconscious patient who developed myonecrosis after prolonged lying, J. Tissue Viability, vol.18, pp.62-71

X. Liu, L. Harlow, Z. A. Graham, W. A. Bauman, and C. Cardozo, Spinal Cord Injury Leads to Hyperoxidation and Nitrosylation of Skeletal Muscle Ryanodine Receptor-1 Associated with Upregulation of Nicotinamide Adenine Dinucleotide Phosphate Oxidase 4, J. Neurotrauma, vol.34, pp.2069-2074, 2017.

S. Loerakker, Aetiology of pressure ulcers, p.31, 2007.

S. Loerakker, E. S. Huisman, H. A. Seelen, J. F. Glatz, F. P. Baaijens et al., Plasma variations of biomarkers for muscle damage in male nondisabled and spinal cord injured subjects, J. Rehabil. Res. Dev, vol.49, pp.361-372, 2012.

B. Lu, T. Li, H. Zhao, X. Li, C. Gao et al., Graphene-based composite materials beneficial to wound healing, Nanoscale, vol.4, pp.2978-2982, 2012.

B. R. Macintosh, R. J. Holash, R. , and J. , Skeletal muscle fatigue -regulation of excitationcontraction coupling to avoid metabolic catastrophe, J Cell Sci, vol.125, pp.2105-2114, 2012.

A. Macron, H. Pillet, J. Doridam, A. Verney, and P. Rohan, Development and evaluation of a new methodology for the fast generation of patient-specific Finite Element models of the buttock for sitting-acquired deep tissue injury prevention, J. Biomech, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02305719

K. Mahadev, X. Wu, H. Motoshima, and B. J. Goldstein, Integration of multiple downstream signals determines the net effect of insulin on MAP kinase vs. PI 3?-kinase activation: potential role of insulin-stimulated H2O2, Cell. Signal, vol.16, pp.323-331, 2004.

K. Mahadev, H. Motoshima, X. Wu, J. M. Ruddy, R. S. Arnold et al., The NAD(P)H Oxidase Homolog Nox4 Modulates Insulin-Stimulated Generation of H2O2 and Plays an Integral Role in Insulin Signal Transduction, Mol. Cell. Biol, vol.24, pp.1844-1854, 2004.

M. Makhsous, F. Lin, A. Pandya, M. S. Pandya, and C. C. Chadwick, Elevation in the serum and urine concentration of injury-related molecules after the formation of deep tissue injury in a rat spinal cord injury pressure ulcer model, PM R, vol.2, pp.1063-1065, 2010.

A. R. Marks, D. W. Landry, and S. Deng, Agents for preventing and treating disorders involving modulation of the ryanodine receptors, 2014.

|. Bibliographie,

M. V. Marshall, J. C. Rasmussen, I. Tan, M. B. Aldrich, K. E. Adams et al., Near-Infrared Fluorescence Imaging in Humans with Indocyanine Green: A Review and Update, Open Surg. Oncol. J. Online, vol.2, pp.12-25, 2010.

S. Matecki, H. Dridi, B. Jung, N. Saint, S. R. Reiken et al., Leaky ryanodine receptors contribute to diaphragmatic weakness during mechanical ventilation, Proc. Natl. Acad. Sci. U. S. A, vol.113, pp.9069-9074, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01800610

W. Mcguiness, E. Vella, H. , and D. , Influence of dressing changes on wound temperature, J. Wound Care, vol.13, pp.383-385, 2004.

E. Mcinnes, A. Jammali-blasi, S. E. Bell-syer, J. C. Dumville, V. Middleton et al., Support surfaces for pressure ulcer prevention, The Cochrane Library, 2015.

E. Meesterberends, R. Halfens, . Dr, C. Lohrmann, and R. De-wit, Pressure ulcer guideline development and dissemination in Europe, J. Clin. Nurs, vol.19, pp.1495-1503, 2010.

G. E. Miller and J. Seale, Lymphatic clearance during compressive loading, Lymphology, vol.14, pp.161-166, 1981.

T. Minematsu, G. Nakagami, Y. Sari, T. Akase, J. Sugama et al., Candidate biomarkers for deep tissue damage from molecular biological and biochemical aspects, J. Tissue Viability, vol.19, pp.77-83, 2010.

Z. Moore, E. Johansen, M. Etten, . Van, H. Strapp et al., Pressure ulcer prevalence and prevention practices: a cross-sectional comparative survey in Norway and Ireland, J. Wound Care, vol.24, pp.333-339, 2015.

F. Morfoisse, Régulations moléculaires des facteurs lymphangiogéniques dans les pathologies vasculaires, vol.3, 2015.

A. Mufti, P. Coutts, and R. G. Sibbald, Validation of commercially available infrared thermometers for measuring skin surface temperature associated with deep and surrounding wound infection, Adv. Skin Wound Care, vol.28, pp.11-16, 2015.

E. Murphy and C. Steenbergen, Mechanisms Underlying Acute Protection From Cardiac Ischemia-Reperfusion Injury, Physiol. Rev, vol.88, pp.581-609, 2008.

G. A. Nader and K. A. Esser, Intracellular signaling specificity in skeletal muscle in response to different modes of exercise, J. Appl. Physiol. Bethesda Md, vol.90, pp.1936-1942, 1985.

G. Nakagami, H. Sanada, S. Iizaka, T. Kadono, T. Higashino et al., Predicting delayed pressure ulcer healing using thermography: a prospective cohort study, J. Wound Care, vol.19, pp.465-472, 2010.

M. Nassaji, Z. Askari, and R. Ghorbani, Cigarette smoking and risk of pressure ulcer in adult intensive care unit patients, Int. J. Nurs. Pract, vol.20, pp.418-423, 2014.

M. Nguyen-tu, A. Begey, J. Decorps, J. Boizot, P. Sommer et al., Skin microvascular response to pressure load in obese mice, Microvasc. Res, vol.90, pp.138-143, 2013.

J. C. Oliver, L. A. Bland, C. W. Oettinger, M. J. Arduino, S. K. Mcallister et al., , 1993.

, Cytokine kinetics in an in vitro whole blood model following an endotoxin challenge, Lymphokine Cytokine Res, vol.12, pp.115-120

C. Oomens, Perspectives of Numerical Modelling in Pressure Ulcer Research, Pressure Ulcer Research: Current and Future Perspectives, pp.149-159, 2005.

C. Oomens, M. Brekelmans, S. Loerakker, and F. Baaijens, Biomechanics: Concepts and Computation, 2018.

C. W. Oomens, M. Maenhout, C. H. Van-oijen, M. R. Drost, and F. P. Baaijens, Finite element modelling of contracting skeletal muscle, Philos. Trans. R. Soc. Lond. B. Biol. Sci, vol.358, pp.1453-1460, 2003.

S. M. Peirce, T. C. Skalak, and G. T. Rodeheaver, Ischemia-reperfusion injury in chronic pressure ulcer formation: a skin model in the rat, Wound Repair Regen. Off. Publ. Wound Heal. Soc. Eur. Tissue Repair Soc, vol.8, pp.68-76, 2000.

A. Perkash and M. Brown, Anemia in patients with traumatic spinal cord injury, J. Am. Paraplegia Soc, vol.9, pp.10-15, 1986.

M. J. Peterson, N. Gravenstein, W. K. Schwab, J. H. Van-oostrom, and L. J. Caruso, Patient repositioning and pressure ulcer risk--monitoring interface pressures of at-risk patients, J. Rehabil. Res. Dev, vol.50, pp.477-488, 2013.

M. A. Petrie, M. Suneja, E. Faidley, and R. K. Shields, Low force contractions induce fatigue consistent with muscle mRNA expression in people with spinal cord injury, Physiol. Rep, vol.2, p.248, 2014.

H. M. Piper, K. Meuter, and C. Schäfer, Cellular mechanisms of ischemia-reperfusion injury, Ann. Thorac. Surg, vol.75, pp.644-648, 2003.

H. M. Piper, Y. Abdallah, and C. Schäfer, The first minutes of reperfusion: a window of opportunity for cardioprotection, Cardiovasc. Res, vol.61, pp.365-371, 2004.

A. Polliack, R. Taylor, and D. Bader, Analysis of sweat during soft tissue breakdown following pressure ischaemia, J. Rehabil. Res. Dev, vol.30, pp.250-259, 1993.

A. Polliack, R. Taylor, and D. Bader, Sweat analysis following pressure ischaemia in a group of debilitated subjects, J. Rehabil. Res. Dev, vol.34, pp.303-308, 1997.

L. A. Pompeii, H. J. Lipscomb, A. L. Schoenfisch, and J. M. Dement, Musculoskeletal injuries resulting from patient handling tasks among hospital workers, Am. J. Ind. Med, vol.52, pp.571-578, 2009.

J. Pottecher, Muscle squelettique et ischémie-reperfusion expérimentale des membres : mécanismes impliqués dans la protection ou les effets délétères de la cyclosporine et facteurs limitant les conditionnements pharmacologique et ischémique, 2012.

R. T. Rebbeck, Y. Karunasekara, P. G. Board, N. A. Beard, M. G. Casarotto et al., Skeletal muscle excitation-contraction coupling: who are the dancing partners?, Int. J. Biochem. Cell Biol, vol.48, pp.28-38, 2014.

N. P. Reddy, Effects of Mechanical Stresses on Lymph and Interstitial Fluid Flows, Pressure Sores -Clinical Practice and Scientific Approach, pp.203-220, 1990.

N. P. Reddy and G. V. Cochran, Interstitial fluid flow as a factor in decubitus ulcer formation, J. Biomech, vol.14, pp.879-881, 1981.

N. P. Reddy, P. , and K. , A mathematical model of flow through the terminal lymphatics, Med. Eng. Phys, vol.17, pp.134-140, 1995.

T. Reffelmann and R. A. Kloner, The "no-reflow" phenomenon: basic science and clinical correlates, Heart, vol.87, pp.162-168, 2002.

S. Reiken, A. Lacampagne, H. Zhou, A. Kherani, S. E. Lehnart et al., PKA phosphorylation activates the calcium release channel (ryanodine receptor) in skeletal muscle, J. Cell Biol, vol.160, pp.919-928, 2003.

E. F. |-bibliographie-ring and K. Ammer, Infrared thermal imaging in medicine, Physiol. Meas, vol.33, pp.33-46, 2012.

G. P. Rodriguez, C. , and J. , Biochemical changes in skin composition in spinal cord injury: a possible contribution to decubitus ulcers, Paraplegia, vol.26, pp.302-309, 1988.

G. P. Rodriguez, J. Claus-walker, M. C. Kent, and H. M. Garza, Collagen metabolite excretion as a predictor of bone-and skin-related complications in spinal cord injury, Arch. Phys. Med. Rehabil, vol.70, pp.442-444, 1989.

R. Rubin, D. S. Strayer, E. Rubin, (. M. Mcdonald, and J. M. , Rubin's Pathology: Clinicopathologic Foundations of Medicine, 2008.

R. Salcido, A. Popescu, A. , and C. , Animal models in pressure ulcer research, J. Spinal Cord Med, vol.30, pp.107-116, 2007.

J. E. Sanders and B. S. Goldstein, Collagen fibril diameters increase and fibril densities decrease in skin subjected to repetitive compressive and shear stresses, J. Biomech, vol.34, pp.1581-1587, 2001.

R. Sartori, G. Milan, M. Patron, C. Mammucari, B. Blaauw et al., Smad2 and 3 transcription factors control muscle mass in adulthood, Am. J. Physiol. Cell Physiol, vol.296, pp.1248-1257, 2009.

A. Sasidharan, L. S. Panchakarla, A. R. Sadanandan, A. Ashokan, P. Chandran et al., Hemocompatibility and macrophage response of pristine and functionalized graphene, Small Weinh. Bergstr. Ger, vol.8, pp.1251-1263, 2012.

J. L. Saumet, , 2005.

, Bull. Acad. Natl. Med, vol.189, pp.105-106

R. Scelsi, Skeletal muscle pathology after spinal cord injury: Our 20 year experience and results on skeletal muscle changes in paraplegics, related to functional rehabilitation, Basic Appl Myol, vol.11, pp.75-85, 2001.

R. Scelsi, L. Scelsi, R. Bocchi, L. , and S. , Morphological changes in the skin microlymphatics in recently injured paraplegic patients with ilio-femoral venous thrombosis, Paraplegia, vol.33, pp.472-475, 1995.

L. Schoonhoven, J. R. Haalboom, M. T. Bousema, A. Algra, D. E. Grobbee et al., Prospective cohort study of routine use of risk assessment scales for prediction of pressure ulcers, BMJ, vol.325, p.797, 2002.

M. Seo, M. Enomoto, N. Ishiyama, P. B. Stathopulos, and M. Ikura, Structural insights into endoplasmic reticulum stored calcium regulation by inositol 1,4,5-trisphosphate and ryanodine receptors, Biochim. Biophys. Acta BBA -Mol. Cell Res, vol.1853, 1980.

V. Serantoni, F. Jourdan, H. Louche, and A. Sultan, Thermomechanical analysis of the surface vascular system -Application to the diabetic foot, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01816018

R. Serra, S. Caroleo, G. Buffone, M. Lugarà, V. Molinari et al., , 2014.

, Low serum albumin level as an independent risk factor for the onset of pressure ulcers in intensive care unit patients, Int. Wound J, vol.11, pp.550-553

N. Shabshin, G. Zoizner, A. Herman, V. Ougortsin, and A. Gefen, Use of weight-bearing MRI for evaluating wheelchair cushions based on internal soft-tissue deformations under ischial tuberosities, J. Rehabil. Res. Dev, vol.47, pp.31-42, 2010.

S. Shahwan, Management and Prevention of Diabetic Pressure Ulcers with Neuropathy, Gen. Med. Open Access, vol.3, pp.1-3, 2015.

V. Shats and S. Kozacov, , 1996.

, Harefuah, vol.131, p.203

S. Shekarforoush, Z. Fatahi, and F. Safari, The effects of pentobarbital, ketamine-pentobarbital and ketamine-xylazine anesthesia in a rat myocardial ischemic reperfusion injury model, Lab. Anim, vol.50, pp.179-184, 2016.

C. Shi, J. C. Dumville, and N. Cullum, Support surfaces for pressure ulcer prevention: A network metaanalysis, PloS One, vol.13, p.192707, 2018.

H. Shi, H. Xie, Y. Zhao, C. Lin, F. Cui et al., Myoprotective effects of bFGF on skeletal muscle injury in pressure-related deep tissue injury in rats, Burns Trauma, vol.4, p.26, 2016.

N. Shoham and A. Gefen, Deformations, mechanical strains and stresses across the different hierarchical scales in weight-bearing soft tissues, J. Tissue Viability, vol.21, pp.39-46, 2012.

N. A. Silva, N. Sousa, R. L. Reis, and A. J. Salgado, From basics to clinical: A comprehensive review on spinal cord injury, Prog. Neurobiol, vol.114, pp.25-57, 2014.

T. K. Sin, X. M. Pei, B. T. Teng, E. W. Tam, B. Y. Yung et al., Oxidative stress and DNA damage signalling in skeletal muscle in pressure-induced deep tissue injury, Pflugers Arch, vol.465, pp.295-317, 2013.

M. D. Siperstein, P. Raskin, and H. Burns, Electron microscopic quantification of diabetic microangiopathy, Diabetes, vol.22, pp.514-527, 1973.

P. M. Siu, B. T. Teng, X. M. Pei, and E. W. Tam, Proteasome inhibition alleviates prolonged moderate compression-induced muscle pathology, BMC Musculoskelet. Disord, vol.12, p.58, 2011.

M. Skoda, I. Dudek, and D. Szukiewicz, Potential and Challenges of Graphene in Medicine, Graphene-Based Materials in Health and Environment: New Paradigms, G. Gonçalves, P. Marques, and M. Vila, pp.3-33, 2016.

R. C. Sloan, M. Rosenbaum, D. O'rourke, K. Oppelt, C. R. Frasier et al., High doses of ketamine-xylazine anesthesia reduce cardiac ischemia-reperfusion injury in guinea pigs, J. Am. Assoc. Lab. Anim. Sci. JAALAS, vol.50, pp.349-354, 2011.

G. C. Slowikowski and M. Funk, Factors associated with pressure ulcers in patients in a surgical intensive care unit, J. Wound Ostomy Cont. Nurs. Off. Publ. Wound Ostomy Cont. Nurses Soc, vol.37, pp.619-626, 2010.

M. J. Smit, H. Beekhuis, A. M. Duursma, J. M. Bouma, and M. Gruber, Catabolism of circulating enzymes: plasma clearance, endocytosis, and breakdown of lactate dehydrogenase-1 in rabbits, Clin. Chem, vol.34, pp.2475-2480, 1988.

A. Smith, G. Hayes, A. Romaschin, and P. Walker, The role of extracellular calcium in ischemia/reperfusion injury in skeletal muscle, J. Surg. Res, vol.49, pp.153-156, 1990.

I. L. Smith, J. Nixon, S. Brown, L. Wilson, C. et al., Pressure ulcer and wounds reporting in NHS hospitals in England part 1: Audit of monitoring systems, J. Tissue Viability, vol.25, pp.3-15, 2016.

J. S. Smith, T. Imagawa, J. Ma, M. Fill, K. P. Campbell et al., Purified ryanodine receptor from rabbit skeletal muscle is the calcium-release channel of sarcoplasmic reticulum, J. Gen. Physiol, vol.92, pp.1-26, 1988.

Y. Son, Y. Cheong, N. Kim, H. Chung, D. G. Kang et al., Mitogen-Activated Protein Kinases and Reactive Oxygen Species: How Can ROS Activate MAPK Pathways?, 2011.

Y. Son, S. Kim, H. Chung, and H. Pae, Reactive oxygen species in the activation of MAP kinases, Methods Enzymol, vol.528, pp.27-48, 2013.

R. Sopher, J. Nixon, C. Gorecki, and A. Gefen, Exposure to internal muscle tissue loads under the ischial tuberosities during sitting is elevated at abnormally high or low body mass indices, J. Biomech, vol.43, pp.280-286, 2010.

P. Srikanthan, A. L. Hevener, and A. S. Karlamangla, Sarcopenia exacerbates obesity-associated insulin resistance and dysglycemia: findings from the National Health and Nutrition Examination Survey III, PloS One, vol.5, p.10805, 2010.

A. Stekelenburg, C. Oomens, and D. Bader, Compression-Induced Tissue Damage: Animal Models, Pressure Ulcer Research, pp.187-204, 2005.

A. Stekelenburg, G. J. Strijkers, H. Parusel, D. L. Bader, K. Nicolay et al., Role of ischemia and deformation in the onset of compression-induced deep tissue injury: MRI-based studies in a rat model, J. Appl. Physiol. Bethesda Md, vol.102, 1985.

O. Stojadinovic, J. Minkiewicz, A. Sawaya, J. W. Bourne, P. Torzilli et al., Deep tissue injury in development of pressure ulcers: a decrease of inflammasome activation and changes in human skin morphology in response to aging and mechanical load, PloS One, vol.8, p.69223, 2013.

L. Suleman, P. , and S. L. , Biofilm-Infected Pressure Ulcers: Current Knowledge and Emerging Treatment Strategies, Biofilm-Based Healthcare-Associated Infections, vol.II, pp.29-43, 2015.

I. Swain, The Measurement of Interface Pressure, pp.51-71, 2005.

I. D. Swain and D. L. Bader, The measurement of interface pressure and its role in soft tissue breakdown, J. Tissue Viability, vol.12, pp.140-146, 2002.

M. Talan, Body temperature of C57BL/6J mice with age, Exp. Gerontol, vol.19, pp.25-29, 1984.

B. T. Teng, X. M. Pei, E. W. Tam, I. F. Benzie, and P. M. Siu, Opposing responses of apoptosis and autophagy to moderate compression in skeletal muscle, Acta Physiol. Oxf. Engl, vol.201, pp.239-254, 2011.

B. T. Teng, E. W. Tam, I. F. Benzie, and P. M. Siu, Protective effect of caspase inhibition on compressioninduced muscle damage, J. Physiol, vol.589, pp.3349-3369, 2011.

C. Vangilder, G. Macfarlane, S. Meyer, and C. Lachenbruch, Body Mass Index, Weight, and Pressure Ulcer Prevalence: An Analysis of the 2006-2007 International Pressure Ulcer Prevalence TM Surveys, J. Nurs. Care Qual, vol.24, p.127, 2009.

C. Vangilder, C. Lachenbruch, C. Algrim-boyle, and S. Meyer, The International Pressure Ulcer Prevalence TM Survey: 2006-2015: A 10-Year Pressure Injury Prevalence and Demographic Trend Analysis by Care Setting, J. Wound Ostomy Cont. Nurs. Off. Publ. Wound Ostomy Cont. Nurses Soc, vol.44, pp.20-28, 2017.

F. Veliev, Z. Han, D. Kalita, A. Briançon-marjollet, V. Bouchiat et al., Recording Spikes Activity in Cultured Hippocampal Neurons Using Flexible or Transparent Graphene Transistors, Front. Neurosci, vol.11, p.466, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01643900

G. A. Vos, J. J. Congleton, J. Steven-moore, A. A. Amendola, and L. Ringer, Postural versus chair design impacts upon interface pressure, Appl. Ergon, vol.37, pp.619-628, 2006.

Y. Wang and J. Sanders, Skin Model Studies, Pressure Ulcer Research: Current and Future Perspectives, pp.263-285, 2005.

Y. Wang and J. E. Sanders, How does skin adapt to repetitive mechanical stress to become load tolerant?, Med. Hypotheses, vol.61, pp.29-35, 2003.

X. Wang, J. Ge, E. E. Tredget, and Y. Wu, The mouse excisional wound splinting model, including applications for stem cell transplantation, Nat. Protoc, vol.8, pp.302-309, 2013.

D. L. Waning, K. S. Mohammad, S. Reiken, W. Xie, D. C. Andersson et al., Excess TGF-? mediates muscle weakness associated with bone metastases in mice, Nat. Med, vol.21, pp.1262-1271, 2015.

X. H. Wehrens, S. E. Lehnart, S. R. Reiken, and A. R. Marks, Ca2+/calmodulin-dependent protein kinase II phosphorylation regulates the cardiac ryanodine receptor, Circ. Res, vol.94, pp.61-70, 2004.

X. H. Wehrens, S. E. Lehnart, S. Reiken, R. Van-der-nagel, R. Morales et al., Enhancing calstabin binding to ryanodine receptors improves cardiac and skeletal muscle function in heart failure, Proc. Natl. Acad. Sci. U. S. A, vol.102, pp.9607-9612, 2005.

C. Weiner, D. Alperovitch-najenson, J. Ribak, and L. Kalichman, Prevention of Nurses' Work-Related Musculoskeletal Disorders Resulting From Repositioning Patients in Bed: Comprehensive Narrative Review. Workplace Health Saf, vol.63, pp.226-232, 2015.

J. Wenk, A. Foitzik, V. Achterberg, A. Sabiwalsky, J. Dissemond et al., Selective pick-up of increased iron by deferoxamine-coupled cellulose abrogates the iron-driven induction of matrix-degrading metalloproteinase 1 and lipid peroxidation in human dermal fibroblasts in vitro: a new dressing concept, J. Invest. Dermatol, vol.116, pp.833-839, 2001.

L. A. Wert, . De, D. L. Bader, C. W. Oomens, L. Schoonhoven et al., A new method to evaluate the effects of shear on the skin, Wound Repair Regen, vol.23, pp.885-890, 2015.

M. J. Westby, J. C. Dumville, M. O. Soares, N. Stubbs, N. et al., Dressings and topical agents for treating pressure ulcers, Cochrane Database Syst. Rev, vol.6, p.11947, 2017.

U. Widegren, C. Wretman, A. Lionikas, G. Hedin, and J. Henriksson, Influence of exercise intensity on ERK/MAP kinase signalling in human skeletal muscle, Pflugers Arch, vol.441, pp.317-322, 2000.

A. J. Williams, N. L. Thomas, G. , and C. H. , The ryanodine receptor: advances in structure and organization, Curr. Opin. Physiol, vol.1, pp.1-6, 2018.

J. W. Witherspoon and K. G. Meilleur, Review of RyR1 pathway and associated pathomechanisms, Acta Neuropathol. Commun, vol.4, 2016.

J. A. Witkowski and L. C. Parish, Histopathology of the decubitus ulcer, J. Am. Acad. Dermatol, vol.6, pp.1014-1021, 1982.

S. Wu, A. J. Applewhite, J. Niezgoda, R. Snyder, J. Shah et al., Oxidized Regenerated Cellulose/Collagen Dressings: Review of Evidence and Recommendations, Adv. Skin Wound Care, vol.30, pp.1-18, 2017.

Y. Wu, D. W. Van-der-schaft, F. P. Baaijens, and C. W. Oomens, Cell death induced by mechanical compression on engineered muscle results from a gradual physiological mechanism, J. Biomech, vol.49, pp.1071-1077, 2016.

T. Yamamoto, N. Yamamoto, S. Azuma, T. Todokoro, and I. Koshima, Handy thermography for bedside evaluation of pressure ulcer, J. Plast. Reconstr. Aesthetic Surg. JPRAS, vol.66, pp.205-206, 2013.

J. Yang, X. Wang, B. Shi, and F. Huang, The angiogenic peptide vascular endothelial growth factor-basic fibroblast growth factor signaling is up-regulated in a rat pressure ulcer model, Anat. Rec. Hoboken NJ, vol.296, pp.1161-1168, 2007.

|. Bibliographie,

Z. Yuchi, S. M. Yuen, K. Lau, A. Q. Underhill, R. L. Cornea et al., Crystal structures of ryanodine receptor SPRY1 and tandem-repeat domains reveal a critical FKBP12 binding determinant, Nat. Commun, vol.6, p.7947, 2015.

R. Zalk, O. B. Clarke, A. Georges, R. A. Grassucci, S. Reiken et al., Structure of a mammalian ryanodine receptor, Nature, vol.517, pp.44-49, 2015.

Y. Zhang, L. Zhou, and C. A. Miller, A splicing variant of a death domain protein that is regulated by a mitogen-activated kinase is a substrate for c-Jun N-terminal kinase in the human central nervous system, Proc. Natl. Acad. Sci. U. S. A, vol.95, pp.2586-2591, 1998.

X. Zhi, H. Fang, C. Bao, G. Shen, J. Zhang et al., The immunotoxicity of graphene oxides and the effect of PVP-coating, Biomaterials, vol.34, pp.5254-5261, 2013.

D. B. Zorov, M. Juhaszova, and S. J. Sollott, Mitochondrial ROS-induced ROS release: an update and review, Biochim. Biophys. Acta, vol.1757, pp.509-517, 2006.