51
|
Allan AN, Zitnay JL, Maas SA, Weiss JA. Development of a continuum damage model to Predict accumulation of sub-failure damage in tendons. J Mech Behav Biomed Mater 2022; 135:105342. [DOI: 10.1016/j.jmbbm.2022.105342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 05/18/2022] [Accepted: 06/26/2022] [Indexed: 11/17/2022]
|
52
|
Ferdousy RN, Kadokawa H. Specific locations and amounts of denatured collagen and collagen-specific chaperone HSP47 in the oviducts and uteri of old cows as compared with those of heifers. Reprod Fertil Dev 2022; 34:619-632. [PMID: 35296375 DOI: 10.1071/rd21130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 01/18/2022] [Indexed: 12/22/2022] Open
Abstract
Collagen, the most abundant extra-cellular matrix in oviducts and uteri, performs critical roles in pregnancies. We hypothesised that the locations and amounts of both denatured collagen and the collagen-specific molecular chaperone 47-kDa heat shock protein (HSP47) in the oviducts and uteri of old cows are different compared with those of young heifers because of repeated pregnancies. Since detecting damaged collagen in tissues is challenging, we developed a new method that uses a denatured collagen detection reagent. Then, we compared damaged collagen in the oviducts and uteri between post-pubertal growing nulliparous heifers (22.1±1.0months old) and old multiparous cows (143.1±15.6months old). Further, we evaluated the relationship between denatured collagen and HSP47 by combining this method with fluorescence immunohistochemistry. Picro-sirius red staining showed collagen in almost all parts of the oviducts and uteri. Expectedly, damaged collagen was increased in the oviducts and uteri of old cows. However, damaged collagen and HSP47 were not located in the same area in old cows. The number of fibroblasts increased, suggesting the presence of fibrosis in the oviducts and uteri of old cows. These organs of old cows showed higher HSP47 protein amounts than those of heifers. However, the uteri, but not oviducts, of old cows had lower HSP47 mRNA amounts than those of heifers. These findings revealed the specific location and amounts of denatured collagen and HSP47 in the oviducts and uteri of old cows compared with those of heifers.
Collapse
Affiliation(s)
- Raihana Nasrin Ferdousy
- Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi-shi, Yamaguchi-ken 1677-1, Japan
| | - Hiroya Kadokawa
- Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi-shi, Yamaguchi-ken 1677-1, Japan
| |
Collapse
|
53
|
Yang DS, Dickerson EE, Zhang LX, Richendrfer H, Karamchedu PN, Badger GJ, Schmidt TA, Fredericks AM, Elsaid KA, Jay GD. Quadruped Gait and Regulation of Apoptotic Factors in Tibiofemoral Joints following Intra-Articular rhPRG4 Injection in Prg4 Null Mice. Int J Mol Sci 2022; 23:ijms23084245. [PMID: 35457064 PMCID: PMC9025840 DOI: 10.3390/ijms23084245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 04/06/2022] [Indexed: 12/03/2022] Open
Abstract
Camptodactyly-arthropathy-coxa vara-pericarditis (CACP) syndrome leads to diarthrodial joint arthropathy and is caused by the absence of lubricin (proteoglycan 4—PRG4), a surface-active mucinous glycoprotein responsible for lubricating articular cartilage. In this study, mice lacking the orthologous gene Prg4 served as a model that recapitulates the destructive arthrosis that involves biofouling of cartilage by serum proteins in lieu of Prg4. This study hypothesized that Prg4-deficient mice would demonstrate a quadruped gait change and decreased markers of mitochondrial dyscrasia, following intra-articular injection of both hindlimbs with recombinant human PRG4 (rhPRG4). Prg4−/− (N = 44) mice of both sexes were injected with rhPRG4 and gait alterations were studied at post-injection day 3 and 6, before joints were harvested for immunohistochemistry for caspase-3 activation. Increased stance and propulsion was shown at 3 days post-injection in male mice. There were significantly fewer caspase-3-positive chondrocytes in tibiofemoral cartilage from rhPRG4-injected mice. The mitochondrial gene Mt-tn, and myosin heavy (Myh7) and light chains (Myl2 and Myl3), known to play a cytoskeletal stabilizing role, were significantly upregulated in both sexes (RNA-Seq) following IA rhPRG4. Chondrocyte mitochondrial dyscrasias attributable to the arthrosis in CACP may be mitigated by IA rhPRG4. In a supporting in vitro crystal microbalance experiment, molecular fouling by albumin did not block the surface activity of rhPRG4.
Collapse
Affiliation(s)
- Daniel S. Yang
- School of Engineering, Brown University, Providence, RI 02912, USA; (D.S.Y.); (G.D.J.)
- Department of Emergency Medicine, Alpert School of Medicine, Brown University, Providence, RI 02903, USA; (L.X.Z.); (H.R.)
| | - Edward E. Dickerson
- North Carolina Agricultural Technical State University, Greensboro, NC 27411, USA;
| | - Ling X. Zhang
- Department of Emergency Medicine, Alpert School of Medicine, Brown University, Providence, RI 02903, USA; (L.X.Z.); (H.R.)
| | - Holly Richendrfer
- Department of Emergency Medicine, Alpert School of Medicine, Brown University, Providence, RI 02903, USA; (L.X.Z.); (H.R.)
| | - Padmini N. Karamchedu
- Department of Orthopedics, Alpert School of Medicine, Brown University, Providence, RI 02903, USA;
| | - Gary J. Badger
- Department of Medical Biostatistics, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA;
| | - Tannin A. Schmidt
- Department of Biomedical Engineering, School of Dental Medicine, University of Connecticut Health, Farmington, CT 06030, USA;
| | - Alger M. Fredericks
- Department of Surgery, Alpert School of Medicine, Brown University, Providence, RI 02903, USA;
| | - Khaled A. Elsaid
- School of Pharmacy, Chapman University, Irvine, CA 92618, USA
- Correspondence:
| | - Gregory D. Jay
- School of Engineering, Brown University, Providence, RI 02912, USA; (D.S.Y.); (G.D.J.)
- Department of Emergency Medicine, Alpert School of Medicine, Brown University, Providence, RI 02903, USA; (L.X.Z.); (H.R.)
- Department of Orthopedics, Alpert School of Medicine, Brown University, Providence, RI 02903, USA;
| |
Collapse
|
54
|
Empirical evidence that bone collagen molecules denature as a result of bone fracture. J Mech Behav Biomed Mater 2022; 131:105220. [DOI: 10.1016/j.jmbbm.2022.105220] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/16/2022] [Accepted: 04/02/2022] [Indexed: 12/11/2022]
|
55
|
Huang R, Yu D, Savage D, Wozniak K, Zheleznyak L, Knox WH, Huxlin KR. Blue-LIRIC in the rabbit cornea: efficacy, tissue effects, and repetition rate scaling. BIOMEDICAL OPTICS EXPRESS 2022; 13:2346-2363. [PMID: 35519279 PMCID: PMC9045900 DOI: 10.1364/boe.448286] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/20/2022] [Accepted: 03/10/2022] [Indexed: 06/14/2023]
Abstract
Laser-induced refractive index change (LIRIC) is being developed as a non-invasive way to alter optical properties of transparent, ophthalmic materials including corneas ex vivo and in vivo. This study examined the optical and biological effects of blue-LIRIC (wavelengths 400-405 nm) of ex-vivo rabbit corneas. Following LIRIC treatment at low and high repetition rates (8.3 MHz and 80 MHz, respectively), we interferometrically measured optical phase change, obtained transmission electron microscopy (TEM) micrographs, and stained histological sections with collagen hybridizing peptides (CHP) to assess the structural and organizational changes caused by LIRIC at different repetition rates. Finally, we performed power and scan speed scaling experiments at three different repetition rates (1 MHz, 8.3 MHz, and 80 MHz) to study their impact on LIRIC efficacy. Histologic co-localization of CHP and LIRIC-generated green autofluorescence signals suggested that collagen denaturation had occurred in the laser-irradiated region. TEM imaging showed different ultrastructural modifications for low and high repetition rate writing, with discrete homogenization of collagen fibrils at 80 MHz, as opposed to contiguous homogenization at 8.3 MHz. Overall, this study confirmed that LIRIC efficacy can be dramatically increased, while still avoiding tissue ablation, by lowering the repetition rate from 80 MHz to 8.3 MHz. Modeling suggests that this is due to a higher, single-pulse, energy density deposition at given laser powers during 8.3 MHz LIRIC.
Collapse
Affiliation(s)
- Ruiting Huang
- The Institute of Optics, University of Rochester, Rochester, NY 14627, USA
| | - Dan Yu
- Materials Science Program, University of Rochester, Rochester, NY 14627, USA
| | - Daniel Savage
- The Institute of Optics, University of Rochester, Rochester, NY 14627, USA
- Flaum Eye Institute, University of Rochester, Rochester, NY 14627, USA
| | - Kaitlin Wozniak
- The Institute of Optics, University of Rochester, Rochester, NY 14627, USA
- Flaum Eye Institute, University of Rochester, Rochester, NY 14627, USA
| | | | - Wayne H. Knox
- The Institute of Optics, University of Rochester, Rochester, NY 14627, USA
- Materials Science Program, University of Rochester, Rochester, NY 14627, USA
- Center for Visual Science, University of Rochester, Rochester, NY 14627, USA
| | - Krystel R. Huxlin
- The Institute of Optics, University of Rochester, Rochester, NY 14627, USA
- Flaum Eye Institute, University of Rochester, Rochester, NY 14627, USA
- Center for Visual Science, University of Rochester, Rochester, NY 14627, USA
| |
Collapse
|
56
|
Wang R, Nisar S, Vogel Z, Liu H, Wang Y. Dentin collagen denaturation status assessed by collagen hybridizing peptide and its effect on bio-stabilization of proanthocyanidins. Dent Mater 2022; 38:748-758. [PMID: 35431088 PMCID: PMC9060396 DOI: 10.1016/j.dental.2022.04.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 03/08/2022] [Accepted: 04/01/2022] [Indexed: 11/03/2022]
Abstract
OBJECTIVE To assess dentin collagen denaturation from phosphoric acid and enzyme treatments using collagen hybridizing peptide (CHP) and to investigate the effect of collagen denaturation on bio-stabilization promoted by proanthocyanidins (PA). METHODS Human molars were sectioned into 7-µm-thick dentin films, demineralized, and assigned to six groups: control with/without PA modification, H3PO4-treated collagen with/without PA modification, enzyme-treated collagen with/without PA modification. PA modification involved immersing collagen films in 0.65% PA for 30 s. H3PO4 and enzyme treatments were used to experimentally induce collagen denaturation, which was quantitated by fluorescence intensity (FI) from the fluorescently-conjugated-CHP (F-CHP) staining (n = 4). FTIR was used to characterize collagen structures. All groups were subject to collagenase digestion to test the bio-stabilization effect of PA on denatured collagen using weight loss analysis and hydroxyproline assay (n = 6). Data were analyzed using two-factor ANOVA and Games-Howell post hoc tests (α = 0.05). RESULTS FTIR showed collagen secondary structural changes after denaturation treatments and confirmed the incorporation and cross-linking of PA in control and treated collagen. F-CHP staining indicated high-degree, medium-degree, and low-degree collagen denaturation from H3PO4-treatment (FI = 83.22), enzyme-treatment (FI = 36.54), and control (FI = 6.01) respectively. PA modification significantly reduced the weight loss and hydroxyproline release of all groups after digestion (p < 0.0001), with the results correlated with FI values at r = 0.96-0.98. SIGNIFICANCE A molecular method CHP is introduced as a sensitive technique to quantitate dentin collagen denaturation for the first time. PA modification is shown to effectively stabilize denatured collagen against collagenase digestion, with the stabilization effect negatively associated with the collagen denaturation degree.
Collapse
|
57
|
An ultrastructural 3D reconstruction method for observing the arrangement of collagen fibrils and proteoglycans in the human aortic wall under mechanical load. Acta Biomater 2022; 141:300-314. [PMID: 35065266 DOI: 10.1016/j.actbio.2022.01.036] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 01/10/2022] [Accepted: 01/14/2022] [Indexed: 12/31/2022]
Abstract
An insight into changes of soft biological tissue ultrastructures under loading conditions is essential to understand their response to mechanical stimuli. Therefore, this study offers an approach to investigate the arrangement of collagen fibrils and proteoglycans (PGs), which are located within the mechanically loaded aortic wall. The human aortic samples were either fixed directly with glutaraldehyde in the load-free state or subjected to a planar biaxial extension test prior to fixation. The aortic ultrastructure was recorded using electron tomography. Collagen fibrils and PGs were segmented using convolutional neural networks, particularly the ESPNet model. The 3D ultrastructural reconstructions revealed a complex organization of collagen fibrils and PGs. In particular, we observed that not all PGs are attached to the collagen fibrils, but some fill the spaces between the fibrils with a clear distance to the collagen. The complex organization cannot be fully captured or can be severely misinterpreted in 2D. The approach developed opens up practical possibilities, including the quantification of the spatial relationship between collagen fibrils and PGs as a function of the mechanical load. Such quantification can also be used to compare tissues under different conditions, e.g., healthy and diseased, to improve or develop new material models. STATEMENT OF SIGNIFICANCE: The developed approach enables the 3D reconstruction of collagen fibrils and proteoglycans as they are embedded in the loaded human aortic wall. This methodological pipeline comprises the knowledge of arterial mechanics, imaging with transmission electron microscopy and electron tomography, segmentation of 3D image data sets with convolutional neural networks and finally offers a unique insight into the ultrastructural changes in the aortic tissue caused by mechanical stimuli.
Collapse
|
58
|
Henderson BS, Cudworth KF, Wale ME, Siegel DN, Lujan TJ. Tensile fatigue strength and endurance limit of human meniscus. J Mech Behav Biomed Mater 2022; 127:105057. [PMID: 35091175 PMCID: PMC9925119 DOI: 10.1016/j.jmbbm.2021.105057] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 12/06/2021] [Accepted: 12/17/2021] [Indexed: 11/29/2022]
Abstract
The knee menisci are prone to mechanical fatigue injury from the cyclic tensile stresses that are generated during daily joint loading. Here we characterize the tensile fatigue behavior of human medial meniscus and investigate the effect of aging on fatigue strength. Test specimens were excised from the medial meniscus of young (under 40 years) and older (over 65 years) fresh-frozen cadaver knees. Cyclic uniaxial tensile loads were applied parallel to the primary circumferential fibers at 70%, 50%, 40%, or 30% of the predicted ultimate tensile strength (UTS) until failure occurred or one million cycles was reached. Equations for fatigue strength (S-N curve) and the probability of fatigue failure (unreliability curves) were created from the measured number of cycles to failure. The mean number of cycles to failure at 70%, 50%, 40%, and 30% of UTS were estimated to be approximately 500, 40000, 340000, and 3 million cycles, respectively. The endurance limit, defined as the tensile stress that can be safely applied for the average lifetime of use (250 million cycles), was estimated to be 10% of UTS (∼1.0 MPa). When cyclic tensile stresses exceeded 30% of UTS (∼3.0 MPa), the probability of fatigue failure rapidly increased. While older menisci were generally weaker and more susceptible to fatigue failures at high-magnitude tensile stresses, both young and older age groups had similar fatigue resistance at low-magnitude tensile stresses. In addition, we found that fatigue failures occurred after the dynamic modulus decreased during cyclic loading by approximately 20%. This experimental study has quantified fundamental fatigue properties that are essential to properly predict and prevent injury in meniscus and other soft fibrous tissues.
Collapse
Affiliation(s)
- Bradley S. Henderson
- Department of Mechanical & Biomedical Engineering, Boise State University, Boise ID, USA
| | - Katelyn F. Cudworth
- Department of Mechanical & Biomedical Engineering, Boise State University, Boise ID, USA
| | - Madison E. Wale
- Department of Mechanical & Biomedical Engineering, Boise State University, Boise ID, USA
| | - Danielle N. Siegel
- Department of Mechanical & Biomedical Engineering, Boise State University, Boise ID, USA
| | - Trevor J. Lujan
- Department of Mechanical & Biomedical Engineering, Boise State University, Boise ID, USA
| |
Collapse
|
59
|
Growth and mechanobiology of the tendon-bone enthesis. Semin Cell Dev Biol 2022; 123:64-73. [PMID: 34362655 PMCID: PMC8810906 DOI: 10.1016/j.semcdb.2021.07.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/17/2021] [Accepted: 07/20/2021] [Indexed: 12/15/2022]
Abstract
Tendons are cable-like connective tissues that transfer both active and passive forces generated by skeletal muscle to bone. In the mature skeleton, the tendon-bone enthesis is an interfacial zone of transitional tissue located between two mechanically dissimilar tissues: compliant, fibrous tendon to rigid, dense mineralized bone. In this review, we focus on emerging areas in enthesis development related to its structure, function, and mechanobiology, as well as highlight established and emerging signaling pathways and physiological processes that influence the formation and adaptation of this important transitional tissue.
Collapse
|
60
|
Humeres C, Shinde AV, Hanna A, Alex L, Hernández SC, Li R, Chen B, Conway SJ, Frangogiannis NG. Smad7 effects on TGF-β and ErbB2 restrain myofibroblast activation and protect from postinfarction heart failure. J Clin Invest 2022; 132:146926. [PMID: 34905511 PMCID: PMC8803336 DOI: 10.1172/jci146926] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 12/09/2021] [Indexed: 01/02/2023] Open
Abstract
Repair of the infarcted heart requires TGF-β/Smad3 signaling in cardiac myofibroblasts. However, TGF-β-driven myofibroblast activation needs to be tightly regulated in order to prevent excessive fibrosis and adverse remodeling that may precipitate heart failure. We hypothesized that induction of the inhibitory Smad, Smad7, may restrain infarct myofibroblast activation, and we examined the molecular mechanisms of Smad7 actions. In a mouse model of nonreperfused infarction, Smad3 activation triggered Smad7 synthesis in α-SMA+ infarct myofibroblasts, but not in α-SMA-PDGFRα+ fibroblasts. Myofibroblast-specific Smad7 loss increased heart failure-related mortality, worsened dysfunction, and accentuated fibrosis in the infarct border zone and in the papillary muscles. Smad7 attenuated myofibroblast activation and reduced synthesis of structural and matricellular extracellular matrix proteins. Smad7 effects on TGF-β cascades involved deactivation of Smad2/3 and non-Smad pathways, without any effects on TGF-β receptor activity. Unbiased transcriptomic and proteomic analysis identified receptor tyrosine kinase signaling as a major target of Smad7. Smad7 interacted with ErbB2 in a TGF-β-independent manner and restrained ErbB1/ErbB2 activation, suppressing fibroblast expression of fibrogenic proteases, integrins, and CD44. Smad7 induction in myofibroblasts serves as an endogenous TGF-β-induced negative feedback mechanism that inhibits postinfarction fibrosis by restraining Smad-dependent and Smad-independent TGF-β responses, and by suppressing TGF-β-independent fibrogenic actions of ErbB2.
Collapse
Affiliation(s)
- Claudio Humeres
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York, USA
| | - Arti V. Shinde
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York, USA
| | - Anis Hanna
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York, USA
| | - Linda Alex
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York, USA
| | - Silvia C. Hernández
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York, USA
| | - Ruoshui Li
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York, USA
| | - Bijun Chen
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York, USA
| | - Simon J. Conway
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Nikolaos G. Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
61
|
Jones B, Debski A, Hans CP, Go MR, Agarwal G. Structurally abnormal collagen fibrils in abdominal aortic aneurysm resist platelet adhesion. J Thromb Haemost 2022; 20:470-477. [PMID: 34714974 DOI: 10.1111/jth.15576] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/16/2021] [Accepted: 10/27/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Platelet adhesion to the subendothelial collagen fibrils is one of the first steps in hemostasis. Understanding how structural perturbations in the collagen fibril affect platelet adhesion can provide novel insights into disruption of hemostasis in various diseases. We have recently identified the presence of abnormal collagen fibrils with compromised D-periodic banding in the extracellular matrix remodeling present in abdominal aortic aneurysms (AAA). OBJECTIVE In this study, we employed multimodal microscopy approaches to characterize how collagen fibril structure impacts platelet adhesion in clinical AAA tissues. METHODS Ultrastructural atomic force microscopy (AFM) analysis was performed on tissue sections after staining with fluorescently labeled collagen hybridizing peptide (CHP) to recognize degraded collagen. Second harmonic generation (SHG) microscopy was used on CHP-stained sections to identify regions of intact versus degraded collagen. Finally, platelet adhesion was identified via SHG and indirect immunofluorescence on the same tissue sections. RESULTS Our results indicate that ultrastructural features characterizing collagen fibril abnormalities coincide with CHP staining. SHG signal was absent from CHP-positive regions. Additionally, platelet binding was primarily localized to regions with SHG signal. Abnormal collagen fibrils present in AAA (in SHG negative regions) were thus found to inhibit platelet adhesion compared to normal fibrils. CONCLUSIONS Our investigations reveal how the collagen fibril structure in the vessel wall can serve as another regulator of platelet-collagen adhesion. These results can be broadly applied to understand the role of collagen fibril structure in regulating thrombosis or bleeding disorders.
Collapse
Affiliation(s)
- Blain Jones
- Biomedical Engineering Graduate Program, The Ohio State University, Columbus, Ohio, USA
| | - Anna Debski
- Department of Material Science and Engineering, The Ohio State University, Columbus, Ohio, USA
| | - Chetan P Hans
- Department of Cardiovascular Medicine, Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Michael R Go
- Division of Vascular Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Gunjan Agarwal
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
62
|
Qi Y, Zhou D, Kessler JL, Qiu R, Yu SM, Li G, Qin Z, Li Y. Terminal repeats impact collagen triple-helix stability through hydrogen bonding. Chem Sci 2022; 13:12567-12576. [PMID: 36382282 PMCID: PMC9629113 DOI: 10.1039/d2sc03666e] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/10/2022] [Indexed: 11/22/2022] Open
Abstract
Nearly 30% of human proteins have tandem repeating sequences. Structural understanding of the terminal repeats is well-established for many repeat proteins with the common α-helix and β-sheet foldings. By contrast, the sequence–structure interplay of the terminal repeats of the collagen triple-helix remains to be fully explored. As the most abundant human repeat protein and the most prevalent structural component of the extracellular matrix, collagen features a hallmark triple-helix formed by three supercoiled polypeptide chains of long repeating sequences of the Gly–X–Y triplets. Here, with CD characterization of 28 collagen-mimetic peptides (CMPs) featuring various terminal motifs, as well as DSC measurements, crystal structure analysis, and computational simulations, we show that CMPs only differing in terminal repeat may have distinct end structures and stabilities. We reveal that the cross-chain hydrogen bonding mediated by the terminal repeat is key to maintaining the triple-helix's end structure, and that disruption of it with a single amide to carboxylate substitution can lead to destabilization as drastic as 19 °C. We further demonstrate that the terminal repeat also impacts how strong the CMP strands form hybrid triple-helices with unfolded natural collagen chains in tissue. Our findings provide a spatial profile of hydrogen bonding within the CMP triple-helix, marking a critical guideline for future crystallographic or NMR studies of collagen, and algorithms for predicting triple-helix stability, as well as peptide-based collagen assemblies and materials. This study will also inspire new understanding of the sequence–structure relationship of many other complex structural proteins with repeating sequences. Collagen mimetic peptides (CMPs) only differing in terminal repeat have distinct stabilities and end structures due to a spatial hydrogen bonding profile that is useful for future crystallography, algorithm prediction, and materials of collagen.![]()
Collapse
Affiliation(s)
- Yingying Qi
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
- Cardiac Surgery and Structural Heart Disease Unit of Cardiovascular Center, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
- Department of Radiology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Daoning Zhou
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Julian L. Kessler
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah 84112, USA
| | - Rongmao Qiu
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - S. Michael Yu
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah 84112, USA
| | - Gang Li
- Cardiac Surgery and Structural Heart Disease Unit of Cardiovascular Center, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Zhao Qin
- Department of Civil & Environmental Engineering, College of Engineering & Computer Science, Syracuse University, Syracuse, New York 13244, USA
| | - Yang Li
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| |
Collapse
|
63
|
Liu L, Huang K, Li W, Qiu R, Fang Y, Huang Y, Zhao S, Lv H, Zhang K, Shan H, Li Y. Molecular Imaging of Collagen Destruction of the Spine. ACS NANO 2021; 15:19138-19149. [PMID: 34738460 DOI: 10.1021/acsnano.1c07112] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
As the leading cause of disability worldwide, low back pain is commonly caused by biomechanical and catabolic disruptions to key structures of the spine, such as intervertebral discs and facet joints. To date, accurate, noninvasive detection of microdestruction within these tissues remains an elusive goal. Here, we report an in vivo imaging approach based on a collagen hybridizing peptide (CHP) that specifically targets disruption to the extracellular matrix architecture at the molecular scale─the denatured collagen molecules. Utilizing fluorescently labeled CHPs, live animal imaging, and light sheet fluorescence microscopy, we mapped collagen destruction in the lumbar spines in 3D, revealing that under normal conditions collagen destruction was localized to load-bearing anatomical structures including annulus fibrosus of the disc and the facet joints, where aging, tensile force (hindlimb suspension), and disc degeneration (needle puncture) escalated the CHP-binding in specific mouse models. We showed that targeting denatured collagen molecules allowed for an accurate, quantifiable interrogation of the structural integrity of these spinal matrixes with a greater sensitivity than anatomical imaging and histology. Finally, we demonstrated CHP's binding to degenerated human discs, suggesting exciting potentials for applying CHP for diagnosing, monitoring, and treating various spinal disorders, including intervertebral disc degeneration, facet joint osteoarthritis, and ankylosing spondylitis.
Collapse
Affiliation(s)
- Lei Liu
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
- Department of Spine Surgery, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Kui Huang
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Wei Li
- Department of Pathology, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Rongmao Qiu
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Yijie Fang
- Department of Radiology, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Yongjie Huang
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Suwen Zhao
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Hai Lv
- Department of Spine Surgery, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Kuibo Zhang
- Department of Spine Surgery, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Hong Shan
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
- Department of Interventional Medicine, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Yang Li
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| |
Collapse
|
64
|
Reversible changes in the 3D collagen fibril architecture during cyclic loading of healthy and degraded cartilage. Acta Biomater 2021; 136:314-326. [PMID: 34563724 PMCID: PMC8631461 DOI: 10.1016/j.actbio.2021.09.037] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 08/19/2021] [Accepted: 09/20/2021] [Indexed: 01/09/2023]
Abstract
Biomechanical changes to the collagen fibrillar architecture in articular cartilage are believed to play a crucial role in enabling normal joint function. However, experimentally there is little quantitative knowledge about the structural response of the Type II collagen fibrils in cartilage to cyclic loading in situ, and the mechanisms that drive the ability of cartilage to withstand long-term repetitive loading. Here we utilize synchrotron small-angle X-ray scattering (SAXS) combined with in-situ cyclic loading of bovine articular cartilage explants to measure the fibrillar response in deep zone articular cartilage, in terms of orientation, fibrillar strain and inter-fibrillar variability in healthy cartilage and cartilage degraded by exposure to the pro-inflammatory cytokine IL-1β. We demonstrate that under repeated cyclic loading the fibrils reversibly change the width of the fibrillar orientation distribution whilst maintaining a largely consistent average direction of orientation. Specifically, the effect on the fibrillar network is a 3-dimensional conical orientation broadening around the normal to the joint surface, inferred by 3D reconstruction of X-ray scattering peak intensity distributions from the 2D pattern. Further, at the intrafibrillar level, this effect is coupled with reversible reduction in fibrillar pre-strain under compression, alongside increase in the variability of fibrillar pre-strain. In IL-1β degraded cartilage, the collagen rearrangement under cyclic loading is disrupted and associated with reduced tissue stiffness. These finding have implications as to how changes in local collagen nanomechanics might drive disease progression or vice versa in conditions such as osteoarthritis and provides a pathway to a mechanistic understanding of such diseases. Statement of significance Structural deterioration in biomechanically loaded musculoskeletal organs, e.g., joint osteoarthritis and back pain, are linked to breakdown and changes in their collagen-rich cartilaginous tissue matrix. A critical component enabling cartilage biomechanics is the ultrastructural collagen fibrillar network in cartilage. However, experimental probes of the dynamic structural response of cartilage collagen to biomechanical loads are limited. Here, we use X-ray scattering during cyclic loading (as during walking) on joint tissue to show that cartilage fibrils resist loading by a reversible, three-dimensional orientation broadening and disordering mechanism at the molecular level, and that inflammation reduces this functionality. Our results will help understand how changes to small-scale tissue mechanisms are linked to ageing and osteoarthritic progression, and development of biomaterials for joint replacements.
Collapse
|
65
|
Brandt ZJ, Collery RF, Besharse JC, Link BA. Ablation of mpeg+ Macrophages Exacerbates mfrp-Related Hyperopia. Invest Ophthalmol Vis Sci 2021; 62:13. [PMID: 34913948 PMCID: PMC8684298 DOI: 10.1167/iovs.62.15.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Proper refractive development of the eye, termed emmetropization, is critical for focused vision and is impacted by both genetic determinants and several visual environment factors. Improper emmetropization caused by genetic variants can lead to congenital hyperopia, which is characterized by small eyes and relatively short ocular axial length. To date, variants in only four genes have been firmly associated with human hyperopia, one of which is MFRP. Zebrafish mfrp mutants also have hyperopia and, similar to reports in mice, exhibit increased macrophage recruitment to the retina. The goal of this research was to examine the effects of macrophage ablation on emmetropization and mfrp-related hyperopia. Methods We utilized a chemically inducible, cell-specific ablation system to deplete macrophages in both wild-type and mfrp mutant zebrafish. Spectral-domain optical coherence tomography was then used to measure components of the eye and determine relative refractive state. Histology, immunohistochemistry, and transmission electron microscopy were used to further study the eyes. Results Although macrophage ablation does not cause significant changes to the relative refractive state of wild-type zebrafish, macrophage ablation in mfrp mutants significantly exacerbates their hyperopic phenotype, resulting in a relative refractive error 1.3 times higher than that of non-ablated mfrp siblings. Conclusions Genetic inactivation of mfrp leads to hyperopia, as well as abnormal accumulation of macrophages in the retina. Ablation of the mpeg1-positive macrophage population exacerbates the hyperopia, suggesting that macrophages may be recruited in an effort help preserve emmetropization and ameliorate hyperopia.
Collapse
Affiliation(s)
- Zachary J Brandt
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Ross F Collery
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Joseph C Besharse
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Brian A Link
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| |
Collapse
|
66
|
Golman M, Birman V, Thomopoulos S, Genin GM. Enthesis strength, toughness and stiffness: an image-based model comparing tendon insertions with varying bony attachment geometries. J R Soc Interface 2021; 18:20210421. [PMID: 34932930 PMCID: PMC8692040 DOI: 10.1098/rsif.2021.0421] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 11/25/2021] [Indexed: 12/24/2022] Open
Abstract
Tendons of the body differ dramatically in their function, mechanics and range of motion, but all connect to bone via an enthesis. Effective force transfer at the enthesis enables joint stability and mobility, with strength and stiffness arising from a fibrous architecture. However, how enthesis toughness arises across tendons with diverse loading orientations remains unclear. To study this, we performed simultaneous imaging of the bone and tendon in entheses that represent the range of tendon-to-bone insertions and extended a mathematical model to account for variations in insertion and bone geometry. We tested the hypothesis that toughness, across a range of tendon entheses, could be explained by differences observed in interactions between fibre architecture and bone architecture. In the model, toughness arose from fibre reorientation, recruitment and rupture, mediated by interactions between fibres at the enthesis and the bony ridge abutting it. When applied to tendons sometimes characterized as either energy-storing or positional, the model predicted that entheses of the former prioritize toughness over strength, while those of the latter prioritize consistent stiffness across loading directions. Results provide insight into techniques for surgical repair of tendon-to-bone attachments, and more broadly into mechanisms for the attachment of highly dissimilar materials.
Collapse
Affiliation(s)
- Mikhail Golman
- Columbia University, Black Building, Room 1408, 650 W 168 ST, New York, NY 10032-3702, USA
| | - Victor Birman
- Missouri University of Science and Technology, Rolla, MO 65409, USA
| | - Stavros Thomopoulos
- Columbia University, Black Building, Room 1408, 650 W 168 ST, New York, NY 10032-3702, USA
| | - Guy M. Genin
- Washington University, Jubel Hall, Room 103F, 1 Brookings Drive, St Louis, MO 63130, USA
| |
Collapse
|
67
|
Golman M, Abraham AC, Kurtaliaj I, Marshall BP, Hu YJ, Schwartz AG, Guo XE, Birman V, Thurner PJ, Genin GM, Thomopoulos S. Toughening mechanisms for the attachment of architectured materials: The mechanics of the tendon enthesis. SCIENCE ADVANCES 2021; 7:eabi5584. [PMID: 34826240 PMCID: PMC8626067 DOI: 10.1126/sciadv.abi5584] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 10/06/2021] [Indexed: 05/09/2023]
Abstract
Architectured materials offer tailored mechanical properties but are limited in engineering applications due to challenges in maintaining toughness across their attachments. The enthesis connects tendon and bone, two vastly different architectured materials, and exhibits toughness across a wide range of loadings. Understanding the mechanisms by which this is achieved could inform the development of engineered attachments. Integrating experiments, simulations, and previously unexplored imaging that enabled simultaneous observation of mineralized and unmineralized tissues, we identified putative mechanisms of enthesis toughening in a mouse model and then manipulated these mechanisms via in vivo control of mineralization and architecture. Imaging uncovered a fibrous architecture within the enthesis that controls trade-offs between strength and toughness. In vivo models of pathology revealed architectural adaptations that optimize these trade-offs through cross-scale mechanisms including nanoscale protein denaturation, milliscale load-sharing, and macroscale energy absorption. Results suggest strategies for optimizing architecture for tough bimaterial attachments in medicine and engineering.
Collapse
Affiliation(s)
- Mikhail Golman
- Department of Orthopedic Surgery, Columbia University, New York, NY 10032, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Adam C. Abraham
- Department of Orthopedic Surgery, Columbia University, New York, NY 10032, USA
| | - Iden Kurtaliaj
- Department of Orthopedic Surgery, Columbia University, New York, NY 10032, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Brittany P. Marshall
- Department of Orthopedic Surgery, Columbia University, New York, NY 10032, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Yizhong Jenny Hu
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Andrea G. Schwartz
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University, St. Louis, MO 63130, USA
| | - X. Edward Guo
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Victor Birman
- Missouri University of Science and Technology, Rolla, MO 65409, USA
| | - Philipp J. Thurner
- Institute of Lightweight Design and Structural Biomechanics, Vienna University of Technology, Vienna, Austria
| | - Guy M. Genin
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University, St. Louis, MO 63130, USA
| | - Stavros Thomopoulos
- Department of Orthopedic Surgery, Columbia University, New York, NY 10032, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| |
Collapse
|
68
|
Converse MI, Nye KS, Dahl MJ, Albertine KH, Monson KL. Stretch-Induced Intimal Failure in Isolated Cerebral Arteries as a Function of Development. Ann Biomed Eng 2021; 49:3540-3549. [PMID: 34725768 DOI: 10.1007/s10439-021-02869-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 09/27/2021] [Indexed: 11/29/2022]
Abstract
Recent clinical studies have shown that traumatic brain injury is a significant risk factor for stroke. Motivated to better understand possible mechanisms of this association, we studied subfailure disruption of the intima in overstretched sheep cerebral arteries, as this has been implicated in the increased risk of stroke following blunt cerebrovascular injury. Middle cerebral arteries from four age groups (ranging from fetal to adult) were stretched axially to failure, and intimal disruption was captured with a video camera. All vessels demonstrated intimal disruption prior to catastrophic failure, with nearly all incurring disruption at stretch values well below those at ultimate stress (means of 1.56 and 1.73, respectively); the lowest stretch associated with intimal disruption was 1.29. The threshold of intimal failure was independent of age. Additional analysis showed that disruption included failure of both the endothelium and internal elastic lamina. Although our experiments were conducted at quasi-static rates, the results likely have important implications for vessel function following trauma. Future work should seek to identify subfailure disruption of the cerebrovasculature in head trauma.
Collapse
Affiliation(s)
- Matthew I Converse
- Department of Mechanical Engineering, University of Utah, 1495 E. 100 S. MEK 1550, Salt Lake City, UT, 84112, USA
| | - Kevin S Nye
- Department of Mechanical Engineering, University of Utah, 1495 E. 100 S. MEK 1550, Salt Lake City, UT, 84112, USA
| | - Mar Janna Dahl
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - Kurt H Albertine
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
- Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT, USA
| | - Kenneth L Monson
- Department of Mechanical Engineering, University of Utah, 1495 E. 100 S. MEK 1550, Salt Lake City, UT, 84112, USA.
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
69
|
Zitnay JL, Lin AH, Weiss JA. Tendons exhibit greater resistance to tissue and molecular-level damage with increasing strain rate during cyclic fatigue. Acta Biomater 2021; 134:435-442. [PMID: 34314889 DOI: 10.1016/j.actbio.2021.07.045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 07/15/2021] [Accepted: 07/21/2021] [Indexed: 11/28/2022]
Abstract
Musculoskeletal soft connective tissues are commonly injured due to repetitive use, but the evolution of mechanical damage to the tissue structure during repeated loading is poorly understood. We investigated the strain-rate dependence of mechanical denaturation of collagen as a form of structural microdamage accumulation during creep fatigue loading of rat tail tendon fascicles. We cycled tendons at three strain rates to the same maximum stress relative to their rate-dependent tensile strength. Collagen denaturation at distinct points during the fatigue process was measured by fluorescence quantification of collagen hybridizing peptide binding. The amount of collagen denaturation was significantly correlated with fascicle creep strain, independent of the cyclic strain rate, supporting our hypothesis that tissue level creep is caused by collagen triple-helix unfolding. Samples that were loaded faster experienced more creep strain and denaturation as a function of the number of loading cycles relative to failure. Although this increased damage capacity at faster rates may serve as a protective measure during high-rate loading events, it may also predispose these tissues to subsequent injury and indicate a mechanism of overuse injury development. These results build on evidence that molecular-level collagen denaturation is the fundamental mechanism of structural damage to tendons during tensile loading. STATEMENT OF SIGNIFICANCE: This study is the first to investigate the accumulation of denatured collagen in tendons throughout fatigue loading when the maximum stress is scaled with the applied strain rate. The amount of denatured collagen was correlated with creep strain, independent of strain rate, but samples that were cycled faster withstood greater amounts of denaturation before failure. Differential accumulation of collagen damage between fast and slow repetitive loading has relevance toward understanding the prevalence of overuse musculoskeletal injuries following sudden changes in activity level. Since collagen is a ubiquitous biological structural component, the basic patterns and mechanisms of loading-induced collagen damage in connective tissues are relevant for understanding injury and disease in other tissues, including those from the cardiovascular and pulmonary systems.
Collapse
Affiliation(s)
- Jared L Zitnay
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA; Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT, USA
| | - Allen H Lin
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA; Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT, USA
| | - Jeffrey A Weiss
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA; Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT, USA; Department of Orthopaedics, University of Utah, Salt Lake City, UT, USA; School of Computing, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
70
|
Mechanical characterisation of the human dura mater, falx cerebri and superior sagittal sinus. Acta Biomater 2021; 134:388-400. [PMID: 34314888 DOI: 10.1016/j.actbio.2021.07.043] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/01/2021] [Accepted: 07/21/2021] [Indexed: 01/14/2023]
Abstract
The cranial meninges have been shown to play a pivotal role in traumatic brain injury mechanopathology. However, while the mechanical response of the brain and its many subregions have been studied extensively, the meninges have conventionally been overlooked. This paper presents the first comparative mechanical analysis of human dura mater, falx cerebri and superior sagittal sinus tissues. Biaxial tensile analysis identified that these tissues are mechanically heterogeneous, in contrast to the assumption that the tissues are mechanically homogeneous which is typically employed in FE model design. A thickness of 0.91 ± 0.05 (standard error) mm for the falx cerebri was also identified. This data can aid in improving the biofidelity of the influential falx structure in FE models. Additionally, the use of a collagen hybridizing peptide on the superior sagittal sinus suggests this structure is particularly susceptible to the effects of circumferential stretch, which may have important implications for clinical treatment of dural venous sinus pathologies. Collectively, this research progresses understanding of meningeal mechanical and structural characteristics and may aid in elucidating the behaviour of these tissues in healthy and diseased conditions. STATEMENT OF SIGNIFICANCE: This study presents the first evaluation of human falx cerebri and superior sagittal sinus mechanical, geometrical and structural properties, along with a comparison to cranial dura mater. To mechanically characterise the tissues, biaxial tensile testing is conducted on the tissues. This analysis identifies, for the first time, mechanical stiffness differences between these tissues. Additionally, geometrical analysis identifies that there are thickness differences between the tissues. The evaluation of human meningeal tissues allows for direct implementation of the novel data to finite element head injury models to enable improved biofidelity of these influential structures in traumatic brain injury simulations. This work also identifies that the superior sagittal sinus may be easily damaged during clinical angioplasty procedures, which may inform the treatment of dural sinus pathologies.
Collapse
|
71
|
Wang Y, Tian X, Liu X, Xing J, Guo C, Du Y, Zhang H, Wang W. Focusing on intramuscular connective tissue: Effect of cooking time and temperature on physical, textual, and structural properties of yak meat. Meat Sci 2021; 184:108690. [PMID: 34656007 DOI: 10.1016/j.meatsci.2021.108690] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 12/20/2022]
Abstract
This study aimed to evaluate the effects of different cooking time (2, 4, and 6 h) and temperature (50, 60, 70, 80, and 90 °C) on physical, textual, and structural properties of longissimus lumborum muscle of yak, and to explore the thermal denaturation process of intramuscular collagen by using a new tool (collagen hybridizing peptide staining, CHP staining). The results showed that tenderness was affected by the interaction of cooking time and temperature and the changes in moisture and collagen composition. In comparison with cooking time, temperature had more obvious effects on cooking loss, moisture content and redness. Scanning electron microscopy showed that as the temperature increased, intramuscular connective tissue gradually degraded, and muscle fibers became more compact. CHP staining showed that the collagen in the perimysium first denatured at 50 °C, and more and more collagen denatured and degraded as the temperature increased.
Collapse
Affiliation(s)
- Yang Wang
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Xiaojing Tian
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin University of Science and Technology, Tianjin 300457, China.
| | - Xinzhu Liu
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Jinfeng Xing
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Chen Guo
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Yuehong Du
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Huan Zhang
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Wenhang Wang
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin University of Science and Technology, Tianjin 300457, China.
| |
Collapse
|
72
|
Mangubat-Medina AE, Ball ZT. Triggering biological processes: methods and applications of photocaged peptides and proteins. Chem Soc Rev 2021; 50:10403-10421. [PMID: 34320043 DOI: 10.1039/d0cs01434f] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
There has been a significant push in recent years to deploy fundamental knowledge and methods of photochemistry toward biological ends. Photoreactive groups have enabled chemists to activate biological function using the concept of photocaging. By granting spatiotemporal control over protein activation, these photocaging methods are fundamental in understanding biological processes. Peptides and proteins are an important group of photocaging targets that present conceptual and technical challenges, requiring precise chemoselectivity in complex polyfunctional environments. This review focuses on recent advances in photocaging techniques and methodologies, as well as their use in living systems. Photocaging methods include genetic and chemical approaches that require a deep understanding of structure-function relationships based on subtle changes in primary structure. Successful implementation of these ideas can shed light on important spatiotemporal aspects of living systems.
Collapse
Affiliation(s)
| | - Zachary T Ball
- Department of Chemistry, Rice University, Houston, TX, 77005, USA.
| |
Collapse
|
73
|
Zhang X, Lee SJ, Wang MM. Hydrolysis of a second Asp-Pro site at the N-terminus of NOTCH3 in inherited vascular dementia. Sci Rep 2021; 11:17246. [PMID: 34446744 PMCID: PMC8390697 DOI: 10.1038/s41598-021-96679-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 08/17/2021] [Indexed: 12/02/2022] Open
Abstract
Cerebrovascular pathology at the biochemical level has been informed by the study of cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), a vascular disorder caused by NOTCH3 mutations. Previous work in CADASIL described N-terminal proteolysis of NOTCH3 generated by specific non-enzymatic cleavage of the first Asp-Pro sequence of the protein. Here, we investigated whether the second Asp-Pro peptide bond (residues 121–122) of NOTCH3 is cleaved in CADASIL. Monospecific antibodies were generated that recognize the neo-epitope predicted to be generated by cleavage after Asp121. These antibodies were used to localize cleavage events at Asp121 in post-mortem CADASIL and control brain tissue and to investigate factors that regulate cleavage at Asp121. We report that cleavage at Asp121 occurs at a high level in the arterial media of CADASIL cerebral arteries. Leptomeningeal arteries demonstrated substantially more cleavage product than penetrating arteries in the white matter, and control vessels harbored only a small amount of cleaved NOTCH3. Proteolysis at Asp121 occurred in purified preparations of NOTCH3 ectodomain, was increased by acidic pH and reductive conditions, and required native protein conformation for cleavage. Increasing the concentration of NOTCH3 EGF-like domain protein elevated the level of proteolysis. On the other hand, several polyanionic chemicals potently blocked cleavage at Asp121. These studies demonstrate that the NOTCH3 protein in CADASIL is cleaved in multiple locations at labile Asp-Pro peptide bonds. As such, chronic brain vascular disease, like other neurodegenerative conditions, features proteolysis of pathological proteins at multiple sites which may generate small pathological peptides.
Collapse
Affiliation(s)
- Xiaojie Zhang
- Department of Neurology, University of Michigan, 7725 Medical Science Building II Box 5622, 1137 Catherine St., Ann Arbor, MI, 48109-5622, USA.,Neurology Service, Department of Veterans Affairs, VA Ann Arbor Healthcare System, Ann Arbor, MI, 48105, USA
| | - Soo Jung Lee
- Department of Neurology, University of Michigan, 7725 Medical Science Building II Box 5622, 1137 Catherine St., Ann Arbor, MI, 48109-5622, USA.,Neurology Service, Department of Veterans Affairs, VA Ann Arbor Healthcare System, Ann Arbor, MI, 48105, USA
| | - Michael M Wang
- Department of Neurology, University of Michigan, 7725 Medical Science Building II Box 5622, 1137 Catherine St., Ann Arbor, MI, 48109-5622, USA. .,Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA. .,Neurology Service, Department of Veterans Affairs, VA Ann Arbor Healthcare System, Ann Arbor, MI, 48105, USA.
| |
Collapse
|
74
|
Marino M, Vairo G, Wriggers P. Mechano-chemo-biological Computational Models for Arteries in Health, Disease and Healing: From Tissue Remodelling to Drug-eluting Devices. Curr Pharm Des 2021; 27:1904-1917. [PMID: 32723253 DOI: 10.2174/1381612826666200728145752] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 06/14/2020] [Indexed: 11/22/2022]
Abstract
This review aims to highlight urgent priorities for the computational biomechanics community in the framework of mechano-chemo-biological models. Recent approaches, promising directions and open challenges on the computational modelling of arterial tissues in health and disease are introduced and investigated, together with in silico approaches for the analysis of drug-eluting stents that promote pharmacological-induced healing. The paper addresses a number of chemo-biological phenomena that are generally neglected in biomechanical engineering models but are most likely instrumental for the onset and the progression of arterial diseases. An interdisciplinary effort is thus encouraged for providing the tools for an effective in silico insight into medical problems. An integrated mechano-chemo-biological perspective is believed to be a fundamental missing piece for crossing the bridge between computational engineering and life sciences, and for bringing computational biomechanics into medical research and clinical practice.
Collapse
Affiliation(s)
- Michele Marino
- Institute of Continuum Mechanics, Leibniz Universität Hannover, An der Universität 1, 30823 Garbsen, Germany
| | - Giuseppe Vairo
- Department of Civil Engineering and Computer Science, University of Rome "Tor Vergata" via del Politecnico 1, 00133 Rome, Italy
| | - Peter Wriggers
- Institute of Continuum Mechanics, Leibniz Universität Hannover, An der Universität 1, 30823 Garbsen, Germany
| |
Collapse
|
75
|
Nallasamy S, Palacios HH, Setlem R, Caraballo MC, Li K, Cao E, Shankaran M, Hellerstein M, Mahendroo M. Transcriptome and proteome dynamics of cervical remodeling in the mouse during pregnancy. Biol Reprod 2021; 105:1257-1271. [PMID: 34309663 DOI: 10.1093/biolre/ioab144] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/02/2021] [Accepted: 07/20/2021] [Indexed: 11/14/2022] Open
Abstract
During gestation, the female reproductive tract must maintain pregnancy while concurrently preparing for parturition. Here, we explore the transitions in gene expression and protein turnover (fractional synthesis rates [FSR]) by which the cervix implements a transition from rigid to compliant. Shifts in gene transcription to achieve immune tolerance and alter epithelial cell programs begin in early pregnancy. Subsequently, in mid-to-late pregnancy transcriptional programs emerge that promote structural reorganization of the extracellular matrix (ECM). Stable isotope labeling revealed a striking slowdown of overall FSRs across the proteome on gestation day 6 that reverses in mid-to-late pregnancy. An exception was soluble fibrillar collagens and proteins of collagen assembly, which exhibit high turnover in non-pregnant cervix compared to other tissues and FSRs that continue throughout pregnancy. This finding provides a mechanism to explain how cross-linked collagen is replaced by newly synthesized, less-cross-linked collagens, which allows increased tissue compliance during parturition. The rapid transition requires a reservoir of newly synthesized, less cross-linked collagens, which is assured by the high FSR of soluble collagens in the cervix. These findings suggest a previously unrecognized form of "metabolic flexibility" for ECM in the cervix that underlies rapid transformation in compliance to allow parturition.
Collapse
Affiliation(s)
- Shanmugasundaram Nallasamy
- Department of Ob/Gyn and Cecil H. and Ida Green Center for Reproductive Biological Science, The University of Texas Southwestern Medical Center, Dallas, TX
| | - Hector H Palacios
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, CA
| | - Rohit Setlem
- Department of Ob/Gyn and Cecil H. and Ida Green Center for Reproductive Biological Science, The University of Texas Southwestern Medical Center, Dallas, TX
| | - Mariano Colon Caraballo
- Department of Ob/Gyn and Cecil H. and Ida Green Center for Reproductive Biological Science, The University of Texas Southwestern Medical Center, Dallas, TX
| | - Kelvin Li
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, CA
| | - Edward Cao
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, CA
| | - Mahalakshmi Shankaran
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, CA
| | - Marc Hellerstein
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, CA
| | - Mala Mahendroo
- Department of Ob/Gyn and Cecil H. and Ida Green Center for Reproductive Biological Science, The University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
76
|
Kessler JL, Kang G, Qin Z, Kang H, Whitby FG, Cheatham TE, Hill CP, Li Y, Yu SM. Peptoid Residues Make Diverse, Hyperstable Collagen Triple-Helices. J Am Chem Soc 2021; 143:10910-10919. [PMID: 34255504 DOI: 10.1021/jacs.1c00708] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
As the only ribosomally encoded N-substituted amino acid, proline promotes distinct secondary protein structures. The high proline content in collagen, the most abundant protein in the human body, is crucial to forming its hallmark structure: the triple-helix. For over five decades, proline has been considered compulsory for synthetic designs aimed at recapitulating collagen's structure and properties. Here we describe that N-substituted glycines (N-glys), also known as peptoid residues, exhibit a general triple-helical propensity similar to or greater than proline, enabling synthesis of stable triple-helical collagen mimetic peptides (CMPs) with unprecedented side chain diversity. Supported by atomic-resolution crystal structures as well as circular dichroism and computational characterizations spanning over 30 N-gly-containing CMPs, we discovered that N-glys stabilize the triple-helix primarily by sterically preorganizing individual chains into the polyproline-II helix. We demonstrated that N-glys with exotic side chains including a "click"-able alkyne and a photosensitive side chain enable CMPs for functional applications including the spatiotemporal control of cell adhesion and migration. The structural principles uncovered in this study open up opportunities for a new generation of collagen-mimetic therapeutics and materials.
Collapse
Affiliation(s)
- Julian L Kessler
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah 84112, United States
| | - Grace Kang
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah 84112, United States
| | - Zhao Qin
- Department of Civil & Environmental Engineering, College of Engineering & Computer Science, Syracuse University, Syracuse, New York 13244, United States
| | - Helen Kang
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah 84112, United States
| | - Frank G Whitby
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, Utah 84112, United States
| | - Thomas E Cheatham
- Department of Medicinal Chemistry, College of Pharmacy, L. S. Skaggs Pharmacy Research Institute, University of Utah, Salt Lake City, Utah 84112, United States
| | - Christopher P Hill
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, Utah 84112, United States
| | - Yang Li
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah 84112, United States
| | - S Michael Yu
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah 84112, United States.,Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, Utah 84112, United States
| |
Collapse
|
77
|
A microstructural model of tendon failure. J Mech Behav Biomed Mater 2021; 122:104665. [PMID: 34311323 DOI: 10.1016/j.jmbbm.2021.104665] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/27/2021] [Accepted: 06/25/2021] [Indexed: 02/07/2023]
Abstract
Collagen fibrils are the most important structural component of tendons. Their crimped structure and parallel arrangement within the tendon lead to a distinctive non-linear stress-strain curve when a tendon is stretched. Microstructural models can be used to relate microscale collagen fibril mechanics to macroscale tendon mechanics, allowing us to identify the mechanisms behind each feature present in the stress-strain curve. Most models in the literature focus on the elastic behaviour of the tendon, and there are few which model beyond the elastic limit without introducing phenomenological parameters. We develop a model, built upon a collagen recruitment approach, that only contains microstructural parameters. We split the stress in the fibrils into elastic and plastic parts, and assume that the fibril yield stretch and rupture stretch are each described by a distribution function, rather than being single-valued. By changing the shapes of the distributions and their regions of overlap, we can produce macroscale tendon stress-strain curves that generate the full range of features observed experimentally, including those that could not be explained using existing models. These features include second linear regions occurring after the tendon has yielded, and step-like failure behaviour present after the stress has peaked. When we compare with an existing model, we find that our model reduces the average root mean squared error from 4.53MPa to 2.29MPa, and the resulting parameter values are closer to those found experimentally. Since our model contains only parameters that have a direct physical interpretation, it can be used to predict how processes such as ageing, disease, and injury affect the mechanical behaviour of tendons, provided we can quantify the effects of these processes on the microstructure.
Collapse
|
78
|
Khajehsaeid H, Tehrani M, Alaghehband N. Anisotropic damage of soft tissues in supra-physiological deformations. J Biomech 2021; 124:110548. [PMID: 34171681 DOI: 10.1016/j.jbiomech.2021.110548] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 05/31/2021] [Indexed: 10/21/2022]
Abstract
Soft tissues may undergo mechanical damage under supra-physiological deformations caused by medical treatments such as balloon-angioplasty and stent deployment. This damage is exhibited as a softening in the mechanical behavior of tissues. In this work, alteration of the collagen network is treated as the origin of damage and loss of stiffness. Inspired by the hierarchical structure of the collagen network, the mechanical properties of collagenous tissues are connected to model parameters. Softening of esophageal and arterial tissues under directional cyclic loading is investigated to determine evolution of the associated material parameters through damage progress. An evolution law is proposed which predicts the mechanical behavior of tissues after excessive deformations. Various deformation measures are examined as the damage parameter to determine the most appropriate one for general 3D loading. It is observed that, if the Green-Lagrange strain in the direction of the fibers is used as the damage parameter, the proposed law well describes the evolution of the collagen network's stiffness. The results not only facilitate prediction of the deformation-induced damage under supra-physiological deformations but also are useful for surgeons in better planning of surgical procedures and stents design.
Collapse
Affiliation(s)
- H Khajehsaeid
- WMG, University of Warwick, Coventry CV4 7AL, United Kingdom; School of Engineering, University of Tabriz, Tabriz, Iran.
| | - M Tehrani
- School of Engineering, University of Tabriz, Tabriz, Iran
| | - N Alaghehband
- School of Engineering, University of Tabriz, Tabriz, Iran
| |
Collapse
|
79
|
Rausch MK, Sugerman GP, Kakaletsis S, Dortdivanlioglu B. Hyper-viscoelastic damage modeling of whole blood clot under large deformation. Biomech Model Mechanobiol 2021; 20:1645-1657. [PMID: 34080080 DOI: 10.1007/s10237-021-01467-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 05/15/2021] [Indexed: 02/07/2023]
Abstract
Blood clots play a diametric role in our bodies as they are both vital as a wound sealant, as well as the source for many devastating diseases. In blood clots' physiological and pathological roles, their mechanics play a critical part. These mechanics are non-trivial owing to blood clots' complex nonlinear, viscoelastic behavior. Casting this behavior into mathematical form is a fundamental step toward a better basic scientific understanding of blood clots, as well as toward diagnostic and prognostic computational models. Here, we identify a hyper-viscoelastic damage model that we fit to original data on the nonlinear, viscoelastic behavior of blood clots. Our model combines the classic Ogden hyperelastic constitutive law, a finite viscoelastic model for large deformations, and a non-local, gradient-enhanced damage formulation. By fitting our model to cyclic tensile test data and extension-to-failure data, we inform the model's nine unknown material parameters. We demonstrate the predictability of our model by validating it against unseen cyclic tensile test and stress-relaxation data. Our original data, model formulation, and the identified constitutive parameters of this model are openly available for others to use, which will aid in developing accurate, quantitative simulations of blood clot mechanics.
Collapse
Affiliation(s)
- Manuel K Rausch
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA.
| | - Gabriella P Sugerman
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Sotirios Kakaletsis
- Department of Aerospace Engineering and Engineering Mechanics, University of Texas at Austin, 2617 Wichita Street, Austin, TX, 78712, USA
| | - Berkin Dortdivanlioglu
- Department of Civil, Architectural and Environmental Engineering, The University of Texas at Austin, Austin, TX, 78712, USA.,Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX, 78712, USA
| |
Collapse
|
80
|
Saghati S, Rahbarghazi R, Baradar Khoshfetrat A, Moharamzadeh K, Tayefi Nasrabadi H, Roshangar L. Phenolated alginate-collagen hydrogel induced chondrogenic capacity of human amniotic mesenchymal stem cells. J Biomater Appl 2021; 36:789-802. [PMID: 34074175 DOI: 10.1177/08853282211021692] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Horseradish peroxidase (HRP)-catalyzed hydrogels are considered to be an important platform for tissue engineering applications. In this study, we investigated the chondrogenic capacity of phenolated (1.2%) alginate-(0.5%) collagen hydrogel on human amniotic mesenchymal stem cells after 21 days. Using NMR, FTIR analyses, and SEM imaging, we studied the phenolation and structure of alginate-collagen hydrogel. For physicochemical evaluations, gelation time, mechanical properties, swelling, and degradation rate were assessed. The survival rate was monitored using the MTT assay and DAPI staining. Western blotting was performed to measure the chondrogenic differentiation of cells. NMR showed successful phenolation of the alginate-collagen hydrogel. FTIR exhibited the interaction between the functional groups of collagen with phenolated alginate. SEM showed the existence of collagen microfibrils in the alginate-collagen hydrogel. Compared to phenolated alginate, the addition of collagen increased hydrogel elasticity by 10%. Both swelling rate and biodegradability were reduced in the presence of collagen. We noted an increased survival rate in phenolated alginate-collagen compared to the control cells (p < 0.05). Western blotting revealed the increase of chondrocyte-associated proteins such as SOX9 and COL2A1 in phenolated-alginate-collagen hydrogels after 21 days. These data showed that phenolated alginate-collagen hydrogel is an appropriate 3 D substrate to induce chondrogenic capacity of human mesenchymal stem cells.
Collapse
Affiliation(s)
- Sepideh Saghati
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Baradar Khoshfetrat
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Hamid Tayefi Nasrabadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Roshangar
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
81
|
Collagen denaturation in the infarcted myocardium involves temporally distinct effects of MT1-MMP-dependent proteolysis and mechanical tension. Matrix Biol 2021; 99:18-42. [PMID: 34048934 DOI: 10.1016/j.matbio.2021.05.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 05/14/2021] [Accepted: 05/19/2021] [Indexed: 12/13/2022]
Abstract
Tissue injury results in profound alterations in the collagen network, associated with unfolding of the collagen triple helix, proteolytic degradation and generation of fragments. In the infarcted myocardium, changes in the collagen network are critically involved in the pathogenesis of left ventricular rupture, adverse remodeling and chronic dysfunction. We hypothesized that myocardial infarction is associated with temporally and spatially restricted patterns of collagen denaturation that may reflect distinct molecular mechanisms of collagen unfolding. We used a mouse model of non-reperfused myocardial infarction, and in vitro assays in fibroblast-populated collagen lattices. In healing infarcts, labeling with collagen hybridizing peptide (CHP) revealed two distinct patterns of collagen denaturation. During the inflammatory and proliferative phases of infarct healing, collagen denaturation was pericellular, localized in close proximity to macrophages and myofibroblasts. qPCR array analysis of genes associated with matrix remodeling showed that Membrane Type 1-Matrix Metalloproteinase (MT1-MMP) is markedly upregulated in infarct macrophages and fibroblasts, suggesting its involvement in pericellular collagen denaturation. In vitro, MT1-MMP-mediated pericellular collagen denaturation is involved in cardiac fibroblast migration. The effects of MT1-MMP on collagen denaturation and fibroblast migration involve the catalytic site, and require hemopexin domain-mediated actions. In contrast, during the maturation phase of infarct healing, extensive collagen denaturation was noted in the hypocellular infarct, in the infarct border zone and in the mitral valve annulus, in the absence of MT1-MMP. In vitro, mechanical tension in attached collagen lattices was sufficient to induce peripheral collagen denaturation. Our study suggests that in healing infarcts, early pericellular collagen denaturation may be important for migration of macrophages and reparative myofibroblasts in the infarct. Extensive denaturation of collagen fibers is noted in mature scars, likely reflecting mechanical tension. Chronic collagen denaturation may increase susceptibility of the matrix to proteolysis, thus contributing to progressive cardiac dilation and post-infarction heart failure.
Collapse
|
82
|
Kobayashi M, Ohara M, Hashimoto Y, Nakamura N, Fujisato T, Kimura T, Kishida A. Effect of luminal surface structure of decellularized aorta on thrombus formation and cell behavior. PLoS One 2021; 16:e0246221. [PMID: 33999919 PMCID: PMC8128234 DOI: 10.1371/journal.pone.0246221] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 05/03/2021] [Indexed: 11/18/2022] Open
Abstract
Due to an increasing number of cardiovascular diseases, artificial heart valves and blood vessels have been developed. Although cardiovascular applications using decellularized tissue have been studied, the mechanisms of their functionality remain unknown. To determine the important factors for preparing decellularized cardiovascular prostheses that show good in vivo performance, the effects of the luminal surface structure of the decellularized aorta on thrombus formation and cell behavior were investigated. Various luminal surface structures of a decellularized aorta were prepared by heating, drying, and peeling. The luminal surface structure and collagen denaturation were evaluated by immunohistological staining, collagen hybridizing peptide (CHP) staining, and scanning electron microscopy (SEM) analysis. To evaluate the effects of luminal surface structure of decellularized aorta on thrombus formation and cell behavior, blood clotting tests and recellularization of endothelial cells and smooth muscle cells were performed. The results of the blood clotting test showed that the closer the luminal surface structure is to the native aorta, the higher the anti-coagulant property. The results of the cell seeding test suggest that vascular cells recognize the luminal surface structure and regulate adhesion, proliferation, and functional expression accordingly. These results provide important factors for preparing decellularized cardiovascular prostheses and will lead to future developments in decellularized cardiovascular applications.
Collapse
Affiliation(s)
- Mako Kobayashi
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Chiyoda-ku, Tokyo, Japan
| | - Masako Ohara
- Department of Bioscience and Engineering, Shibaura Institute of Technology, Minuma-ku, Saitama-shi, Saitama, Japan
| | - Yoshihide Hashimoto
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Chiyoda-ku, Tokyo, Japan
| | - Naoko Nakamura
- Department of Bioscience and Engineering, Shibaura Institute of Technology, Minuma-ku, Saitama-shi, Saitama, Japan
| | - Toshiya Fujisato
- Department of Biomedical Engineering, Osaka Institute of Technology, Asahi-ku, Osaka, Japan
| | - Tsuyoshi Kimura
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Chiyoda-ku, Tokyo, Japan
| | - Akio Kishida
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Chiyoda-ku, Tokyo, Japan
| |
Collapse
|
83
|
Computational model of damage-induced growth in soft biological tissues considering the mechanobiology of healing. Biomech Model Mechanobiol 2021; 20:1297-1315. [PMID: 33768359 PMCID: PMC8298377 DOI: 10.1007/s10237-021-01445-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 03/04/2021] [Indexed: 11/01/2022]
Abstract
Healing in soft biological tissues is a chain of events on different time and length scales. This work presents a computational framework to capture and couple important mechanical, chemical and biological aspects of healing. A molecular-level damage in collagen, i.e., the interstrand delamination, is addressed as source of plastic deformation in tissues. This mechanism initiates a biochemical response and starts the chain of healing. In particular, damage is considered to be the stimulus for the production of matrix metalloproteinases and growth factors which in turn, respectively, degrade and produce collagen. Due to collagen turnover, the volume of the tissue changes, which can result either in normal or pathological healing. To capture the mechanisms on continuum scale, the deformation gradient is multiplicatively decomposed in inelastic and elastic deformation gradients. A recently proposed elasto-plastic formulation is, through a biochemical model, coupled with a growth and remodeling description based on homogenized constrained mixtures. After the discussion of the biological species response to the damage stimulus, the framework is implemented in a mixed nonlinear finite element formulation and a biaxial tension and an indentation tests are conducted on a prestretched flat tissue sample. The results illustrate that the model is able to describe the evolutions of growth factors and matrix metalloproteinases following damage and the subsequent growth and remodeling in the respect of equilibrium. The interplay between mechanical and chemo-biological events occurring during healing is captured, proving that the framework is a suitable basis for more detailed simulations of damage-induced tissue response.
Collapse
|
84
|
Wang WM, Yu CH, Chang JY, Chen TH, Chen YC, Sun YT, Wang SH, Jao SC, Cheng RP. Insertion of Pro-Hyp-Gly provides 2 kcal mol -1 stability but attenuates the specific assembly of ABC heterotrimeric collagen triple helices. Org Biomol Chem 2021; 19:1860-1866. [PMID: 33565556 DOI: 10.1039/d0ob02190c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Collagen is a major structural component of the extracellular matrix and connective tissue. The key structural feature of collagen is the collagen triple helix, with a Xaa-Yaa-Gly (glycine) repeating pattern. The most frequently occurring triplet is Pro (proline)-Hyp (hydroxyproline)-Gly. The reversible thermal folding and unfolding of a series of heterotrimeric collagen triple helices with varying number of Pro-Hyp-Gly triplets were monitored by circular dichroism spectroscopy to determine the unfolding thermodynamic parameters Tm (midpoint transition temperature), ΔHTm (unfolding enthalpy), and ΔGunfold (unfolding free energy). The Tm and ΔGunfold of the heterotrimeric collagen triple helices increased with increasing number of Pro-Hyp-Gly triplets. The ΔGunfold increased by 2.0 ± 0.2 kcal mol-1 upon inserting one Pro-Hyp-Gly triplet into all three chains. The Tm difference between the most stable ABC combination and the second most stable BCC combination decreased with increasing number of Pro-Hyp-Gly triplets, even though the ΔGunfold difference remained the same. These results should be useful for tuning the stability of collagen triple helical peptides for hydrogel formation, recognition of denatured collagen triple helices as diagnostics and therapeutics, and targeted drug delivery.
Collapse
Affiliation(s)
- Wei-Ming Wang
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan.
| | - Chen-Hsu Yu
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan.
| | - Jing-Yuan Chang
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan.
| | - Ting-Hsuan Chen
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan.
| | - Yan-Chen Chen
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan.
| | - Yi-Ting Sun
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan.
| | - Szu-Huan Wang
- Department of Academic Affairs and Instrument Service, and Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Shu-Chuan Jao
- Department of Academic Affairs and Instrument Service, and Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Richard P Cheng
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan.
| |
Collapse
|
85
|
Brunet J, Pierrat B, Badel P. Review of Current Advances in the Mechanical Description and Quantification of Aortic Dissection Mechanisms. IEEE Rev Biomed Eng 2021; 14:240-255. [PMID: 31905148 DOI: 10.1109/rbme.2019.2950140] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Aortic dissection is a life-threatening event associated with a very poor outcome. A number of complex phenomena are involved in the initiation and propagation of the disease. Advances in the comprehension of the mechanisms leading to dissection have been made these last decades, thanks to improvements in imaging and experimental techniques. However, the micro-mechanics involved in triggering such rupture events remains poorly described and understood. It constitutes the primary focus of the present review. Towards the goal of detailing the dissection phenomenon, different experimental and modeling methods were used to investigate aortic dissection, and to understand the underlying phenomena involved. In the last ten years, research has tended to focus on the influence of microstructure on initiation and propagation of the dissection, leading to a number of multiscale models being developed. This review brings together all these materials in an attempt to identify main advances and remaining questions.
Collapse
|
86
|
Xu Y, Kirchner M. Collagen Mimetic Peptides. Bioengineering (Basel) 2021; 8:5. [PMID: 33466358 PMCID: PMC7824840 DOI: 10.3390/bioengineering8010005] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/24/2020] [Accepted: 12/31/2020] [Indexed: 12/20/2022] Open
Abstract
Since their first synthesis in the late 1960s, collagen mimetic peptides (CMPs) have been used as a molecular tool to study collagen, and as an approach to develop novel collagen mimetic biomaterials. Collagen, a major extracellular matrix (ECM) protein, plays vital roles in many physiological and pathogenic processes. Applications of CMPs have advanced our understanding of the structure and molecular properties of a collagen triple helix-the building block of collagen-and the interactions of collagen with important molecular ligands. The accumulating knowledge is also paving the way for developing novel CMPs for biomedical applications. Indeed, for the past 50 years, CMP research has been a fast-growing, far-reaching interdisciplinary field. The major development and achievement of CMPs were documented in a few detailed reviews around 2010. Here, we provided a brief overview of what we have learned about CMPs-their potential and their limitations. We focused on more recent developments in producing heterotrimeric CMPs, and CMPs that can form collagen-like higher order molecular assemblies. We also expanded the traditional view of CMPs to include larger designed peptides produced using recombinant systems. Studies using recombinant peptides have provided new insights on collagens and promoted progress in the development of collagen mimetic fibrillar self-assemblies.
Collapse
Affiliation(s)
- Yujia Xu
- Department of Chemistry, Hunter College of the City University of New York, 695 Park Ave., New York, NY 10065, USA;
| | | |
Collapse
|
87
|
Siadat SM, Zamboulis DE, Thorpe CT, Ruberti JW, Connizzo BK. Tendon Extracellular Matrix Assembly, Maintenance and Dysregulation Throughout Life. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1348:45-103. [PMID: 34807415 DOI: 10.1007/978-3-030-80614-9_3] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
In his Lissner Award medal lecture in 2000, Stephen Cowin asked the question: "How is a tissue built?" It is not a new question, but it remains as relevant today as it did when it was asked 20 years ago. In fact, research on the organization and development of tissue structure has been a primary focus of tendon and ligament research for over two centuries. The tendon extracellular matrix (ECM) is critical to overall tissue function; it gives the tissue its unique mechanical properties, exhibiting complex non-linear responses, viscoelasticity and flow mechanisms, excellent energy storage and fatigue resistance. This matrix also creates a unique microenvironment for resident cells, allowing cells to maintain their phenotype and translate mechanical and chemical signals into biological responses. Importantly, this architecture is constantly remodeled by local cell populations in response to changing biochemical (systemic and local disease or injury) and mechanical (exercise, disuse, and overuse) stimuli. Here, we review the current understanding of matrix remodeling throughout life, focusing on formation and assembly during the postnatal period, maintenance and homeostasis during adulthood, and changes to homeostasis in natural aging. We also discuss advances in model systems and novel tools for studying collagen and non-collagenous matrix remodeling throughout life, and finally conclude by identifying key questions that have yet to be answered.
Collapse
Affiliation(s)
| | - Danae E Zamboulis
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Chavaunne T Thorpe
- Comparative Biomedical Sciences, The Royal Veterinary College, University of London, London, UK
| | - Jeffrey W Ruberti
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Brianne K Connizzo
- Department of Biomedical Engineering, Boston University, Boston, MA, USA.
| |
Collapse
|
88
|
Ghosh AK. Pharmacological activation of PPAR-γ: a potential therapy for skin fibrosis. Int J Dermatol 2020; 60:376-383. [PMID: 33377189 DOI: 10.1111/ijd.15388] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 11/23/2020] [Accepted: 12/01/2020] [Indexed: 12/28/2022]
Abstract
Skin fibrosis caused by excessive collagen synthesis and deposition in the dermis affects the quality of daily life of hundreds of thousands of people around the world. The skin quality, including its smoothness in young age and wrinkly during the aging process, depends largely on the levels of extracellular matrix proteins such as collagen in skin. As physiological levels of collagen are desirable for skin homeostasis, beauty, and its flexibility, too much collagen deposition in the skin is associated with tight hard skin, loss of adipose layer, and flexibility, the pathological manifestations of skin fibrosis in fibrotic diseases such as scleroderma. To understand the molecular basis of skin fibrosis and in search of its therapy, different cellular, molecular, epigenetic, and preclinical studies have been undertaken to control abnormal excessive synthesis and accumulation of matrix protein collagen. Over the last two decades, numerous phase 1 through 3 clinical trials have been conducted to test the safety and efficacy of a wide variety of compounds in amelioration of skin fibrosis and other pathologies in scleroderma, yet, no effective therapy for skin fibrosis is available. This article solely focuses on the role of a nuclear receptor and transcription factor, peroxisome proliferator-activated receptor-gamma (PPAR-γ), as an anti-skin fibrotic driving force and the potential therapeutic efficacies of PPAR-γ-specific ligands/agonists including antidiabetic drugs and other natural or semi-synthetic compounds derived from cannabis in amelioration of skin fibrosis in scleroderma. The underlying molecular basis of agonist-activated PPAR-γ-mediated suppression of profibrogenic signaling and skin fibrogenesis is also highlighted.
Collapse
Affiliation(s)
- Asish K Ghosh
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
89
|
Lin AH, Allan AN, Zitnay JL, Kessler JL, Yu SM, Weiss JA. Collagen denaturation is initiated upon tissue yield in both positional and energy-storing tendons. Acta Biomater 2020; 118:153-160. [PMID: 33035697 DOI: 10.1016/j.actbio.2020.09.056] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 09/04/2020] [Accepted: 09/30/2020] [Indexed: 12/12/2022]
Abstract
Tendons are collagenous soft tissues that transmit loads between muscles and bones. Depending on their anatomical function, tendons are classified as positional or energy-storing with differing biomechanical and biochemical properties. We recently demonstrated that during monotonic stretch of positional tendons, permanent denatured collagen begins accumulating upon departing the linear region of the stress-strain curve. However, it is unknown if this observation is true during mechanical overload of other types of tendons. Therefore, the purpose of this study was to investigate the onset of collagen denaturation relative to applied strain, and whether it differs between the two tendon types. Rat tail tendon (RTT) fascicles and rat flexor digitorum longus (FDL) tendons represented positional and energy-storing tendons, respectively. The samples were stretched to incremental levels of strain, then stained with fluorescently labeled collagen hybridizing peptides (CHPs); the CHP fluorescence was measured to quantify denatured collagen. Denatured collagen in both positional and energy-storing tendons began to increase at the yield strain, upon leaving the linear region of the stress-strain curve as the sample started to permanently deform. Despite significant differences between the two tendon types, it appears that collagen denaturation is initiated at tissue yield during monotonic stretch, and the fundamental mechanism of failure is the same for the two types of tendons. At tissue failure, positional tendons had double the percentage of denatured collagen compared to energy-storing tendons, with no difference between 0% control groups. These results help to elucidate the etiology of subfailure injury and rupture in functionally distinct tendons.
Collapse
Affiliation(s)
- Allen H Lin
- Department of Biomedical Engineering, University of Utah, United States; Scientific Computing and Imaging Institute, University of Utah, United States
| | - Alexandra N Allan
- Department of Biomedical Engineering, University of Utah, United States; Scientific Computing and Imaging Institute, University of Utah, United States
| | - Jared L Zitnay
- Department of Biomedical Engineering, University of Utah, United States; Scientific Computing and Imaging Institute, University of Utah, United States
| | - Julian L Kessler
- Department of Biomedical Engineering, University of Utah, United States
| | - S Michael Yu
- Department of Biomedical Engineering, University of Utah, United States; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, United States
| | - Jeffrey A Weiss
- Department of Biomedical Engineering, University of Utah, United States; Scientific Computing and Imaging Institute, University of Utah, United States; Department of Orthopaedics, University of Utah, United States.
| |
Collapse
|
90
|
Hoy RC, D'Erminio DN, Krishnamoorthy D, Natelson DM, Laudier DM, Illien‐Jünger S, Iatridis JC. Advanced glycation end products cause RAGE-dependent annulus fibrosus collagen disruption and loss identified using in situ second harmonic generation imaging in mice intervertebral disk in vivo and in organ culture models. JOR Spine 2020; 3:e1126. [PMID: 33392460 PMCID: PMC7770195 DOI: 10.1002/jsp2.1126] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 08/12/2020] [Accepted: 09/09/2020] [Indexed: 12/13/2022] Open
Abstract
Aging and diabetes are associated with increased low-back pain and intervertebral disk (IVD) degeneration yet causal mechanisms remain uncertain. Advanced glycation end products (AGEs), which accumulate in IVDs from aging and are implicated in diabetes-related disorders, alter collagen and induce proinflammatory conditions. A need exists for methods that assess IVD collagen quality and degradation in order to better characterize specific structural changes in IVDs due to AGE accumulation and to identify roles for the receptor for AGEs (RAGE). We used multiphoton microscopy with second harmonic generation (SHG), collagen-hybridizing peptide (CHP), and image analysis methods to characterize effects of AGEs and RAGE on collagen quality and quantity in IVD annulus fibrosus (AF). First, we used SHG imaging on thin sections with an in vivo dietary mouse model and determined that high-AGE (H-AGE) diets increased AF fibril disruption and collagen degradation resulting in decreased total collagen content, suggesting an early degenerative cascade. Next, we used in situ SHG imaging with an ex vivo IVD organ culture model of AGE challenge on wild type and RAGE-knockout (RAGE-KO) mice and determined that early degenerative changes to collagen quality and degradation were RAGE dependent. We conclude that AGE accumulation leads to RAGE-dependent collagen disruption in the AF and can initiate molecular and tissue level collagen disruption. Furthermore, SHG and CHP analyzes were sensitive to collagenous alterations at multiple hierarchical levels due to AGE and may be useful in identifying additional contributors to collagen damage in IVD degeneration processes.
Collapse
Affiliation(s)
- Robert C. Hoy
- Leni & Peter W. May Department of OrthopaedicsIcahn School of Medicine at Mount SinaiNYUnited StatesUSA
| | - Danielle N. D'Erminio
- Leni & Peter W. May Department of OrthopaedicsIcahn School of Medicine at Mount SinaiNYUnited StatesUSA
| | - Divya Krishnamoorthy
- Leni & Peter W. May Department of OrthopaedicsIcahn School of Medicine at Mount SinaiNYUnited StatesUSA
| | - Devorah M. Natelson
- Leni & Peter W. May Department of OrthopaedicsIcahn School of Medicine at Mount SinaiNYUnited StatesUSA
| | - Damien M. Laudier
- Leni & Peter W. May Department of OrthopaedicsIcahn School of Medicine at Mount SinaiNYUnited StatesUSA
| | | | - James C. Iatridis
- Leni & Peter W. May Department of OrthopaedicsIcahn School of Medicine at Mount SinaiNYUnited StatesUSA
| |
Collapse
|
91
|
Nesbitt DQ, Siegel DN, Nelson SJ, Lujan TJ. Effect of age on the failure properties of human meniscus: High-speed strain mapping of tissue tears. J Biomech 2020; 115:110126. [PMID: 33359919 DOI: 10.1016/j.jbiomech.2020.110126] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/04/2020] [Indexed: 12/30/2022]
Abstract
The knee meniscus is a soft fibrous tissue with a high incidence of injury in older populations. The objective of this study was to determine the effect of age on the failure behavior of human knee meniscus when applying uniaxial tensile loads parallel or perpendicular to the primary circumferential fiber orientation. Two age groups were tested: under 40 and over 65 years old. We paired high-speed video with digital image correlation to quantify for the first time the planar strains occurring in the tear region at precise time points, including at ultimate tensile stress, when the tissue begins losing load-bearing capacity. On average, older meniscus specimens loaded parallel to the fiber axis had approximately one-third less ultimate tensile strain and absorbed 60% less energy to failure within the tear region than younger specimens (p < 0.05). Older specimens also had significantly reduced strength and material toughness when loaded perpendicular to the fibers (p < 0.05). These age-related changes indicate a loss of collagen fiber extensibility and weakening of the non-fibrous matrix with age. In addition, we found that when loaded perpendicular to the circumferential fibers, tears propagated near the planes of maximum tensile stress and strain. Whereas when loaded parallel to the circumferential fibers, tears propagated oblique to the loading axis, closer to the planes of maximum shear stress and strain. Our experimental results can assist the selection of valid failure criteria for meniscus, and provide insight into the effect of age on the failure mechanisms of soft fibrous tissue.
Collapse
Affiliation(s)
- Derek Q Nesbitt
- Boise State University, Mechanical and Biomedical Engineering, 1910 University Drive, Boise, ID 83725-2085, United States
| | - Danielle N Siegel
- Boise State University, Mechanical and Biomedical Engineering, 1910 University Drive, Boise, ID 83725-2085, United States
| | - Sean J Nelson
- Boise State University, Mechanical and Biomedical Engineering, 1910 University Drive, Boise, ID 83725-2085, United States
| | - Trevor J Lujan
- Boise State University, Mechanical and Biomedical Engineering, 1910 University Drive, Boise, ID 83725-2085, United States.
| |
Collapse
|
92
|
Qin J, Sloppy JD, Kiick KL. Fine structural tuning of the assembly of ECM peptide conjugates via slight sequence modifications. SCIENCE ADVANCES 2020; 6:eabd3033. [PMID: 33028534 PMCID: PMC7541060 DOI: 10.1126/sciadv.abd3033] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 08/26/2020] [Indexed: 05/07/2023]
Abstract
The self-assembly of nanostructures from conjugates of elastin-like peptides and collagen-like peptides (ELP-CLP) has been studied as means to produce thermoresponsive, collagen-binding drug delivery vehicles. Motivated by our previous work in which ELP-CLP conjugates successfully self-assembled into vesicles and platelet-like nanostructures, here, we extend our library of ELP-CLP bioconjugates to a series of tryptophan/phenylalanine-containing ELPs and GPO-based CLPs [W2F x -b-(GPO) y ] with various domain lengths to determine the impact of these modifications on the thermoresponsiveness and morphology. The lower transition temperature of the conjugates with longer ELP or CLP domains enables the formation of well-defined nanoparticles near physiological temperature. Moreover, the morphological transition from vesicles to platelet-like nanostructures occurred when the ratio of the lengths of ELP/CLP decreased. Given the previously demonstrated ability of many ELP-CLP bioconjugates to bind to both hydrophobic drugs and collagen-containing materials, our results suggest new opportunities for designing specific thermoresponsive nanostructures for targeted biological applications.
Collapse
Affiliation(s)
- Jingya Qin
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA
| | - Jennifer D Sloppy
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA
| | - Kristi L Kiick
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA.
- Delaware Biotechnology Institute, Newark, DE 19711, USA
| |
Collapse
|
93
|
Khajuria DK, Soliman M, Elfar JC, Lewis GS, Abraham T, Kamal F, Elbarbary RA. Aberrant structure of fibrillar collagen and elevated levels of advanced glycation end products typify delayed fracture healing in the diet-induced obesity mouse model. Bone 2020; 137:115436. [PMID: 32439570 PMCID: PMC7938873 DOI: 10.1016/j.bone.2020.115436] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/17/2020] [Accepted: 05/14/2020] [Indexed: 12/16/2022]
Abstract
Impaired fracture healing in patients with obesity-associated type 2 diabetes (T2D) is a significant unmet clinical problem that affects millions of people worldwide. However, the underlying causes are poorly understood. Additionally, limited clinical information is available on how pre-diabetic hyperglycemia in obese individuals impacts bone healing. Here, we use the diet-induced obesity (DIO) mouse (C57BL/6J) model to study the impact of obesity-associated pre-diabetic hyperglycemia on bone healing and fibrillar collagen organization as healing proceeds from one phase to another. We show that DIO mice exhibit defective healing characterized by reduced bone mineral density, bone volume, and bone volume density. Differences in the healing pattern between lean and DIO mice occur early in the healing process as evidenced by faster resorption of the fibrocartilaginous callus in DIO mice. However, the major differences between lean and DIO mice occur during the later phases of endochondral ossification and bone remodeling. Comprehensive analyses of fibrillar collagen microstructure and expression pattern during these phases, using a set of complementary techniques that include histomorphometry, immunofluorescence staining, and second harmonic generation microscopy, demonstrate significant defects in DIO mice. Defects include strikingly sparse and disorganized collagen fibers, as well as pathological accumulation of unfolded collagen triple helices. We also demonstrate that DIO-associated changes in fibrillar collagen structure are attributable, at least in part, to the accumulation of advanced glycation end products, which increase the collagen-fiber crosslink density. These major changes impair fibrillar collagens functions, culminating in defective callus mineralization, remodeling, and strength. Our data extend the understanding of mechanisms by which obesity and its associated hyperglycemia impair fracture healing and underline defective fibrillar collagen microstructure as a novel and important contributor.
Collapse
Affiliation(s)
- Deepak Kumar Khajuria
- Department of Orthopaedics and Rehabilitation, The Pennsylvania State University College of Medicine, Hershey, PA, USA; Center for Orthopaedic Research and Translational Science (CORTS), The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Marwa Soliman
- Department of Orthopaedics and Rehabilitation, The Pennsylvania State University College of Medicine, Hershey, PA, USA; Center for Orthopaedic Research and Translational Science (CORTS), The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - John C Elfar
- Department of Orthopaedics and Rehabilitation, The Pennsylvania State University College of Medicine, Hershey, PA, USA; Center for Orthopaedic Research and Translational Science (CORTS), The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Gregory S Lewis
- Department of Orthopaedics and Rehabilitation, The Pennsylvania State University College of Medicine, Hershey, PA, USA; Center for Orthopaedic Research and Translational Science (CORTS), The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Thomas Abraham
- Microscopy Imaging Facility, The Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Neural and Behavioural Sciences, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Fadia Kamal
- Department of Orthopaedics and Rehabilitation, The Pennsylvania State University College of Medicine, Hershey, PA, USA; Center for Orthopaedic Research and Translational Science (CORTS), The Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Reyad A Elbarbary
- Department of Orthopaedics and Rehabilitation, The Pennsylvania State University College of Medicine, Hershey, PA, USA; Center for Orthopaedic Research and Translational Science (CORTS), The Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, USA; Center for RNA Molecular Biology, Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
94
|
Zitnay JL, Jung GS, Lin AH, Qin Z, Li Y, Yu SM, Buehler MJ, Weiss JA. Accumulation of collagen molecular unfolding is the mechanism of cyclic fatigue damage and failure in collagenous tissues. SCIENCE ADVANCES 2020; 6:eaba2795. [PMID: 32923623 PMCID: PMC7455178 DOI: 10.1126/sciadv.aba2795] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 07/14/2020] [Indexed: 05/04/2023]
Abstract
Overuse injuries to dense collagenous tissues are common, but their etiology is poorly understood. The predominant hypothesis that micro-damage accumulation exceeds the rate of biological repair is missing a mechanistic explanation. Here, we used collagen hybridizing peptides to measure collagen molecular damage during tendon cyclic fatigue loading and computational simulations to identify potential explanations for our findings. Our results revealed that triple-helical collagen denaturation accumulates with increasing cycles of fatigue loading, and damage is correlated with creep strain independent of the cyclic strain rate. Finite-element simulations demonstrated that biphasic fluid flow is a possible fascicle-level mechanism to explain the rate dependence of the number of cycles and time to failure. Molecular dynamics simulations demonstrated that triple-helical unfolding is rate dependent, revealing rate-dependent mechanisms at multiple length scales in the tissue. The accumulation of collagen molecular denaturation during cyclic loading provides a long-sought "micro-damage" mechanism for the development of overuse injuries.
Collapse
Affiliation(s)
- Jared L. Zitnay
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Gang Seob Jung
- Laboratory for Atomistic and Molecular Mechanics, Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Allen H. Lin
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Zhao Qin
- Laboratory for Atomistic and Molecular Mechanics, Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yang Li
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
| | - S. Michael Yu
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84132, USA
| | - Markus J. Buehler
- Laboratory for Atomistic and Molecular Mechanics, Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jeffrey A. Weiss
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT 84112, USA
- Department of Orthopaedics and School of Computing, University of Utah, Salt Lake City, UT 84112, USA
- Corresponding author.
| |
Collapse
|
95
|
Norris EG, Dalecki D, Hocking DC. Using Acoustic Fields to Fabricate ECM-Based Biomaterials for Regenerative Medicine Applications. RECENT PROGRESS IN MATERIALS 2020; 2:1-24. [PMID: 33604591 PMCID: PMC7889011 DOI: 10.21926/rpm.2003018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Ultrasound is emerging as a promising tool for both characterizing and fabricating engineered biomaterials. Ultrasound-based technologies offer a diverse toolbox with outstanding capacity for optimization and customization within a variety of therapeutic contexts, including improved extracellular matrix-based materials for regenerative medicine applications. Non-invasive ultrasound fabrication tools include the use of thermal and mechanical effects of acoustic waves to modify the structure and function of extracellular matrix scaffolds both directly, and indirectly via biochemical and cellular mediators. Materials derived from components of native extracellular matrix are an essential component of engineered biomaterials designed to stimulate cell and tissue functions and repair or replace injured tissues. Thus, continued investigations into biological and acoustic mechanisms by which ultrasound can be used to manipulate extracellular matrix components within three-dimensional hydrogels hold much potential to enable the production of improved biomaterials for clinical and research applications.
Collapse
Affiliation(s)
- Emma G Norris
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York, USA
| | - Diane Dalecki
- Department of Biomedical Engineering, University of Rochester, Rochester, New York, USA
| | - Denise C Hocking
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York, USA
- Department of Biomedical Engineering, University of Rochester, Rochester, New York, USA
| |
Collapse
|
96
|
Lim N, Wen C, Vincent T. Molecular and structural imaging in surgically induced murine osteoarthritis. Osteoarthritis Cartilage 2020; 28:874-884. [PMID: 32305526 PMCID: PMC7327515 DOI: 10.1016/j.joca.2020.03.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 03/15/2020] [Accepted: 03/23/2020] [Indexed: 02/02/2023]
Abstract
Preclinical imaging in osteoarthritis is a rapidly growing area with three principal objectives: to provide rapid, sensitive tools to monitor the course of experimental OA longitudinally; to describe the temporal relationship between tissue-specific pathologies over the course of disease; and to use molecular probes to measure disease activity in vivo. Research in this area can be broadly divided into those techniques that monitor structural changes in tissues (microCT, microMRI, ultrasound) and those that detect molecular disease activity (positron emission tomography (PET), optical and optoacoustic imaging). The former techniques have largely evolved from experience in human joint imaging and have been refined for small animal use. Some of the latter tools, such as optical imaging, have been developed in preclinical models and may have translational benefit in the future for patient stratification and for monitoring disease progression and response to treatment. In this narrative review we describe these methodologies and discuss the benefits to animal research, understanding OA pathogenesis, and in the development of human biomarkers.
Collapse
Affiliation(s)
- N.H. Lim
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, University of Oxford, UK,Address correspondence and reprint requests to: N.H. Lim, Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, University of Oxford, UK.
| | - C. Wen
- Department of Biomedical Engineering, Hong Kong Polytechnic University, Hong Kong
| | - T.L. Vincent
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, University of Oxford, UK
| |
Collapse
|
97
|
Rega R, Mugnano M, Oleandro E, Tkachenko V, del Giudice D, Bagnato G, Ferraro P, Grilli S, Gangemi S. Detecting Collagen Molecules at Picogram Level through Electric Field-Induced Accumulation. SENSORS (BASEL, SWITZERLAND) 2020; 20:E3567. [PMID: 32599740 PMCID: PMC7349194 DOI: 10.3390/s20123567] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/19/2020] [Accepted: 06/22/2020] [Indexed: 12/24/2022]
Abstract
The demand for sensors capable of measuring low-abundant collagen in human fluids has highly increased in recent years. Indeed, collagen is expected to be a biomarker for chronic diseases and could monitor their progression. Here we show detection of highly diluted samples of collagen at picogram level thanks to an innovative pyro-electrohydrodynamic jet (p-jet) system. Through the intense electric fields generated by the pyroelectric effect in a ferroelectric crystal, the collagen solution was concentrated on a small area of a slide that was appropriately functionalized to bind proteins. The collagen molecules were labeled by an appropriate fluorophore to show how the number of tiny droplets influences the limit of detection of the technique. The results show that the p-jet is extremely promising for overcoming the current detection limits of collagen-based products in human fluids, performing 10 times better than the enzyme-linked immunosorbent assay (ELISA) and thus paving the way for the early diagnosis of related chronic diseases.
Collapse
Affiliation(s)
- Romina Rega
- Department of Physical Science and Technology of Matter, Institute of Applied Sciences and Intelligent Systems (ISASI), National Research Council of Italy (CNR), 80078 Pozzuoli (NA), Italy; (M.M.); (E.O.); (V.T.); (D.d.G.); (P.F.); (S.G.)
| | - Martina Mugnano
- Department of Physical Science and Technology of Matter, Institute of Applied Sciences and Intelligent Systems (ISASI), National Research Council of Italy (CNR), 80078 Pozzuoli (NA), Italy; (M.M.); (E.O.); (V.T.); (D.d.G.); (P.F.); (S.G.)
| | - Emilia Oleandro
- Department of Physical Science and Technology of Matter, Institute of Applied Sciences and Intelligent Systems (ISASI), National Research Council of Italy (CNR), 80078 Pozzuoli (NA), Italy; (M.M.); (E.O.); (V.T.); (D.d.G.); (P.F.); (S.G.)
- Department of Mathematics and Physics, University of Campania, 81100 Caserta, Italy
| | - Volodymyr Tkachenko
- Department of Physical Science and Technology of Matter, Institute of Applied Sciences and Intelligent Systems (ISASI), National Research Council of Italy (CNR), 80078 Pozzuoli (NA), Italy; (M.M.); (E.O.); (V.T.); (D.d.G.); (P.F.); (S.G.)
| | - Danila del Giudice
- Department of Physical Science and Technology of Matter, Institute of Applied Sciences and Intelligent Systems (ISASI), National Research Council of Italy (CNR), 80078 Pozzuoli (NA), Italy; (M.M.); (E.O.); (V.T.); (D.d.G.); (P.F.); (S.G.)
- Department of Mathematics and Physics, University of Campania, 81100 Caserta, Italy
| | - Gianluca Bagnato
- Division of Pneumology, Papardo Hospital, Contrada Papardo, 98122 Messina, Italy;
| | - Pietro Ferraro
- Department of Physical Science and Technology of Matter, Institute of Applied Sciences and Intelligent Systems (ISASI), National Research Council of Italy (CNR), 80078 Pozzuoli (NA), Italy; (M.M.); (E.O.); (V.T.); (D.d.G.); (P.F.); (S.G.)
| | - Simonetta Grilli
- Department of Physical Science and Technology of Matter, Institute of Applied Sciences and Intelligent Systems (ISASI), National Research Council of Italy (CNR), 80078 Pozzuoli (NA), Italy; (M.M.); (E.O.); (V.T.); (D.d.G.); (P.F.); (S.G.)
| | - Sebastiano Gangemi
- School and Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy;
| |
Collapse
|
98
|
Kwa KAA, van Haasterecht L, Elgersma A, Breederveld RS, Groot ML, van Zuijlen PPM, Boekema BKHL. Effective enzymatic debridement of burn wounds depends on the denaturation status of collagen. Wound Repair Regen 2020; 28:666-675. [PMID: 32570295 DOI: 10.1111/wrr.12827] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 04/30/2020] [Accepted: 05/05/2020] [Indexed: 12/21/2022]
Abstract
The treatment of burn wounds by enzymatic debridement using bromelain has shown promising results in our burn center. However, inadequate debridement occurred in a few cases in which the etiology of the burn was attributed to relatively low temperature burns. We hypothesized that bromelain is ineffective in burns in which collagen denaturation, which occurs approximately at 65°C, has not taken place. Our objective was to assess whether there is a relationship between the denaturation of collagen and the ability of bromelain to debride acute scald burn wounds of different temperatures. Ex vivo human skin from four different donors was cut into 1x1 cm samples, and scald burns were produced by immersion in water at temperatures of 40°C, 50°C, 60°C, 70°C, and 100°C for 20 minutes. Denaturation of collagen was assessed with histology, using hematoxylin and eosin (H&E) staining and a fluorescently labeled collagen hybridizing peptide (CHP), and with second harmonic generation (SHG) microscopy. Burned samples and one control sample (room temperature) were weighed before and after application of enzymatic debridement to assess the efficacy of enzymatic debridement. After enzymatic debridement, a weight reduction of 80% was seen in the samples heated to 70°C and 100°C, whereas the other samples showed a reduction of 20%. Unfolding of collagen, loss of basket-weave arrangement, and necrosis was seen in samples heated to 60°C or higher. Evident CHP fluorescence, indicative of collagen denaturation, was seen in samples of 60°C, 70°C and 100°C. SHG intensity, signifying intact collagen, was significantly lower in the 70°C and 100°C group (P <.05) compared to the lower temperatures. In conclusion, denaturation of collagen in skin samples occurred between 60°C and 70°C and strongly correlated with the efficacy of enzymatic debridement. Therefore, enzymatic debridement with the use of bromelain is ineffective in scald burns lower than 60°C.
Collapse
Affiliation(s)
- Kelly A A Kwa
- Burn Center, Red Cross Hospital, Beverwijk, The Netherlands.,Department of Traumasurgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Ludo van Haasterecht
- LaserLaB Amsterdam, Department of Physics and Astronomy, Faculty of Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Amsterdam UMC Location VUmc, Department of Plastic, Reconstructive and Hand Surgery, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Anouk Elgersma
- Preclinical Research, Association of Dutch Burn Centers, Beverwijk, The Netherlands
| | - Roelf S Breederveld
- Burn Center, Red Cross Hospital, Beverwijk, The Netherlands.,Department of Traumasurgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Marie Louise Groot
- LaserLaB Amsterdam, Department of Physics and Astronomy, Faculty of Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Paul P M van Zuijlen
- Burn Center, Red Cross Hospital, Beverwijk, The Netherlands.,Amsterdam UMC Location VUmc, Department of Plastic, Reconstructive and Hand Surgery, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Bouke K H L Boekema
- Preclinical Research, Association of Dutch Burn Centers, Beverwijk, The Netherlands
| |
Collapse
|
99
|
Bourne JW, Shi L, Torzilli PA. Collagen peptide simulated bending after applied axial deformation. J Mech Behav Biomed Mater 2020; 108:103835. [PMID: 32469728 DOI: 10.1016/j.jmbbm.2020.103835] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 04/10/2020] [Accepted: 04/26/2020] [Indexed: 01/19/2023]
Abstract
Structural proteins in the extracellular matrix are subjected to a range of mechanical loading conditions, including varied directions of force application. Molecular modeling suggests that these mechanical forces directly affect collagen's conformation and the subsequent mechanical response at the molecular level is complex. For example, tensile forces in the axial direction result in collagen triple helix elongation and unwinding, while perpendicular forces can cause local triple helix disruption. However, the effects of more complicated mechanical loading, such as the effect of axial pretension on collagen bending and triple helix microunfolding are unknown. In this study we used steered molecular dynamics to first model a collagen peptide under axial tension and then apply a perpendicular bending force. Axial tension causes molecular elongation and increased the subsequent perpendicular bending stiffness, but surprisingly did not increase the predicted collagen triple helix microunfolding threshold. We believe these results elucidate a key potential mechanism by which microscale mechanical loads translate from cellular and micro scales down to the nano and atomistic. Further, these data predict that cryptic force-induced collagen triple helix unwinding is axial-deformation independent, supporting the possibility that cell traction forces could be a key molecular mechanism to alter the cellular matrix microenvironment to facilitate collagen enzymatic degradation and subsequent cellular migration, such as in tumor extravasation.
Collapse
Affiliation(s)
- Jonathan W Bourne
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, 535 E. 70(th) St., New York, NY, 10021, USA.
| | - Lei Shi
- Dept. of Physiology and Biophysics (Box 75), Weill Cornell Medical College, 1300 York Ave., New York, NY, 10065, USA
| | - Peter A Torzilli
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, 535 E. 70(th) St., New York, NY, 10021, USA; Physiology, Biophysics, and Systems Biology Program (Box 75), Weill Cornell Graduate School of Medical Sciences, 1300 York Ave., New York, NY, 10065, USA
| |
Collapse
|
100
|
Zapp C, Obarska-Kosinska A, Rennekamp B, Kurth M, Hudson DM, Mercadante D, Barayeu U, Dick TP, Denysenkov V, Prisner T, Bennati M, Daday C, Kappl R, Gräter F. Mechanoradicals in tensed tendon collagen as a source of oxidative stress. Nat Commun 2020; 11:2315. [PMID: 32385229 PMCID: PMC7210969 DOI: 10.1038/s41467-020-15567-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 03/10/2020] [Indexed: 12/31/2022] Open
Abstract
As established nearly a century ago, mechanoradicals originate from homolytic bond scission in polymers. The existence, nature and biological relevance of mechanoradicals in proteins, instead, are unknown. We here show that mechanical stress on collagen produces radicals and subsequently reactive oxygen species, essential biological signaling molecules. Electron-paramagnetic resonance (EPR) spectroscopy of stretched rat tail tendon, atomistic molecular dynamics simulations and quantum-chemical calculations show that the radicals form by bond scission in the direct vicinity of crosslinks in collagen. Radicals migrate to adjacent clusters of aromatic residues and stabilize on oxidized tyrosyl radicals, giving rise to a distinct EPR spectrum consistent with a stable dihydroxyphenylalanine (DOPA) radical. The protein mechanoradicals, as a yet undiscovered source of oxidative stress, finally convert into hydrogen peroxide. Our study suggests collagen I to have evolved as a radical sponge against mechano-oxidative damage and proposes a mechanism for exercise-induced oxidative stress and redox-mediated pathophysiological processes. The existence, nature and biological relevance of mechanoradicals in proteins are unknown. Here authors show that mechanical stress on collagen produces radicals and subsequently reactive oxygen species and suggest that collagen I evolved as a radical sponge against mechano-oxidative damage.
Collapse
Affiliation(s)
- Christopher Zapp
- Heidelberg Institute for Theoretical Studies, Schloss-Wolfsbrunnenweg 35, 69118, Heidelberg, Germany.,Institute for Theoretical Physics, Heidelberg University, Philosophenweg 16, 69120, Heidelberg, Germany
| | - Agnieszka Obarska-Kosinska
- Heidelberg Institute for Theoretical Studies, Schloss-Wolfsbrunnenweg 35, 69118, Heidelberg, Germany.,Hamburg Unit c/o DESY, European Molecular Biology Laboratory, Notkestrasse 85, 22607, Hamburg, Germany
| | - Benedikt Rennekamp
- Heidelberg Institute for Theoretical Studies, Schloss-Wolfsbrunnenweg 35, 69118, Heidelberg, Germany.,Institute for Theoretical Physics, Heidelberg University, Philosophenweg 16, 69120, Heidelberg, Germany
| | - Markus Kurth
- Heidelberg Institute for Theoretical Studies, Schloss-Wolfsbrunnenweg 35, 69118, Heidelberg, Germany
| | - David M Hudson
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Davide Mercadante
- Biochemical Institute, University of Zuerich, Winterthurerstr. 190, 8057, Zuerich, Switzerland
| | - Uladzimir Barayeu
- Faculty of Biosciences, Heidelberg University, Im Neuenheimer Feld 234, 69120, Heidelberg, Germany.,Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Tobias P Dick
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Vasyl Denysenkov
- Institute of Physical and Theoretical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 7, 60438, Frankfurt am Main, Germany
| | - Thomas Prisner
- Institute of Physical and Theoretical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 7, 60438, Frankfurt am Main, Germany
| | - Marina Bennati
- Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany
| | - Csaba Daday
- Heidelberg Institute for Theoretical Studies, Schloss-Wolfsbrunnenweg 35, 69118, Heidelberg, Germany.,Interdisciplinary Center for Scientific Computing, Heidelberg University, INF 205, 69120, Heidelberg, Germany
| | - Reinhard Kappl
- Institute for Biophysics, Saarland University Medical Center, CIPMM Geb. 48, 66421, Homburg/Saar, Germany
| | - Frauke Gräter
- Heidelberg Institute for Theoretical Studies, Schloss-Wolfsbrunnenweg 35, 69118, Heidelberg, Germany. .,Interdisciplinary Center for Scientific Computing, Heidelberg University, INF 205, 69120, Heidelberg, Germany.
| |
Collapse
|