Pub Date : 2024-02-28DOI: 10.1016/j.matbio.2024.02.008
Elisavet Angeli , Maria Jordan , Mandy Otto , Stevan D. Stojanović , Morten Karsdal , Johann Bauersachs , Thomas Thum , Jan Fiedler , Federica Genovese
Cardiomyopathies encompass a spectrum of heart disorders with diverse causes and presentations. Fibrosis stands out as a shared hallmark among various cardiomyopathies, reflecting a common thread in their pathogenesis. This prevalent fibrotic response is intricately linked to the consequences of dysregulated extracellular matrix (ECM) remodeling, emphasizing its significance in the development and progression the disease. This review explores the ECM involvement in various cardiomyopathies and its impact on myocardial stiffness and fibrosis. Additionally, we discuss the potential of ECM fragments as early diagnosis, prognosis, and risk stratification. Biomarkers deriving from turnover of collagens and other ECM proteins hold promise in clinical applications. We outline current clinical management, future directions, and the potential for personalized ECM-targeted therapies with specific focus on microRNAs. In summary, this review examines the role of the fibrosis in cardiomyopathies, highlighting the potential of ECM-derived biomarkers in improving disease management with implications for precision medicine.
{"title":"The role of fibrosis in cardiomyopathies: An opportunity to develop novel biomarkers of disease activity","authors":"Elisavet Angeli , Maria Jordan , Mandy Otto , Stevan D. Stojanović , Morten Karsdal , Johann Bauersachs , Thomas Thum , Jan Fiedler , Federica Genovese","doi":"10.1016/j.matbio.2024.02.008","DOIUrl":"10.1016/j.matbio.2024.02.008","url":null,"abstract":"<div><p>Cardiomyopathies encompass a spectrum of heart disorders with diverse causes and presentations. Fibrosis stands out as a shared hallmark among various cardiomyopathies, reflecting a common thread in their pathogenesis. This prevalent fibrotic response is intricately linked to the consequences of dysregulated extracellular matrix (ECM) remodeling, emphasizing its significance in the development and progression the disease. This review explores the ECM involvement in various cardiomyopathies and its impact on myocardial stiffness and fibrosis. Additionally, we discuss the potential of ECM fragments as early diagnosis, prognosis, and risk stratification. Biomarkers deriving from turnover of collagens and other ECM proteins hold promise in clinical applications. We outline current clinical management, future directions, and the potential for personalized ECM-targeted therapies with specific focus on microRNAs. In summary, this review examines the role of the fibrosis in cardiomyopathies, highlighting the potential of ECM-derived biomarkers in improving disease management with implications for precision medicine.</p></div>","PeriodicalId":49851,"journal":{"name":"Matrix Biology","volume":"128 ","pages":"Pages 65-78"},"PeriodicalIF":6.9,"publicationDate":"2024-02-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139998106","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-02-28DOI: 10.1016/j.matbio.2024.02.009
Pratyusha Chitturi, Andrew Leask
The largest mammalian organ, skin, consisting of a dermal connective tissue layer that underlies and supports the epidermis, acts as a protective barrier that excludes external pathogens and disseminates sensory signals emanating from the local microenvironment. Dermal connective tissue is comprised of a collagen-rich extracellular matrix (ECM) that is produced by connective tissue fibroblasts resident within the dermis. When wounded, a tissue repair program is induced whereby fibroblasts, in response to alterations in the microenvironment, produce new ECM components, resulting in the formation of a scar. Failure to terminate the normal tissue repair program causes fibrotic conditions including: hypertrophic scars, keloids, and the systemic autoimmune connective tissue disease scleroderma (systemic sclerosis, SSc). Histological and single-cell RNA sequencing (scRNAseq) studies have revealed that fibroblasts are heterogeneous and highly plastic. Understanding how this diversity contributes to dermal homeostasis, wounding, fibrosis, and cancer may ultimately result in novel anti-fibrotic therapies and personalized medicine. This review summarizes studies supporting this concept.
{"title":"The role of positional information in determining dermal fibroblast diversity","authors":"Pratyusha Chitturi, Andrew Leask","doi":"10.1016/j.matbio.2024.02.009","DOIUrl":"10.1016/j.matbio.2024.02.009","url":null,"abstract":"<div><p>The largest mammalian organ, skin, consisting of a dermal connective tissue layer that underlies and supports the epidermis, acts as a protective barrier that excludes external pathogens and disseminates sensory signals emanating from the local microenvironment. Dermal connective tissue is comprised of a collagen-rich extracellular matrix (ECM) that is produced by connective tissue fibroblasts resident within the dermis. When wounded, a tissue repair program is induced whereby fibroblasts, in response to alterations in the microenvironment, produce new ECM components, resulting in the formation of a scar. Failure to terminate the normal tissue repair program causes fibrotic conditions including: hypertrophic scars, keloids, and the systemic autoimmune connective tissue disease scleroderma (systemic sclerosis, SSc). Histological and single-cell RNA sequencing (scRNAseq) studies have revealed that fibroblasts are heterogeneous and highly plastic. Understanding how this diversity contributes to dermal homeostasis, wounding, fibrosis, and cancer may ultimately result in novel anti-fibrotic therapies and personalized medicine. This review summarizes studies supporting this concept.</p></div>","PeriodicalId":49851,"journal":{"name":"Matrix Biology","volume":"128 ","pages":"Pages 31-38"},"PeriodicalIF":6.9,"publicationDate":"2024-02-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0945053X24000362/pdfft?md5=39f70e589c6770fd750b2ae39b89e2e3&pid=1-s2.0-S0945053X24000362-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139998107","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-02-21DOI: 10.1016/j.matbio.2024.02.007
Mahsima Khoshneviszadeh , Solveig Henneicke , Daniel Pirici , Akilashree Senthilnathan , Lorena Morton , Philipp Arndt , Rahul Kaushik , Oula Norman , Jari Jukkola , Ildiko Rita Dunay , Constanze Seidenbecher , Anne Heikkinen , Stefanie Schreiber , Alexander Dityatev
Collagen type XVIII (COL18) is an abundant heparan sulfate proteoglycan in vascular basement membranes. Here, we asked (i) if the loss of COL18 would result in blood-brain barrier (BBB) breakdown, pathological alterations of small arteries and capillaries and neuroinflammation as found in cerebral small vessel disease (CSVD) and (ii) if such changes may be associated with remodeling of synapses and neural extracellular matrix (ECM). We found that 5-month-old Col18a1−/− mice had elevated BBB permeability for mouse IgG in the deep gray matter, and intravascular erythrocyte accumulations were observed brain-wide in capillaries and arterioles. BBB permeability increased with age and affected cortical regions and the hippocampus in 12-month-old Col18a1−/− mice. None of the Col18a1−/− mice displayed hallmarks of advanced CSVD, such as hemorrhages, and did not show perivascular space enlargement. Col18a1 deficiency-induced BBB leakage was accompanied by activation of microglia and astrocytes, a loss of aggrecan in the ECM of perineuronal nets associated with fast-spiking inhibitory interneurons and accumulation of the perisynaptic ECM proteoglycan brevican and the microglial complement protein C1q at excitatory synapses. As the pathway underlying these regulations, we found increased signaling through the TGF-ß1/Smad3/TIMP-3 cascade. We verified the pivotal role of COL18 for small vessel wall structure in CSVD by demonstrating the protein's involvement in vascular remodeling in autopsy brains from patients with cerebral hypertensive arteriopathy. Our study highlights an association between the alterations of perivascular ECM, extracellular proteolysis, and perineuronal/perisynaptic ECM, as a possible substrate of synaptic and cognitive alterations in CSVD.
{"title":"Microvascular damage, neuroinflammation and extracellular matrix remodeling in Col18a1 knockout mice as a model for early cerebral small vessel disease","authors":"Mahsima Khoshneviszadeh , Solveig Henneicke , Daniel Pirici , Akilashree Senthilnathan , Lorena Morton , Philipp Arndt , Rahul Kaushik , Oula Norman , Jari Jukkola , Ildiko Rita Dunay , Constanze Seidenbecher , Anne Heikkinen , Stefanie Schreiber , Alexander Dityatev","doi":"10.1016/j.matbio.2024.02.007","DOIUrl":"10.1016/j.matbio.2024.02.007","url":null,"abstract":"<div><p>Collagen type XVIII (COL18) is an abundant heparan sulfate proteoglycan in vascular basement membranes. Here, we asked (i) if the loss of COL18 would result in blood-brain barrier (BBB) breakdown, pathological alterations of small arteries and capillaries and neuroinflammation as found in cerebral small vessel disease (CSVD) and (ii) if such changes may be associated with remodeling of synapses and neural extracellular matrix (ECM). We found that 5-month-old <em>Col18a1<sup>−/−</sup></em> mice had elevated BBB permeability for mouse IgG in the deep gray matter, and intravascular erythrocyte accumulations were observed brain-wide in capillaries and arterioles. BBB permeability increased with age and affected cortical regions and the hippocampus in 12-month-old <em>Col18a1<sup>−/−</sup></em> mice. None of the <em>Col18a1<sup>−/−</sup></em> mice displayed hallmarks of advanced CSVD, such as hemorrhages, and did not show perivascular space enlargement. <em>Col18a1</em> deficiency-induced BBB leakage was accompanied by activation of microglia and astrocytes, a loss of aggrecan in the ECM of perineuronal nets associated with fast-spiking inhibitory interneurons and accumulation of the perisynaptic ECM proteoglycan brevican and the microglial complement protein C1q at excitatory synapses. As the pathway underlying these regulations, we found increased signaling through the TGF-ß1/Smad3/TIMP-3 cascade. We verified the pivotal role of COL18 for small vessel wall structure in CSVD by demonstrating the protein's involvement in vascular remodeling in autopsy brains from patients with cerebral hypertensive arteriopathy. Our study highlights an association between the alterations of perivascular ECM, extracellular proteolysis, and perineuronal/perisynaptic ECM, as a possible substrate of synaptic and cognitive alterations in CSVD.</p></div>","PeriodicalId":49851,"journal":{"name":"Matrix Biology","volume":"128 ","pages":"Pages 39-64"},"PeriodicalIF":6.9,"publicationDate":"2024-02-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0945053X2400026X/pdfft?md5=f5e54f1667086f5aee9e56f6e36957f1&pid=1-s2.0-S0945053X2400026X-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139917019","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-02-19DOI: 10.1016/j.matbio.2024.02.006
Paul Hiebert , Giuseppe Antoniazzi , Matthew Aronoff , Sabine Werner , Helma Wennemers
Tissue repair and fibrosis involve the dynamic remodeling of collagen, and accurate detection of these sites is of utmost importance. Here, we use a collagen peptide sensor (1) to visualize collagen formation and remodeling during wound healing in mice and humans. We show that the probe binds selectively to sites of collagen formation and remodeling at different stages of healing. Compared to conventional methods, the peptide sensor localizes preferentially to areas of collagen synthesis and remodeling at the wound edge and not in matured fibrillar collagen. We also demonstrate its applicability for in vivo wound imaging and for discerning differential remodeling in wounds of transgenic mice with altered collagen dynamics. Our findings show the value of 1 as a diagnostic tool to rapidly identify the sites of matrix remodeling in tissue sections, which will aid in the conception of new therapeutic strategies for fibrotic disorders and defective tissue repair.
{"title":"A lysyl oxidase-responsive collagen peptide illuminates collagen remodeling in wound healing","authors":"Paul Hiebert , Giuseppe Antoniazzi , Matthew Aronoff , Sabine Werner , Helma Wennemers","doi":"10.1016/j.matbio.2024.02.006","DOIUrl":"10.1016/j.matbio.2024.02.006","url":null,"abstract":"<div><p>Tissue repair and fibrosis involve the dynamic remodeling of collagen, and accurate detection of these sites is of utmost importance. Here, we use a collagen peptide sensor (<strong>1</strong>) to visualize collagen formation and remodeling during wound healing in mice and humans. We show that the probe binds selectively to sites of collagen formation and remodeling at different stages of healing. Compared to conventional methods, the peptide sensor localizes preferentially to areas of collagen synthesis and remodeling at the wound edge and not in matured fibrillar collagen. We also demonstrate its applicability for <em>in vivo</em> wound imaging and for discerning differential remodeling in wounds of transgenic mice with altered collagen dynamics. Our findings show the value of <strong>1</strong> as a diagnostic tool to rapidly identify the sites of matrix remodeling in tissue sections, which will aid in the conception of new therapeutic strategies for fibrotic disorders and defective tissue repair.</p></div>","PeriodicalId":49851,"journal":{"name":"Matrix Biology","volume":"128 ","pages":"Pages 11-20"},"PeriodicalIF":6.9,"publicationDate":"2024-02-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0945053X24000258/pdfft?md5=9c7f01eb15a3e7eb570c79764f015938&pid=1-s2.0-S0945053X24000258-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139916920","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-02-18DOI: 10.1016/j.matbio.2024.02.005
Johanna Lena Schön , Victoria Elisabeth Groß , Willem Berend Post , Alexandra Daum , Daniel Matúš , Johanna Pilz , Rene Schnorr , Susanne Horn , Miriam Bäumers , Stefanie Weidtkamp-Peters , Samantha Hughes , Torsten Schöneberg , Simone Prömel
The extracellular matrix (ECM) is a network of macromolecules that presents a vital scaffold for cells and enables multiple ways of cellular communication. Thus, it is essential for many physiological processes such as development, tissue morphogenesis, homeostasis, the shape and partially the size of the body and its organs. To ensure these, the composition of the ECM is tissue-specific and highly dynamic. ECM homeostasis is therefore tightly controlled by several mechanisms.
Here, we show that FMI-1, the homolog of the Adhesion GPCR Flamingo/CELSR/ADGRC in the nematode Caenorhabditis elegans, modulates the composition of the ECM by controlling the production both of ECM molecules such as collagens and also of ECM modifying enzymes. Thereby, FMI-1 affects the morphology and functionality of the nematode´s cuticle, which is mainly composed of ECM, and also modulates the body size. Mechanistic analyses highlight the fact that FMI-1 exerts its function from neurons non-cell autonomously (trans) solely via its extracellular N terminus. Our data support a model, by which the activity of the receptor, which has a well-described role in the planar cell polarity (PCP) pathway, involves the PCP molecule VANG-1, but seems to be independent of the DBL-1/BMP pathway.
{"title":"The adhesion GPCR and PCP component flamingo (FMI-1) alters body size and regulates the composition of the extracellular matrix","authors":"Johanna Lena Schön , Victoria Elisabeth Groß , Willem Berend Post , Alexandra Daum , Daniel Matúš , Johanna Pilz , Rene Schnorr , Susanne Horn , Miriam Bäumers , Stefanie Weidtkamp-Peters , Samantha Hughes , Torsten Schöneberg , Simone Prömel","doi":"10.1016/j.matbio.2024.02.005","DOIUrl":"10.1016/j.matbio.2024.02.005","url":null,"abstract":"<div><p>The extracellular matrix (ECM) is a network of macromolecules that presents a vital scaffold for cells and enables multiple ways of cellular communication. Thus, it is essential for many physiological processes such as development, tissue morphogenesis, homeostasis, the shape and partially the size of the body and its organs. To ensure these, the composition of the ECM is tissue-specific and highly dynamic. ECM homeostasis is therefore tightly controlled by several mechanisms.</p><p>Here, we show that FMI-1, the homolog of the Adhesion GPCR Flamingo/CELSR/ADGRC in the nematode <em>Caenorhabditis elegans</em>, modulates the composition of the ECM by controlling the production both of ECM molecules such as collagens and also of ECM modifying enzymes. Thereby, FMI-1 affects the morphology and functionality of the nematode´s cuticle, which is mainly composed of ECM, and also modulates the body size. Mechanistic analyses highlight the fact that FMI-1 exerts its function from neurons non-cell autonomously (<em>trans</em>) solely via its extracellular N terminus. Our data support a model, by which the activity of the receptor, which has a well-described role in the planar cell polarity (PCP) pathway, involves the PCP molecule VANG-1, but seems to be independent of the DBL-1/BMP pathway.</p></div>","PeriodicalId":49851,"journal":{"name":"Matrix Biology","volume":"128 ","pages":"Pages 1-10"},"PeriodicalIF":6.9,"publicationDate":"2024-02-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0945053X24000246/pdfft?md5=a8bd278d8e87c1f01e376f3063d345f1&pid=1-s2.0-S0945053X24000246-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139913960","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-02-09DOI: 10.1016/j.matbio.2024.02.004
Mei Sun , Ana Carolina Acosta , Victoria Emerick , Sheila Adams , Marcel Y Avila , Curtis E Margo , Edgar M Espana
Patients with classical Ehlers Danlos syndrome (cEDS) suffer impaired wound healing and from scars formed after injuries that are atrophic and difficult to close surgically. Haploinsufficiency in COL5A1 creates systemic morphological and functional alterations in the entire body. We investigated mechanisms that impair wound healing from corneal lacerations (full thickness injuries) in a mouse model of cEDS (Col5a1+/−). We found that collagen V reexpression in this model is upregulated during corneal tissue repair and that wound healing is delayed, impaired, and results in large atrophic corneal scars. We noted that in a matrix with a 50 % content of collagen V, activation of latent Transforming Growth Factor (TGF) β is dysregulated. Corneal myofibroblasts with a haploinsufficiency of collagen V failed to mechanically activate latent TGF β. Second harmonic imaging microscopy showed a disorganized, undulated, and denser collagen matrix in our Col5a1+/- model that suggested alterations in the extracellular matrix structure and function. We hypothesize that a regenerated collagen matrix with only 50 % content of collagen V is not resistant enough mechanically to allow adequate activation of latent TGF β by fibroblasts and myofibroblasts.
典型埃勒斯-丹洛斯综合征(cEDS)患者的伤口愈合能力受损,受伤后形成的疤痕萎缩且难以通过手术缝合。COL5A1单倍体缺陷会导致全身形态和功能改变。我们研究了cEDS(Col5a1+/-)小鼠模型中影响角膜裂伤(全厚损伤)伤口愈合的机制。我们发现,在该模型中,胶原蛋白 V 的再表达在角膜组织修复过程中上调,伤口愈合延迟、受损,并导致大面积萎缩性角膜疤痕。我们注意到,在胶原蛋白 V 含量为 50% 的基质中,潜伏的转化生长因子 (TGF) β 的激活失调。二次谐波成像显微镜显示,在我们的 Col5a1+/- 模型中,胶原基质变得杂乱、起伏和致密,这表明细胞外基质的结构和功能发生了改变。我们推测,再生的胶原基质中胶原 V 的含量仅为 50%,机械抵抗力不足,无法充分激活成纤维细胞和肌成纤维细胞潜伏的 TGF β。
{"title":"Dysfunctional latent transforming growth factor β activation after corneal injury in a classical Ehlers–Danlos model","authors":"Mei Sun , Ana Carolina Acosta , Victoria Emerick , Sheila Adams , Marcel Y Avila , Curtis E Margo , Edgar M Espana","doi":"10.1016/j.matbio.2024.02.004","DOIUrl":"10.1016/j.matbio.2024.02.004","url":null,"abstract":"<div><p>Patients with classical Ehlers Danlos syndrome (cEDS) suffer impaired wound healing and from scars formed after injuries that are atrophic and difficult to close surgically. Haploinsufficiency in COL5A1 creates systemic morphological and functional alterations in the entire body. We investigated mechanisms that impair wound healing from corneal lacerations (full thickness injuries) in a mouse model of cEDS (<em>Col5a1</em><sup>+/−</sup>). We found that collagen V reexpression in this model is upregulated during corneal tissue repair and that wound healing is delayed, impaired, and results in large atrophic corneal scars. We noted that in a matrix with a 50 % content of collagen V, activation of latent Transforming Growth Factor (TGF) β is dysregulated. Corneal myofibroblasts with a haploinsufficiency of collagen V failed to mechanically activate latent TGF β. Second harmonic imaging microscopy showed a disorganized, undulated, and denser collagen matrix in our <em>Col5a1</em><sup>+/-</sup> model that suggested alterations in the extracellular matrix structure and function. We hypothesize that a regenerated collagen matrix with only 50 % content of collagen V is not resistant enough mechanically to allow adequate activation of latent TGF β by fibroblasts and myofibroblasts.</p></div>","PeriodicalId":49851,"journal":{"name":"Matrix Biology","volume":"128 ","pages":"Pages 21-30"},"PeriodicalIF":6.9,"publicationDate":"2024-02-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139716562","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-02-08DOI: 10.1016/j.matbio.2024.02.003
Whitney M. Longmate , Emily Norton , Giesse Albeche Duarte , Lei Wu , Mathieu R. DiPersio , John M. Lamar , C. Michael DiPersio
The development of wound therapy targeting integrins is hampered by inadequate understanding of integrin function in cutaneous wound healing and the wound microenvironment. Following cutaneous injury, keratinocytes migrate to restore the skin barrier, and macrophages aid in debris clearance. Thus, both keratinocytes and macrophages are critical to the coordination of tissue repair. Keratinocyte integrins have been shown to participate in this coordinated effort by regulating secreted factors, some of which crosstalk to distinct cells in the wound microenvironment. Epidermal integrin α3β1 is a receptor for laminin-332 in the cutaneous basement membrane. Here we show that wounds deficient in epidermal α3β1 express less epidermal-derived macrophage colony-stimulating factor 1 (CSF-1), the primary macrophage-stimulating growth factor. α3β1-deficient wounds also have fewer wound-proximal macrophages, suggesting that keratinocyte α3β1 may stimulate wound macrophages through the regulation of CSF-1. Indeed, using a set of immortalized keratinocytes, we demonstrate that keratinocyte-derived CSF-1 supports macrophage growth, and that α3β1 regulates Csf1 expression through Src-dependent stimulation of Yes-associated protein (YAP)-Transcriptional enhanced associate domain (TEAD)-mediated transcription. Consistently, α3β1-deficient wounds in vivo display a substantially reduced number of keratinocytes with YAP-positive nuclei. Overall, our current findings identify a novel role for epidermal integrin α3β1 in regulating the cutaneous wound microenvironment by mediating paracrine crosstalk from keratinocytes to wound macrophages, implicating α3β1 as a potential target of wound therapy.
{"title":"Keratinocyte integrin α3β1 induces expression of the macrophage stimulating factor, CSF-1, through a YAP/TEAD-dependent mechanism.","authors":"Whitney M. Longmate , Emily Norton , Giesse Albeche Duarte , Lei Wu , Mathieu R. DiPersio , John M. Lamar , C. Michael DiPersio","doi":"10.1016/j.matbio.2024.02.003","DOIUrl":"10.1016/j.matbio.2024.02.003","url":null,"abstract":"<div><p>The development of wound therapy targeting integrins is hampered by inadequate understanding of integrin function in cutaneous wound healing and the wound microenvironment. Following cutaneous injury, keratinocytes migrate to restore the skin barrier, and macrophages aid in debris clearance. Thus, both keratinocytes and macrophages are critical to the coordination of tissue repair. Keratinocyte integrins have been shown to participate in this coordinated effort by regulating secreted factors, some of which crosstalk to distinct cells in the wound microenvironment. Epidermal integrin α3β1 is a receptor for laminin-332 in the cutaneous basement membrane. Here we show that wounds deficient in epidermal α3β1 express less epidermal-derived macrophage colony-stimulating factor 1 (CSF-1), the primary macrophage-stimulating growth factor. α3β1-deficient wounds also have fewer wound-proximal macrophages, suggesting that keratinocyte α3β1 may stimulate wound macrophages through the regulation of CSF-1. Indeed, using a set of immortalized keratinocytes, we demonstrate that keratinocyte-derived CSF-1 supports macrophage growth, and that α3β1 regulates <em>Csf1</em> expression through Src-dependent stimulation of Yes-associated protein (YAP)-Transcriptional enhanced associate domain (TEAD)-mediated transcription. Consistently, α3β1-deficient wounds <em>in vivo</em> display a substantially reduced number of keratinocytes with YAP-positive nuclei. Overall, our current findings identify a novel role for epidermal integrin α3β1 in regulating the cutaneous wound microenvironment by mediating paracrine crosstalk from keratinocytes to wound macrophages, implicating α3β1 as a potential target of wound therapy.</p></div>","PeriodicalId":49851,"journal":{"name":"Matrix Biology","volume":"127 ","pages":"Pages 48-56"},"PeriodicalIF":6.9,"publicationDate":"2024-02-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139716563","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-02-07DOI: 10.1016/j.matbio.2024.02.002
Christine Chew , Oliver J Brand , Tomohiko Yamamura , Craig Lawless , Mychel Raony Paiva Teixeira Morais , Leo Zeef , I-Hsuan Lin , Gareth Howell , Sylvia Lui , Franziska Lausecker , Christopher Jagger , Tovah N Shaw , Siddharth Krishnan , Flora A McClure , Hayley Bridgeman , Kelly Wemyss , Joanne E Konkel , Tracy Hussell , Rachel Lennon
Background
The kidney contains distinct glomerular and tubulointerstitial compartments with diverse cell types and extracellular matrix components. The role of immune cells in glomerular environment is crucial for dampening inflammation and maintaining homeostasis. Macrophages are innate immune cells that are influenced by their tissue microenvironment. However, the multifunctional role of kidney macrophages remains unclear.
Methods
Flow and imaging cytometry were used to determine the relative expression of CD81 and CX3CR1 (C-X3-C motif chemokine receptor 1) in kidney macrophages. Monocyte replenishment was assessed in Cx3cr1CreER X R26-yfp-reporter and shielded chimeric mice. Bulk RNA-sequencing and mass spectrometry-based proteomics were performed on isolated kidney macrophages from wild type and Col4a5−/− (Alport) mice. RNAscope was used to visualize transcripts and macrophage purity in bulk RNA assessed by CIBERSORTx analyses.
Results
In wild type mice we identified three distinct kidney macrophage subsets using CD81 and CX3CR1 and these subsets showed dependence on monocyte replenishment. In addition to their immune function, bulk RNA-sequencing of macrophages showed enrichment of biological processes associated with extracellular matrix. Proteomics identified collagen IV and laminins in kidney macrophages from wild type mice whilst other extracellular matrix proteins including cathepsins, ANXA2 and LAMP2 were enriched in Col4a5−/− (Alport) mice. A subset of kidney macrophages co-expressed matrix and macrophage transcripts.
Conclusions
We identified CD81 and CX3CR1 positive kidney macrophage subsets with distinct dependence for monocyte replenishment. Multiomic analysis demonstrated that these cells have diverse functions that underscore the importance of macrophages in kidney health and disease.
{"title":"Kidney resident macrophages have distinct subsets and multifunctional roles","authors":"Christine Chew , Oliver J Brand , Tomohiko Yamamura , Craig Lawless , Mychel Raony Paiva Teixeira Morais , Leo Zeef , I-Hsuan Lin , Gareth Howell , Sylvia Lui , Franziska Lausecker , Christopher Jagger , Tovah N Shaw , Siddharth Krishnan , Flora A McClure , Hayley Bridgeman , Kelly Wemyss , Joanne E Konkel , Tracy Hussell , Rachel Lennon","doi":"10.1016/j.matbio.2024.02.002","DOIUrl":"10.1016/j.matbio.2024.02.002","url":null,"abstract":"<div><h3>Background</h3><p>The kidney contains distinct glomerular and tubulointerstitial compartments with diverse cell types and extracellular matrix components. The role of immune cells in glomerular environment is crucial for dampening inflammation and maintaining homeostasis. Macrophages are innate immune cells that are influenced by their tissue microenvironment. However, the multifunctional role of kidney macrophages remains unclear.</p></div><div><h3>Methods</h3><p>Flow and imaging cytometry were used to determine the relative expression of CD81 and CX<sub>3</sub>CR1 (C-X3-C motif chemokine receptor 1) in kidney macrophages. Monocyte replenishment was assessed in <em>Cx3cr1</em><sup>CreER</sup> X <em>R26-yfp</em>-reporter and shielded chimeric mice. Bulk RNA-sequencing and mass spectrometry-based proteomics were performed on isolated kidney macrophages from wild type and <em>Col4a5</em><sup>−/−</sup> (Alport) mice. RNAscope was used to visualize transcripts and macrophage purity in bulk RNA assessed by CIBERSORTx analyses.</p></div><div><h3>Results</h3><p>In wild type mice we identified three distinct kidney macrophage subsets using CD81 and CX<sub>3</sub>CR1 and these subsets showed dependence on monocyte replenishment. In addition to their immune function, bulk RNA-sequencing of macrophages showed enrichment of biological processes associated with extracellular matrix. Proteomics identified collagen IV and laminins in kidney macrophages from wild type mice whilst other extracellular matrix proteins including cathepsins, ANXA2 and LAMP2 were enriched in <em>Col4a5<sup>−/−</sup></em> (Alport) mice. A subset of kidney macrophages co-expressed matrix and macrophage transcripts.</p></div><div><h3>Conclusions</h3><p>We identified CD81 and CX<sub>3</sub>CR1 positive kidney macrophage subsets with distinct dependence for monocyte replenishment. Multiomic analysis demonstrated that these cells have diverse functions that underscore the importance of macrophages in kidney health and disease.</p></div>","PeriodicalId":49851,"journal":{"name":"Matrix Biology","volume":"127 ","pages":"Pages 23-37"},"PeriodicalIF":6.9,"publicationDate":"2024-02-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0945053X24000210/pdfft?md5=07a30ae5fc61894613edbbab00b8f6ca&pid=1-s2.0-S0945053X24000210-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139708384","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Volumetric muscle loss (VML) represents a clinical challenge due to the limited regenerative capacity of skeletal muscle. Most often, it results in scar tissue formation and loss of function, which cannot be prevented by current therapies. Decellularized extracellular matrix (DEM) has emerged as a native biomaterial for the enhancement of tissue repair. Here, we report the generation and characterization of hydrogels derived from DEM prepared from WT or thrombospondin (TSP)-2 null muscle tissue. TSP2-null hydrogels, when compared to WT, displayed altered architecture, protein composition, and biomechanical properties and allowed enhanced invasion of C2C12 myocytes and chord formation by endothelial cells. They also displayed enhanced cell invasion, innervation, and angiogenesis following subcutaneous implantation. To evaluate their regenerative capacity, WT or TSP2 null hydrogels were used to treat VML injury to tibialis anterior muscles and the latter induced greater recruitment of repair cells, innervation, and blood vessel formation and reduced inflammation. Taken together, these observations indicate that TSP2-null hydrogels enhance angiogenesis and promote muscle repair in a VML model.
{"title":"Novel muscle-derived extracellular matrix hydrogel promotes angiogenesis and neurogenesis in volumetric muscle loss","authors":"Zhuoyue Chen , Yaqing Huang , Hao Xing , Tiffany Tseng , Hailey Edelman , Rachel Perry , Themis R. Kyriakides","doi":"10.1016/j.matbio.2024.02.001","DOIUrl":"10.1016/j.matbio.2024.02.001","url":null,"abstract":"<div><p>Volumetric muscle loss (VML) represents a clinical challenge due to the limited regenerative capacity of skeletal muscle. Most often, it results in scar tissue formation and loss of function, which cannot be prevented by current therapies. Decellularized extracellular matrix (DEM) has emerged as a native biomaterial for the enhancement of tissue repair. Here, we report the generation and characterization of hydrogels derived from DEM prepared from WT or thrombospondin (TSP)-2 null muscle tissue. TSP2-null hydrogels, when compared to WT, displayed altered architecture, protein composition, and biomechanical properties and allowed enhanced invasion of C2C12 myocytes and chord formation by endothelial cells. They also displayed enhanced cell invasion, innervation, and angiogenesis following subcutaneous implantation. To evaluate their regenerative capacity, WT or TSP2 null hydrogels were used to treat VML injury to tibialis anterior muscles and the latter induced greater recruitment of repair cells, innervation, and blood vessel formation and reduced inflammation. Taken together, these observations indicate that TSP2-null hydrogels enhance angiogenesis and promote muscle repair in a VML model.</p></div>","PeriodicalId":49851,"journal":{"name":"Matrix Biology","volume":"127 ","pages":"Pages 38-47"},"PeriodicalIF":6.9,"publicationDate":"2024-02-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139690506","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-02-01DOI: 10.1016/j.matbio.2024.01.004
Fengying Tang , Stephen R. Reeves , Jourdan E. Brune , Mary Y. Chang , Christina K. Chan , Peter Waldron , Sheona P. Drummond , Caroline M. Milner , Kimberly M. Alonge , Stavros Garantziotis , Anthony J. Day , William A. Altemeier , Charles W. Frevert
<div><p>The inter-alpha-trypsin inhibitor (IαI) complex is composed of the bikunin core protein with a single chondroitin sulfate (CS) attached and one or two heavy chains (HCs) covalently linked to the CS chain. The HCs from IαI can be transferred to hyaluronan (HA) through a TNFα-stimulated gene-6 (TSG-6) dependent process to form an HC•HA matrix. Previous studies reported increased IαI, HA, and HC•HA complexes in mouse bronchoalveolar lavage fluid (BALF) post-influenza infection. However, the expression and incorporation of HCs into the HA matrix of the lungs during the clinical course of influenza A virus (IAV) infection and the biological significance of the HC•HA matrix are poorly understood. The present study aimed to better understand the composition of HC•HA matrices in mice infected with IAV and how these matrices regulate the host pulmonary immune response. In IAV infected mice bikunin, HC1–3, TSG-6, and HAS1–3 all show increased gene expression at various times during a 12-day clinical course. The increased accumulation of IαI and HA was confirmed in the lungs of infected mice using immunohistochemistry and quantitative digital pathology. Western blots confirmed increases in the IαI components in BALF and lung tissue at 6 days post-infection (dpi). Interestingly, HCs and bikunin recovered from BALF and plasma from mice 6 dpi with IAV, displayed differences in the HC composition by Western blot analysis and differences in bikunin's CS chain sulfation patterns by mass spectrometry analysis. This strongly suggests that the IαI components were synthesized in the lungs rather than translocated from the vascular compartment. HA was significantly increased in BALF at 6 dpi, and the HA recovered in BALF and lung tissues were modified with HCs indicating the presence of an HC•HA matrix. <em>In vitro</em> experiments using polyinosinic-polycytidylic acid (poly(I:C)) treated mouse lung fibroblasts (MLF) showed that modification of HA with HCs increased cell-associated HA, and that this increase was due to the retention of HA in the MLF glycocalyx. <em>In vitro</em> studies of leukocyte adhesion showed differential binding of lymphoid (Hut78), monocyte (U937), and neutrophil (dHL60) cell lines to HA and HC•HA matrices. Hut78 cells adhered to immobilized HA in a size and concentration-dependent manner. In contrast, the binding of dHL60 and U937 cells depended on generating a HC•HA matrix by MLF. Our <em>in vivo</em> findings, using multiple bronchoalveolar lavages, correlated with our <em>in vitro</em> findings in that lymphoid cells bound more tightly to the HA-glycocalyx in the lungs of influenza-infected mice than neutrophils and mononuclear phagocytes (MNPs). The neutrophils and MNPs were associated with a HC•HA matrix and were more readily lavaged from the lungs. In conclusion, this work shows increased IαI and HA accumulation and the formation of a HC•HA matrix in mouse lungs post-IAV infection. The formation of HA and HC•HA matrices could p
α-胰蛋白酶间抑制物(IαI)复合物由带有单个硫酸软骨素(CS)的比库宁核心蛋白和与 CS 链共价连接的一条或两条重链(HCs)组成。IαI 中的 HCs 可通过 TNFα-stimulated gene-6 (TSG-6) 依赖过程转移到透明质酸(HA)上,形成 HC-HA 基质。之前的研究报告称,流感感染后小鼠支气管肺泡灌洗液(BALF)中的 IαI、HA 和 HC-HA 复合物增加。然而,人们对甲型流感病毒(IAV)感染的临床过程中 HCs 的表达、HA 与肺部 HA 基质的结合以及 HC-HA 基质的生物学意义知之甚少。本研究旨在更好地了解感染 IAV 的小鼠体内 HC-HA 基质的组成以及这些基质如何调节宿主肺部免疫反应。在感染 IAV 的小鼠中,bikunin、HC1-3、TSG-6 和 HAS1-3 在 12 天的临床过程中的不同时期均出现基因表达增加。使用免疫组化和定量数字病理学方法证实了 IαI 和 HA 在感染小鼠肺部的积累增加。Western 印迹证实,在感染后 6 天(dpi),BALF 和肺组织中的 IαI 成分有所增加。有趣的是,从感染 IAV 6 dpi 小鼠的 BALF 和血浆中回收的 HC 和 bikunin,通过 Western 印迹分析显示出 HC 成分的差异,通过质谱分析显示出 bikunin 的 CS 链硫酸化模式的差异。这有力地表明,IαI 成分是在肺中合成的,而不是从血管区转移过来的。6 dpi时,BALF中的HA明显增加,BALF和肺组织中回收的HA被HC修饰,表明存在HC-HA基质。使用聚肌苷酸-聚胞苷酸(poly(I:C))处理的小鼠肺成纤维细胞(MLF)进行的体外实验表明,用 HCs 修饰 HA 可增加细胞相关的 HA,这种增加是由于 HA 被保留在 MLF 糖萼中。白细胞粘附的体外研究表明,淋巴细胞(Hut78)、单核细胞(U937)和中性粒细胞(dHL60)与 HA 和 HC-HA 基质的粘附程度不同。Hut78 细胞以大小和浓度依赖的方式粘附在固定的 HA 上。相比之下,dHL60 和 U937 细胞的粘附取决于通过 MLF 生成 HC-HA 基质。我们通过多次支气管肺泡灌洗获得的体内研究结果与体外研究结果一致,即淋巴细胞比中性粒细胞和单核吞噬细胞(MNPs)更紧密地与流感感染小鼠肺部的HA-糖萼结合。中性粒细胞和单核吞噬细胞与 HC-HA 基质相关联,更容易从肺部洗出。总之,这项研究表明,IIAV 感染后,小鼠肺部的 IαI 和 HA 积累增加,并形成 HC-HA 基质。HA 和 HC-HA 基质的形成有可能在 IAV 感染期间为免疫细胞的招募和激活创造特定的肺部微环境。
{"title":"Inter-alpha-trypsin inhibitor (IαI) and hyaluronan modifications enhance the innate immune response to influenza virus in the lung","authors":"Fengying Tang , Stephen R. Reeves , Jourdan E. Brune , Mary Y. Chang , Christina K. Chan , Peter Waldron , Sheona P. Drummond , Caroline M. Milner , Kimberly M. Alonge , Stavros Garantziotis , Anthony J. Day , William A. Altemeier , Charles W. Frevert","doi":"10.1016/j.matbio.2024.01.004","DOIUrl":"10.1016/j.matbio.2024.01.004","url":null,"abstract":"<div><p>The inter-alpha-trypsin inhibitor (IαI) complex is composed of the bikunin core protein with a single chondroitin sulfate (CS) attached and one or two heavy chains (HCs) covalently linked to the CS chain. The HCs from IαI can be transferred to hyaluronan (HA) through a TNFα-stimulated gene-6 (TSG-6) dependent process to form an HC•HA matrix. Previous studies reported increased IαI, HA, and HC•HA complexes in mouse bronchoalveolar lavage fluid (BALF) post-influenza infection. However, the expression and incorporation of HCs into the HA matrix of the lungs during the clinical course of influenza A virus (IAV) infection and the biological significance of the HC•HA matrix are poorly understood. The present study aimed to better understand the composition of HC•HA matrices in mice infected with IAV and how these matrices regulate the host pulmonary immune response. In IAV infected mice bikunin, HC1–3, TSG-6, and HAS1–3 all show increased gene expression at various times during a 12-day clinical course. The increased accumulation of IαI and HA was confirmed in the lungs of infected mice using immunohistochemistry and quantitative digital pathology. Western blots confirmed increases in the IαI components in BALF and lung tissue at 6 days post-infection (dpi). Interestingly, HCs and bikunin recovered from BALF and plasma from mice 6 dpi with IAV, displayed differences in the HC composition by Western blot analysis and differences in bikunin's CS chain sulfation patterns by mass spectrometry analysis. This strongly suggests that the IαI components were synthesized in the lungs rather than translocated from the vascular compartment. HA was significantly increased in BALF at 6 dpi, and the HA recovered in BALF and lung tissues were modified with HCs indicating the presence of an HC•HA matrix. <em>In vitro</em> experiments using polyinosinic-polycytidylic acid (poly(I:C)) treated mouse lung fibroblasts (MLF) showed that modification of HA with HCs increased cell-associated HA, and that this increase was due to the retention of HA in the MLF glycocalyx. <em>In vitro</em> studies of leukocyte adhesion showed differential binding of lymphoid (Hut78), monocyte (U937), and neutrophil (dHL60) cell lines to HA and HC•HA matrices. Hut78 cells adhered to immobilized HA in a size and concentration-dependent manner. In contrast, the binding of dHL60 and U937 cells depended on generating a HC•HA matrix by MLF. Our <em>in vivo</em> findings, using multiple bronchoalveolar lavages, correlated with our <em>in vitro</em> findings in that lymphoid cells bound more tightly to the HA-glycocalyx in the lungs of influenza-infected mice than neutrophils and mononuclear phagocytes (MNPs). The neutrophils and MNPs were associated with a HC•HA matrix and were more readily lavaged from the lungs. In conclusion, this work shows increased IαI and HA accumulation and the formation of a HC•HA matrix in mouse lungs post-IAV infection. The formation of HA and HC•HA matrices could p","PeriodicalId":49851,"journal":{"name":"Matrix Biology","volume":"126 ","pages":"Pages 25-42"},"PeriodicalIF":6.9,"publicationDate":"2024-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139474576","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}