1
|
Stins MF, Mtaja A, Mulendele E, Mwimbe D, Pinilla-Monsalve GD, Mutengo M, Pardo CA, Chipeta J. Inflammation and Elevated Osteopontin in Plasma and CSF in Cerebral Malaria Compared to Plasmodium-Negative Neurological Infections. Int J Mol Sci 2024; 25:9620. [PMID: 39273566 PMCID: PMC11394774 DOI: 10.3390/ijms25179620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 08/26/2024] [Accepted: 09/01/2024] [Indexed: 09/15/2024] Open
Abstract
Cerebral malaria in young African children is associated with high mortality, and persisting neurological deficits often remain in survivors. Sequestered Plasmodium-infected red blood cells lead to cerebrovascular inflammation and subsequent neuroinflammation. Brain inflammation can play a role in the pathogenesis of neurologic sequelae. Therefore, we assessed a select set of proinflammatory analytes (IP10, IL23, MIP3α, GRO, MCP-1, and osteopontin in both the plasma and cerebrospinal fluid(CSF) of Zambian children with cerebral malaria and compared this with children with neurological symptoms that were negative for Plasmodium falciparum (non-cerebral malaria). Several similarities in plasma and CSF levels were found, as were some striking differences. We confirmed that IP10 levels were higher in the plasma of cerebral malaria patients, but this was not found in CSF. Levels of osteopontin were elevated in both the plasma and CSF of CM patients compared to the non-CM patients. These results show again a highly inflammatory environment in both groups but a different profile for CM when compared to non-cerebral malaria. Osteopontin may play an important role in neurological inflammation in CM and the resulting sequelae. Therefore, osteopontin could be a valid target for further biomarker research and potentially for therapeutic interventions in neuroinflammatory infections.
Collapse
Affiliation(s)
- Monique F. Stins
- Malaria Research Institute, Johns Hopkins School of Public Health, 615N Wolfe Street, Baltimore, MD 21205, USA
- Biomedical Research Institute of Southern California, Oceanside, CA 92046, USA
| | - Agnes Mtaja
- University Teaching Hospital Malaria Research Unit (SMUTH-MRU), Department of Pediatrics and Child Health, University of Zambia School of Medicine, Lusaka P.O. Box 50110, Zambia
| | - Evans Mulendele
- University Teaching Hospital Malaria Research Unit (SMUTH-MRU), Department of Pediatrics and Child Health, University of Zambia School of Medicine, Lusaka P.O. Box 50110, Zambia
| | - Daniel Mwimbe
- University Teaching Hospital Malaria Research Unit (SMUTH-MRU), Department of Pediatrics and Child Health, University of Zambia School of Medicine, Lusaka P.O. Box 50110, Zambia
| | - Gabriel D. Pinilla-Monsalve
- Division of Neuroimmunology and Neuroinfectious Diseases, Department of Neurology, Johns Hopkins School of Medicine, 600 N Wolfe Street, Baltimore, MD 21285, USA; (G.D.P.-M.); (C.A.P.)
- Department of Radiology, Faculty of Medicine, University of Montreal, 2900 Edouard Montpetit Blvd, Montreal, QC H3T 1J4, Canada
| | - Mable Mutengo
- University Teaching Hospital Malaria Research Unit (SMUTH-MRU), Department of Pediatrics and Child Health, University of Zambia School of Medicine, Lusaka P.O. Box 50110, Zambia
- Institute of Basic and Biomedical Sciences, Levy Mwanawasa Medical University, Lusaka P.O. Box 33991, Zambia
| | - Carlos A. Pardo
- Division of Neuroimmunology and Neuroinfectious Diseases, Department of Neurology, Johns Hopkins School of Medicine, 600 N Wolfe Street, Baltimore, MD 21285, USA; (G.D.P.-M.); (C.A.P.)
| | - James Chipeta
- University Teaching Hospital Malaria Research Unit (SMUTH-MRU), Department of Pediatrics and Child Health, University of Zambia School of Medicine, Lusaka P.O. Box 50110, Zambia
| |
Collapse
|
2
|
Pal D, Das P, Mukherjee P, Roy S, Chaudhuri S, Kesh SS, Ghosh D, Nandi SK. Biomaterials-Based Strategies to Enhance Angiogenesis in Diabetic Wound Healing. ACS Biomater Sci Eng 2024; 10:2725-2741. [PMID: 38630965 DOI: 10.1021/acsbiomaterials.4c00216] [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] [Indexed: 04/19/2024]
Abstract
Amidst the present healthcare issues, diabetes is unique as an emerging class of affliction with chronicity in a majority of the population. To check and control its effects, there have been huge turnover and constant development of management strategies, and though a bigger part of the health care area is involved in achieving its control and the related issues such as the effect of diabetes on wound healing and care and many of the works have reached certain successful outcomes, still there is a huge lack in managing it, with maximum effect yet to be attained. Studying pathophysiology and involvement of various treatment options, such as tissue engineering, application of hydrogels, drug delivery methods, and enhancing angiogenesis, are at constantly developing stages either direct or indirect. In this review, we have gathered a wide field of information and different new therapeutic methods and targets for the scientific community, paving the way toward more settled ideas and research advances to cure diabetic wounds and manage their outcomes.
Collapse
Affiliation(s)
- Debajyoti Pal
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal & Fishery Sciences, Kolkata 700037, India
| | - Pratik Das
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal & Fishery Sciences, Kolkata 700037, India
| | - Prasenjit Mukherjee
- Department of Veterinary Clinical Complex, West Bengal University of Animal & Fishery Sciences, Kolkata 700037, India
| | - Subhasis Roy
- Department of Veterinary Clinical Complex, West Bengal University of Animal & Fishery Sciences, Kolkata 700037, India
| | - Shubhamitra Chaudhuri
- Department of Veterinary Clinical Complex, West Bengal University of Animal & Fishery Sciences, Kolkata 700037, India
| | - Shyam Sundar Kesh
- Department of Veterinary Clinical Complex, West Bengal University of Animal & Fishery Sciences, Kolkata 700037, India
| | - Debaki Ghosh
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal & Fishery Sciences, Kolkata 700037, India
| | - Samit Kumar Nandi
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal & Fishery Sciences, Kolkata 700037, India
| |
Collapse
|
3
|
Abhinav K, Lee AG, Pendharkar AV, Bigder M, Bet A, Rosenberg-Hasson Y, Cheng MY, Steinberg GK. Comprehensive Profiling of Secreted Factors in the Cerebrospinal Fluid of Moyamoya Disease Patients. Transl Stroke Res 2024; 15:399-408. [PMID: 36745304 PMCID: PMC10891229 DOI: 10.1007/s12975-023-01135-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/27/2023] [Accepted: 01/30/2023] [Indexed: 02/07/2023]
Abstract
Moyamoya disease (MMD) is characterized by progressive occlusion of the intracranial internal carotid arteries, leading to ischemic and hemorrhagic events. Significant clinical differences exist between ischemic and hemorrhagic MMD. To understand the molecular profiles in the cerebrospinal fluid (CSF) of MMD patients, we investigated 62 secreted factors in both MMD subtypes (ischemic and hemorrhagic) and examined their relationship with preoperative perfusion status, the extent of postoperative angiographic revascularization, and functional outcomes. Intraoperative CSF was collected from 32 control and 71 MMD patients (37 ischemic and 34 hemorrhagic). Multiplex Luminex assay analysis showed that 41 molecules were significantly elevated in both MMD subtypes when compared to controls, including platelet-derived growth factor-BB (PDGF-BB), plasminogen activator inhibitor 1 (PAI-1), and intercellular adhesion molecule 1 (ICAM1) (p < 0.001). Many of these secreted proteins have not been previously reported in MMD, including interleukins (IL-2, IL-4, IL-5, IL-7, IL-8, IL-9, IL-17, IL-18, IL-22, and IL-23) and C-X-C motif chemokines (CXCL1 and CXCL9). Pathway analysis indicated that both MMD subtypes exhibited similar cellular/molecular functions and pathways, including cellular activation, migration, and inflammatory response. While neuroinflammation and dendritic cell pathways were activated in MMD patients, lipid signaling pathways involving nuclear receptors, peroxisome proliferator-activated receptor (PPAR), and liver X receptors (LXR)/retinoid X receptors (RXR) signaling were inhibited. IL-13 and IL-2 were negatively correlated with preoperative cerebral perfusion status, while 7 factors were positively correlated with the extent of postoperative revascularization. These elevated cytokines, chemokines, and growth factors in CSF may contribute to the pathogenesis of MMD and represent potential future therapeutic targets.
Collapse
Affiliation(s)
- Kumar Abhinav
- Department of Neurosurgery, Stanford University School of Medicine, 1201 Welch Road, MSLS P305, Stanford, CA, 94305, USA
- Stanford Stroke Center, Stanford University School of Medicine, Stanford, CA, USA
- Department of Neurosurgery, Bristol Institute of Clinical Neuroscience, Southmead Hospital, Bristol, UK
| | - Alex G Lee
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Arjun V Pendharkar
- Department of Neurosurgery, Stanford University School of Medicine, 1201 Welch Road, MSLS P305, Stanford, CA, 94305, USA
- Stanford Stroke Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Mark Bigder
- Department of Neurosurgery, Stanford University School of Medicine, 1201 Welch Road, MSLS P305, Stanford, CA, 94305, USA
- Stanford Stroke Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Anthony Bet
- Department of Neurosurgery, Stanford University School of Medicine, 1201 Welch Road, MSLS P305, Stanford, CA, 94305, USA
| | - Yael Rosenberg-Hasson
- Human Immune Monitoring Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Michelle Y Cheng
- Department of Neurosurgery, Stanford University School of Medicine, 1201 Welch Road, MSLS P305, Stanford, CA, 94305, USA
- Stanford Stroke Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Gary K Steinberg
- Department of Neurosurgery, Stanford University School of Medicine, 1201 Welch Road, MSLS P305, Stanford, CA, 94305, USA.
- Stanford Stroke Center, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
4
|
Nawaz S, Kulyar MFEA, Mo Q, Yao W, Iqbal M, Li J. Homeostatic Regulation of Pro-Angiogenic and Anti-Angiogenic Proteins via Hedgehog, Notch Grid, and Ephrin Signaling in Tibial Dyschondroplasia. Animals (Basel) 2023; 13:3750. [PMID: 38136788 PMCID: PMC10740744 DOI: 10.3390/ani13243750] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/21/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
Precise coupling of two fundamental mechanisms, chondrogenesis and osteogenesis via angiogenesis, plays a crucial role during rapid proliferation of growth plates, and alteration in their balance might lead to pathogenic conditions. Tibial dyschondroplasia (TD) is characterized by an avascular, non-mineralized, jade-white "cartilaginous wedge" with impaired endochondral ossification and chondrocyte proliferation at the proximal end of a tibial bone in rapidly growing poultry birds. Developing vascular structures are dynamic with cartilage growth and are regulated through homeostatic balance among pro and anti-angiogenic proteins and cytokines. Pro-angiogenic factors involves a wide spectrum of multifactorial mitogens, such as vascular endothelial growth factors (VEGF), platelet-derived growth factors (PDGF), basic fibroblast growth factor (bFGF), placental growth factors, transforming growth factor-β (TGF-β), and TNF-α. Considering their regulatory role via the sonic hedgehog, notch-gridlock, and ephrin-B2/EphB4 pathways and inhibition through anti-angiogenic proteins like angiostatin, endostatin, decoy receptors, vasoinhibin, thrombospondin, PEX, and troponin, their possible role in persisting inflammatory conditions like TD was studied in the current literature review. Balanced apoptosis and angiogenesis are vital for physiological bone growth. Any homeostatic imbalance among apoptotic, angiogenetic, pro-angiogenic, or anti-angiogenic proteins ultimately leads to pathological bone conditions like TD and osteoarthritis. The current review might substantiate solid grounds for developing innovative therapeutics for diseases governed by the disproportion of angiogenesis and anti-angiogenesis proteins.
Collapse
Affiliation(s)
- Shah Nawaz
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (S.N.); (M.F.-e.-A.K.); (W.Y.); (M.I.)
| | - Muhammad Fakhar-e-Alam Kulyar
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (S.N.); (M.F.-e.-A.K.); (W.Y.); (M.I.)
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Quan Mo
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (S.N.); (M.F.-e.-A.K.); (W.Y.); (M.I.)
| | - Wangyuan Yao
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (S.N.); (M.F.-e.-A.K.); (W.Y.); (M.I.)
| | - Mudassar Iqbal
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (S.N.); (M.F.-e.-A.K.); (W.Y.); (M.I.)
- Faculty of Veterinary and Animal Sciences, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Jiakui Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (S.N.); (M.F.-e.-A.K.); (W.Y.); (M.I.)
| |
Collapse
|
5
|
Lei XX, Zou CY, Hu JJ, Jiang YL, Zhang XZ, Zhao LM, He T, Zhang QY, Li YX, Li-Ling J, Xie HQ. Click-crosslinked in-situ hydrogel improves the therapeutic effect in wound infections through antibacterial, antioxidant and anti-inflammatory activities. CHEMICAL ENGINEERING JOURNAL 2023; 461:142092. [DOI: 10.1016/j.cej.2023.142092] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/25/2024]
|
6
|
Muller AJ, Mondal A, Dey S, Prendergast GC. IDO1 and inflammatory neovascularization: bringing new blood to tumor-promoting inflammation. Front Oncol 2023; 13:1165298. [PMID: 37182174 PMCID: PMC10172587 DOI: 10.3389/fonc.2023.1165298] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/12/2023] [Indexed: 05/16/2023] Open
Abstract
In parallel with the genetic and epigenetic changes that accumulate in tumor cells, chronic tumor-promoting inflammation establishes a local microenvironment that fosters the development of malignancy. While knowledge of the specific factors that distinguish tumor-promoting from non-tumor-promoting inflammation remains inchoate, nevertheless, as highlighted in this series on the 'Hallmarks of Cancer', it is clear that tumor-promoting inflammation is essential to neoplasia and metastatic progression making identification of specific factors critical. Studies of immunometabolism and inflamometabolism have revealed a role for the tryptophan catabolizing enzyme IDO1 as a core element in tumor-promoting inflammation. At one level, IDO1 expression promotes immune tolerance to tumor antigens, thereby helping tumors evade adaptive immune control. Additionally, recent findings indicate that IDO1 also promotes tumor neovascularization by subverting local innate immunity. This newly recognized function for IDO1 is mediated by a unique myeloid cell population termed IDVCs (IDO1-dependent vascularizing cells). Initially identified in metastatic lesions, IDVCs may exert broader effects on pathologic neovascularization in various disease settings. Mechanistically, induction of IDO1 expression in IDVCs by the inflammatory cytokine IFNγ blocks the antagonistic effect of IFNγ on neovascularization by stimulating the expression of IL6, a powerful pro-angiogenic cytokine. By contributing to vascular access, this newly ascribed function for IDO1 aligns with its involvement in other cancer hallmark functionalities, (tumor-promoting inflammation, immune escape, altered cellular metabolism, metastasis), which may stem from an underlying role in normal physiological functions such as wound healing and pregnancy. Understanding the nuances of how IDO1 involvement in these cancer hallmark functionalities varies between different tumor settings will be crucial to the future development of successful IDO1-directed therapies.
Collapse
Affiliation(s)
- Alexander J. Muller
- Lankenau Institute for Medical Research, Wynnewood, PA, United States
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
- *Correspondence: Alexander J. Muller,
| | - Arpita Mondal
- Arbutus Biopharma, Inc., Warminster, PA, United States
| | - Souvik Dey
- Wuxi Advanced Therapeutics, Inc., Philadelphia, PA, United States
| | - George C. Prendergast
- Lankenau Institute for Medical Research, Wynnewood, PA, United States
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
7
|
de Almeida BM, dos Santos IDD, de Carvalho FMA, Correa LC, Cunha JLS, Dariva C, Severino P, Cardoso JC, Souto EB, de Albuquerque-Júnior RLC. Himatanthus bracteatus-Composed In Situ Polymerizable Hydrogel for Wound Healing. Int J Mol Sci 2022; 23:ijms232315176. [PMID: 36499503 PMCID: PMC9739771 DOI: 10.3390/ijms232315176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/27/2022] [Accepted: 10/18/2022] [Indexed: 12/12/2022] Open
Abstract
The Himatanthus genus presents anti-inflammatory, antioxidant activities, suggesting potential wound-healing properties. This study aimed to develop and analyze the wound-healing properties of a photopolymerizable gelatin-based hydrogel (GelMA) containing an ethanolic extract of Himatanthus bracteatus in a murine model. The extract was obtained under high pressure conditions, incorporated (2%) into the GelMA (GelMA-HB), and physically characterized. The anti-inflammatory activity of the extract was assessed using a carrageenan-induced pleurisy model and the GelMA-HB scarring properties in a wound-healing assay. The extract reduced IL-1β and TNF-α levels (48.5 ± 6.7 and 64.1 ± 4.9 pg/mL) compared to the vehicle (94.4 ± 2.3 pg/mL and 106.3 ± 5.7 pg/mL; p < 0.001). GelMA-HB depicted significantly lower swelling and increased resistance to mechanical compression compared to GelMA (p < 0.05). GelMA-HB accelerated wound closure over the time course of the experiment (p < 0.05) and promoted a significantly greater peak of myofibroblast differentiation (36.1 ± 6.6 cells) and microvascular density (23.1 ± 0.7 microvessels) on day 7 in comparison to GelMA (31.9 ± 5.3 cells and 20.2 ± 0.6 microvessels) and the control (25.8 ± 4.6 cells and 17.5 ± 0.5 microvessels) (p < 0.05). In conclusion, GelMA-HB improved wound healing in rodents, probably by modulating the inflammatory response and myofibroblastic and microvascular differentiation.
Collapse
Affiliation(s)
- Bernadeth M. de Almeida
- Biotechnological Postgraduate Program—RENORBIO, Tiradentes University, Aracaju 49010-390, SE, Brazil
| | | | - Felipe M. A. de Carvalho
- Postgraduate Program in Health and Environment, Tiradentes University, Aracaju 49032-490, SE, Brazil
| | - Luana C. Correa
- School of Physiotherapy, Tiradentes University, Aracaju 49032-490, SE, Brazil
| | - John L. S. Cunha
- Department of Odontology, Paraiba State University, Campina Grande 58429 500, PB, Brazil
| | - Claudio Dariva
- Laboratory for Colloidal Systems Studies, Institute of Technology and Research (ITP), Tiradentes University, Aracaju 49010-390, SE, Brazil
| | - Patricia Severino
- Biotechnological Postgraduate Program—RENORBIO, Tiradentes University, Aracaju 49010-390, SE, Brazil
| | - Juliana C. Cardoso
- Biotechnological Postgraduate Program—RENORBIO, Tiradentes University, Aracaju 49010-390, SE, Brazil
- Postgraduate Program in Health and Environment, Tiradentes University, Aracaju 49032-490, SE, Brazil
| | - Eliana B. Souto
- Biotechnological Postgraduate Program—RENORBIO, Tiradentes University, Aracaju 49010-390, SE, Brazil
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, nº. 228, 4050-313 Porto, Portugal
- REQUIMTE/UCIBIO, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, nº. 228, 4050-313 Porto, Portugal
- Correspondence: (E.B.S.); (R.L.C.d.A.-J.)
| | | |
Collapse
|
8
|
Li S, Shi S, Xia F, Luo B, Ha Y, Luisi J, Gupta PK, Merkley KH, Motamedi M, Liu H, Zhang W. CXCR3 deletion aggravates corneal neovascularization in a corneal alkali-burn model. Exp Eye Res 2022; 225:109265. [PMID: 36206861 PMCID: PMC10191246 DOI: 10.1016/j.exer.2022.109265] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/25/2022] [Accepted: 09/21/2022] [Indexed: 01/16/2023]
Abstract
Corneal neovascularization can cause devastating consequences including vision impairment and even blindness. Corneal inflammation is a crucial factor for the induction of corneal neovascularization. Current anti-inflammatory approaches are of limited value with poor therapeutic effects. Therefore, there is an urgent need to develop new therapies that specifically modulate inflammatory pathways and inhibit neovascularization in the cornea. The interaction of chemokines and their receptors plays a key role in regulating leukocyte migration during inflammatory response. CXCR3 is essential for mediating the recruitment of activated T cells and microglia/macrophages, but the role of CXCR3 in the initiation and promotion of corneal neovascularization remains unclear. Here, we showed that the expression of CXCL10 and CXCR3 was significantly increased in the cornea after alkali burn. Compared with WT mice, CXCR3-/- mice exhibited significantly increased corneal hemangiogenesis and lymphangiogenesis after alkali burn. In addition, exaggerated leukocyte infiltration and leukostasis, and elevated expression of inflammatory cytokines and angiogenic factor were also found in the corneas of CXCR3-/- mice subjected to alkali burn. With bone marrow (BM) transplantation, we further demonstrated that the deletion of CXCR3 in BM-derived leukocytes plays a key role in the acceleration of alkali burn-induced corneal neovascularization. Taken together, our results suggest that upregulation of CXCR3 does not exhibit its conventional action as a proinflammatory cytokine but instead serves as a self-protective mechanism for the modulation of inflammation and maintenance of corneal avascularity after corneal alkali burn.
Collapse
Affiliation(s)
- Shengguo Li
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Shuizhen Shi
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Fan Xia
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Ban Luo
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Yonju Ha
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Jonathan Luisi
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA; Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Praveena K Gupta
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Kevin H Merkley
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Massoud Motamedi
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Hua Liu
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA.
| | - Wenbo Zhang
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA; Departments of Neuroscience, Cell Biology & Anatomy, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
9
|
Wells A. Role of CXCR3 in fibrotic tissue responses. Int J Biochem Cell Biol 2022; 152:106311. [PMID: 36195287 DOI: 10.1016/j.biocel.2022.106311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 09/22/2022] [Accepted: 09/28/2022] [Indexed: 11/20/2022]
Abstract
Development of fibrosis leads to end stage diseases that defy treatments across all organs. This ensues as chronic inflammation is not dampened by physiologic processes that issue in the resolution phase of wound healing. Thus, these conditions can be considered diseases of "failure to heal". In the absence of broadly viable treatments, it is proposed to examine key switches in wound healing resolution to seek insights into novel approaches. Signaling through the GPCR CXCR3 has been shown to be one such critical player in this physiologic transition that limits and even reverses early fibrosis. As such, a number of investigators and early stage technology companies have posited that triggering this signaling network would limit fibrosis. While there are some conflicting results, a consensus is emerging that pharmacologic interventions that promote signaling through this pathway represent innovative ways to limit fibrotic diseases.
Collapse
Affiliation(s)
- Alan Wells
- Departments of Pathology, Bioengineering, and Computational & Systems Biology, and McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; R&D Service, Pittsburgh VA Health System, Pittsburgh, PA 15213, USA.
| |
Collapse
|
10
|
Tregub PP, Averchuk AS, Baranich TI, Ryazanova MV, Salmina AB. Physiological and Pathological Remodeling of Cerebral Microvessels. Int J Mol Sci 2022; 23:12683. [PMID: 36293539 PMCID: PMC9603917 DOI: 10.3390/ijms232012683] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/10/2022] [Accepted: 10/18/2022] [Indexed: 11/13/2022] Open
Abstract
There is growing evidence that the remodeling of cerebral microvessels plays an important role in plastic changes in the brain associated with development, experience, learning, and memory consolidation. At the same time, abnormal neoangiogenesis, and deregulated regulation of microvascular regression, or pruning, could contribute to the pathogenesis of neurodevelopmental diseases, stroke, and neurodegeneration. Aberrant remodeling of microvesselsis associated with blood-brain barrier breakdown, development of neuroinflammation, inadequate microcirculation in active brain regions, and leads to the dysfunction of the neurovascular unit and progressive neurological deficits. In this review, we summarize current data on the mechanisms of blood vessel regression and pruning in brain plasticity and in Alzheimer's-type neurodegeneration. We discuss some novel approaches to modulating cerebral remodeling and preventing degeneration-coupled aberrant microvascular activity in chronic neurodegeneration.
Collapse
Affiliation(s)
- Pavel P. Tregub
- Federal State Budgetary Scientific Institution Research Center of Neurology, 125367 Moscow, Russia
| | | | | | | | | |
Collapse
|
11
|
Abstract
Angiogenesis, or the growth of new blood vessels from the preexisting vasculature, is a visible and important component of wound repair. When tissue damage occurs, disruption of the vasculature structure leads to hypoxia. The restoration of normoxia is essential for appropriate and durable tissue repair. Angiogenesis in wounds is regulated by endogenous proangiogenic mediators, which cause rapid growth of a new vascular bed that is much denser than that of normal tissue. Such rapid growth of the capillary bed results in capillaries that are abnormal, and the newly formed vessels are tortuous, dilated, and immature. During wound resolution, this substantial neocapillary bed is pruned back to normal density with attendant maturation. Many poorly healing wounds, including nonhealing ulcers and scars, exhibit an aberrant angiogenic response. The fine-tuning of capillary regrowth in wounds is an area of significant therapeutic potential.
Collapse
Affiliation(s)
- Chen Han
- Center for Wound Healing and Tissue Regeneration, Colleges of Dentistry and Medicine, University of Illinois Chicago, Chicago, Illinois 60612, USA
| | - May Barakat
- Center for Wound Healing and Tissue Regeneration, Colleges of Dentistry and Medicine, University of Illinois Chicago, Chicago, Illinois 60612, USA
| | - Luisa A DiPietro
- Center for Wound Healing and Tissue Regeneration, Colleges of Dentistry and Medicine, University of Illinois Chicago, Chicago, Illinois 60612, USA
| |
Collapse
|
12
|
Masood F, Bhattaram R, Rosenblatt MI, Kazlauskas A, Chang JH, Azar DT. Lymphatic Vessel Regression and Its Therapeutic Applications: Learning From Principles of Blood Vessel Regression. Front Physiol 2022; 13:846936. [PMID: 35392370 PMCID: PMC8980686 DOI: 10.3389/fphys.2022.846936] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/25/2022] [Indexed: 02/03/2023] Open
Abstract
Aberrant lymphatic system function has been increasingly implicated in pathologies such as lymphedema, organ transplant rejection, cardiovascular disease, obesity, and neurodegenerative diseases including Alzheimer's disease and Parkinson's disease. While some pathologies are exacerbated by lymphatic vessel regression and dysfunction, induced lymphatic regression could be therapeutically beneficial in others. Despite its importance, our understanding of lymphatic vessel regression is far behind that of blood vessel regression. Herein, we review the current understanding of blood vessel regression to identify several hallmarks of this phenomenon that can be extended to further our understanding of lymphatic vessel regression. We also summarize current research on lymphatic vessel regression and an array of research tools and models that can be utilized to advance this field. Additionally, we discuss the roles of lymphatic vessel regression and dysfunction in select pathologies, highlighting how an improved understanding of lymphatic vessel regression may yield therapeutic insights for these disease states.
Collapse
|
13
|
T cell apoptosis characterizes severe Covid-19 disease. Cell Death Differ 2022; 29:1486-1499. [PMID: 35066575 PMCID: PMC8782710 DOI: 10.1038/s41418-022-00936-x] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 01/06/2022] [Accepted: 01/10/2022] [Indexed: 02/02/2023] Open
Abstract
Severe SARS-CoV-2 infections are characterized by lymphopenia, but the mechanisms involved are still elusive. Based on our knowledge of HIV pathophysiology, we hypothesized that SARS-CoV-2 infection-mediated lymphopenia could also be related to T cell apoptosis. By comparing intensive care unit (ICU) and non-ICU COVID-19 patients with age-matched healthy donors, we found a strong positive correlation between plasma levels of soluble FasL (sFasL) and T cell surface expression of Fas/CD95 with the propensity of T cells to die and CD4 T cell counts. Plasma levels of sFasL and T cell death are correlated with CXCL10 which is part of the signature of 4 biomarkers of disease severity (ROC, 0.98). We also found that members of the Bcl-2 family had modulated in the T cells of COVID-19 patients. More importantly, we demonstrated that the pan-caspase inhibitor, Q-VD, prevents T cell death by apoptosis and enhances Th1 transcripts. Altogether, our results are compatible with a model in which T-cell apoptosis accounts for T lymphopenia in individuals with severe COVID-19. Therefore, a strategy aimed at blocking caspase activation could be beneficial for preventing immunodeficiency in COVID-19 patients.
Collapse
|
14
|
Al-Shammari AM, Al-Mudhafr MA, Chalap Al- Grawi ED, Al-Hili ZA, Yaseen N. Newcastle disease virus suppresses angiogenesis in mammary adenocarcinoma models. BULGARIAN JOURNAL OF VETERINARY MEDICINE 2022. [DOI: 10.15547/bjvm.2020-0019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Cancer cells heavily utilise angiogenesis process to increase vascularisation for tumour mass growth and spread, so targeting this process is important to create an effective therapy. The AMHA1 strain of Newcastle disease virus (NDV) is an RNA virus with natural oncotropism. NDV induces direct tumour cytolysis, apoptosis, and immune stimulation. This work aimed to test NDV anti-angiogenic activity in a breast cancer model. To evaluate NDV’s antitumour effect in vivo, NDV was tested against mammary adenocarcinoma AN3 transplanted in syngeneic immunocompetent mice. In vivo antiangiogenic activity was evaluated by quantifying the blood vessels in treated and control tumour sections. In vitro experiments that exposed AMN3 mammary adenocarcinoma cells and Hep-2 laryngeal carcinoma cells to NDV at different time intervals were performed to identify the exact mechanism of anti-angiogenesis by using angiogenesis microarray slides. In vivo results showed significant tumour regression and significant decrease in blood vessel formation in treated tumour sections. The in vitro microarray analysis of 14 different angiogenesis factors revealed that NDV downregulated angiopoietin-1, angiopoietin-2, and epidermal growth factor in mammary adenocarcinoma cells. However, NDV elicited a different effect on Hep-2 as represented by the downregulation of inducible protein 10, intracellular adhesion molecule-1, and basic fibroblast growth factor beta in NDV-infected tumour cells. It was found out that microarray analysis results helped interpret the in vivo data. The results suggested that the NDV oncolytic strain reduced angiogenesis by interfering with angiogenesis factors that might reduce tumour cell proliferation, infiltration, and invasion.
Collapse
Affiliation(s)
- A. M. Al-Shammari
- University of Mustansiriyah, Iraqi Center for Cancer and Medical Genetic Research, Experimental Therapy Department, Baghdad, Iraq
| | - M. A. Al-Mudhafr
- University of Kufa, Faculty of Veterinary Medicine, Department of Microbiology
| | | | - Z. A. Al-Hili
- University of Mustansiriyah, Iraqi Center for Cancer and Medical Genetic Research, Experimental Therapy Department, Baghdad, Iraq
| | - N. Yaseen
- University of Mustansiriyah, Iraqi Center for Cancer and Medical Genetic Research, Experimental Therapy Department, Baghdad, Iraq
| |
Collapse
|
15
|
Qiao X, Zhang W, Zhao W. Role of CXCL10 in Spinal Cord Injury. Int J Med Sci 2022; 19:2058-2070. [PMID: 36483597 PMCID: PMC9724238 DOI: 10.7150/ijms.76694] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 11/03/2022] [Indexed: 11/24/2022] Open
Abstract
Spinal cord injury (SCI) results in acute inflammatory responses and secondary damages, including neuronal and glial cell death, axonal damage and demyelination, and blood-brain barrier (BBB) damage, eventually leading to neuronal dysfunction and other complications. C-X-C motif Chemokine Ligand 10 (CXCL10) is expressed after the injury, playing multiple roles in the development and progression of SCI. Moreover, the CXCL10 antagonist can restrict inflammatory immune responses and promote neuronal regeneration and functional recovery. In this review, we summarize the structure and biological functions of CXCL10, and the roles of the CXCL10 / CXCR3 axis in acute inflammatory responses, secondary damages, and complications during SCI, thus providing a potential theoretical basis by highlighting CXCL10 as a new potential drug target for the treatment of SCI.
Collapse
Affiliation(s)
- Xinyu Qiao
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Wei Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, China.,Department of Pathogen Biology, Guizhou Nursing Vocational College, Guiyang, China
| | - Weijiang Zhao
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, China.,Cell Biology Department, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, China
| |
Collapse
|
16
|
Wang P, Zhao Y, Wang J, Wu Z, Sui B, Mao X, Shi S, Kou X. Dephosphorylation of Caveolin-1 Controls C-X-C Motif Chemokine Ligand 10 Secretion in Mesenchymal Stem Cells to Regulate the Process of Wound Healing. Front Cell Dev Biol 2021; 9:725630. [PMID: 34790658 PMCID: PMC8592036 DOI: 10.3389/fcell.2021.725630] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 10/11/2021] [Indexed: 12/24/2022] Open
Abstract
Mesenchymal stem cells (MSCs) secrete cytokines in a paracrine or autocrine manner to regulate immune response and tissue regeneration. Our previous research revealed that MSCs use the complex of Fas/Fas-associated phosphatase-1 (Fap-1)/caveolin-1 (Cav-1) mediated exocytotic process to regulate cytokine and small extracellular vesicles (EVs) secretion, which contributes to accelerated wound healing. However, the detailed underlying mechanism of cytokine secretion controlled by Cav-1 remains to be explored. We show that Gingiva-derived MSCs (GMSCs) could secrete more C-X-C motif chemokine ligand 10 (CXCL10) but showed lower phospho-Cav-1 (p-Cav-1) expression than skin-derived MSCs (SMSCs). Moreover, dephosphorylation of Cav-1 by a Src kinase inhibitor PP2 significantly enhances CXCL10 secretion, while activating phosphorylation of Cav-1 by H2O2 restraints CXCL10 secretion in GMSCs. We also found that Fas and Fap-1 contribute to the dephosphorylation of Cav-1 to elevate CXCL10 secretion. Tumor necrosis factor-α serves as an activator to up-regulate Fas, Fap-1, and down-regulate p-Cav-1 expression to promote CXCL10 release. Furthermore, local applying p-Cav-1 inhibitor PP2 could accelerate wound healing, reduce the expression of α-smooth muscle actin and increase cleaved-caspase 3 expression. These results indicated that dephosphorylation of Cav-1 could inhibit fibrosis during wound healing. The present study establishes a previously unknown role of p-Cav-1 in controlling cytokine release of MSC and may present a potential therapeutic approach for promoting scarless wound healing.
Collapse
Affiliation(s)
- Panpan Wang
- South China Center of Craniofacial Stem Cell Research, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Yingji Zhao
- South China Center of Craniofacial Stem Cell Research, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Juan Wang
- South China Center of Craniofacial Stem Cell Research, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Zhiying Wu
- Department of Microbiology, Zhongshan School of Medicine, Key Laboratory for Tropical Diseases Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Bingdong Sui
- South China Center of Craniofacial Stem Cell Research, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.,Department of Microbiology, Zhongshan School of Medicine, Key Laboratory for Tropical Diseases Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Xueli Mao
- South China Center of Craniofacial Stem Cell Research, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.,Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, China
| | - Songtao Shi
- South China Center of Craniofacial Stem Cell Research, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.,Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, China
| | - Xiaoxing Kou
- South China Center of Craniofacial Stem Cell Research, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.,Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, China
| |
Collapse
|
17
|
Majumder N, Velayutham M, Bitounis D, Kodali VK, Hasan Mazumder MH, Amedro J, Khramtsov VV, Erdely A, Nurkiewicz T, Demokritou P, Kelley EE, Hussain S. Oxidized carbon black nanoparticles induce endothelial damage through C-X-C chemokine receptor 3-mediated pathway. Redox Biol 2021; 47:102161. [PMID: 34624601 PMCID: PMC8502956 DOI: 10.1016/j.redox.2021.102161] [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] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/27/2021] [Accepted: 10/02/2021] [Indexed: 01/19/2023] Open
Abstract
Oxidation of engineered nanomaterials during application in various industrial sectors can alter their toxicity. Oxidized nanomaterials also have widespread industrial and biomedical applications. In this study, we evaluated the cardiopulmonary hazard posed by these nanomaterials using oxidized carbon black (CB) nanoparticles (CBox) as a model particle. Particle surface chemistry was characterized by X-ray photo electron spectroscopy (XPS) and Fourier-transform infrared spectroscopy (FTIR). Colloidal characterization and in vitro dosimetry modeling (particle kinetics, fate and transport modeling) were performed. Lung inflammation was assessed following oropharyngeal aspiration of CB or oxidized CBox particles (20 μg per mouse) in C57BL/6J mice. Toxicity and functional assays were also performed on murine macrophage (RAW 264.7) and endothelial cell lines (C166) with and without pharmacological inhibitors. Oxidant generation was assessed by electron paramagnetic resonance spectroscopy (EPR) and via flow cytometry. Endothelial toxicity was evaluated by quantifying pro-inflammatory mRNA expression, monolayer permeability, and wound closure. XPS and FTIR spectra indicated surface modifications, the appearance of new functionalities, and greater oxidative potential (both acellular and in vitro) of CBox particles. Treatment with CBox demonstrated greater in vivo inflammatory potentials (lavage neutrophil counts, secreted cytokine, and lung tissue mRNA expression) and air-blood barrier disruption (lavage proteins). Oxidant-dependent pro-inflammatory signaling in macrophages led to the production of CXCR3 ligands (CXCL9,10,11). Conditioned medium from CBox-treated macrophages induced significant elevation in endothelial cell pro-inflammatory mRNA expression, enhanced monolayer permeability and impairment of scratch healing in CXCR3 dependent manner. In summary, this study mechanistically demonstrated an increased biological potency of CBox particles and established the role of macrophage-released chemical mediators in endothelial damage.
Collapse
Affiliation(s)
- Nairrita Majumder
- Department of Physiology and Pharmacology, West Virginia University, School of Medicine, USA; Center for Inhalation Toxicology (iTOX), West Virginia University, School of Medicine, USA
| | - Murugesan Velayutham
- Center for Inhalation Toxicology (iTOX), West Virginia University, School of Medicine, USA; Department of Biochemistry, West Virginia University, School of Medicine, USA
| | - Dimitrios Bitounis
- Center for Nanotechnology and Nanotoxicology, Department of Environmental Health, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Vamsi K Kodali
- Department of Physiology and Pharmacology, West Virginia University, School of Medicine, USA; Center for Inhalation Toxicology (iTOX), West Virginia University, School of Medicine, USA; National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Md Habibul Hasan Mazumder
- Department of Physiology and Pharmacology, West Virginia University, School of Medicine, USA; Center for Inhalation Toxicology (iTOX), West Virginia University, School of Medicine, USA
| | - Jessica Amedro
- Department of Physiology and Pharmacology, West Virginia University, School of Medicine, USA; Center for Inhalation Toxicology (iTOX), West Virginia University, School of Medicine, USA
| | - Valery V Khramtsov
- Department of Biochemistry, West Virginia University, School of Medicine, USA
| | - Aaron Erdely
- Department of Physiology and Pharmacology, West Virginia University, School of Medicine, USA; Center for Inhalation Toxicology (iTOX), West Virginia University, School of Medicine, USA; National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Timothy Nurkiewicz
- Department of Physiology and Pharmacology, West Virginia University, School of Medicine, USA; Center for Inhalation Toxicology (iTOX), West Virginia University, School of Medicine, USA; National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Philip Demokritou
- Center for Nanotechnology and Nanotoxicology, Department of Environmental Health, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Eric E Kelley
- Department of Physiology and Pharmacology, West Virginia University, School of Medicine, USA; Center for Inhalation Toxicology (iTOX), West Virginia University, School of Medicine, USA
| | - Salik Hussain
- Department of Physiology and Pharmacology, West Virginia University, School of Medicine, USA; Center for Inhalation Toxicology (iTOX), West Virginia University, School of Medicine, USA.
| |
Collapse
|
18
|
The Involvement of CXC Motif Chemokine Ligand 10 (CXCL10) and Its Related Chemokines in the Pathogenesis of Coronary Artery Disease and in the COVID-19 Vaccination: A Narrative Review. Vaccines (Basel) 2021; 9:vaccines9111224. [PMID: 34835155 PMCID: PMC8623875 DOI: 10.3390/vaccines9111224] [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] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 10/16/2021] [Accepted: 10/18/2021] [Indexed: 12/11/2022] Open
Abstract
Coronary artery disease (CAD) and coronary heart disease (CHD) constitute two of the leading causes of death in Europe, USA and the rest of the world. According to the latest reports of the Iranian National Health Ministry, CAD is the main cause of death in Iranian patients with an age over 35 years despite a significant reduction in mortality due to early interventional treatments in the context of an acute coronary syndrome (ACS). Inflammation plays a fundamental role in coronary atherogenesis, atherosclerotic plaque formation, acute coronary thrombosis and CAD establishment. Chemokines are well-recognized mediators of inflammation involved in several bio-functions such as leucocyte migration in response to inflammatory signals and oxidative vascular injury. Different chemokines serve as chemo-attractants for a wide variety of cell types including immune cells. CXC motif chemokine ligand 10 (CXCL10), also known as interferon gamma-induced protein 10 (IP-10/CXLC10), is a chemokine with inflammatory features whereas CXC chemokine receptor 3 (CXCR3) serves as a shared receptor for CXCL9, 10 and 11. These chemokines mediate immune responses through the activation and recruitment of leukocytes, eosinophils, monocytes and natural killer (NK) cells. CXCL10, interleukin (IL-15) and interferon (IFN-g) are increased after a COVID-19 vaccination with a BNT162b2 mRNA (Pfizer/BioNTech) vaccine and are enriched by tumor necrosis factor alpha (TNF-α) and IL-6 after the second vaccination. The aim of the present study is the presentation of the elucidation of the crucial role of CXCL10 in the patho-physiology and pathogenesis of CAD and in identifying markers associated with the vaccination resulting in antibody development.
Collapse
|
19
|
From remodeling to quiescence: The transformation of the vascular network. Cells Dev 2021; 168:203735. [PMID: 34425253 DOI: 10.1016/j.cdev.2021.203735] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 07/14/2021] [Accepted: 08/16/2021] [Indexed: 12/15/2022]
Abstract
The vascular system is essential for embryogenesis, healing, and homeostasis. Dysfunction or deregulated blood vessel function contributes to multiple diseases, including diabetic retinopathy, cancer, hypertension, or vascular malformations. A balance between the formation of new blood vessels, vascular remodeling, and vessel quiescence is fundamental for tissue growth and function. Whilst the major mechanisms contributing to the formation of new blood vessels have been well explored in recent years, vascular remodeling and quiescence remain poorly understood. In this review, we highlight the cellular and molecular mechanisms responsible for vessel remodeling and quiescence during angiogenesis. We further underline how impaired remodeling and/or destabilization of vessel networks can contribute to vascular pathologies. Finally, we speculate how addressing the molecular mechanisms of vascular remodeling and stabilization could help to treat vascular-related disorders.
Collapse
|
20
|
Groblewska M, Mroczko B. Pro- and Antiangiogenic Factors in Gliomas: Implications for Novel Therapeutic Possibilities. Int J Mol Sci 2021; 22:ijms22116126. [PMID: 34200145 PMCID: PMC8201226 DOI: 10.3390/ijms22116126] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/01/2021] [Accepted: 06/03/2021] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis, a complex, multistep process of forming new blood vessels, plays crucial role in normal development, embryogenesis, and wound healing. Malignant tumors characterized by increased proliferation also require new vasculature to provide an adequate supply of oxygen and nutrients for developing tumor. Gliomas are among the most frequent primary tumors of the central nervous system (CNS), characterized by increased new vessel formation. The processes of neoangiogenesis, necessary for glioma development, are mediated by numerous growth factors, cytokines, chemokines and other proteins. In contrast to other solid tumors, some biological conditions, such as the blood–brain barrier and the unique interplay between immune microenvironment and tumor, represent significant challenges in glioma therapy. Therefore, the objective of the study was to present the role of various proangiogenic factors in glioma angiogenesis as well as the differences between normal and tumoral angiogenesis. Another goal was to present novel therapeutic options in oncology approaches. We performed a thorough search via the PubMed database. In this paper we describe various proangiogenic factors in glioma vasculature development. The presented paper also reviews various antiangiogenic factors necessary in maintaining equilibrium between pro- and antiangiogenic processes. Furthermore, we present some novel possibilities of antiangiogenic therapy in this type of tumors.
Collapse
Affiliation(s)
- Magdalena Groblewska
- Department of Biochemical Diagnostics, University Hospital in Białystok, 15-269 Białystok, Poland;
| | - Barbara Mroczko
- Department of Biochemical Diagnostics, University Hospital in Białystok, 15-269 Białystok, Poland;
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, 15-269 Białystok, Poland
- Correspondence: ; Tel.: +48-858318785
| |
Collapse
|
21
|
Coperchini F, Chiovato L, Ricci G, Croce L, Magri F, Rotondi M. The cytokine storm in COVID-19: Further advances in our understanding the role of specific chemokines involved. Cytokine Growth Factor Rev 2021; 58:82-91. [PMID: 33573850 PMCID: PMC7837329 DOI: 10.1016/j.cytogfr.2020.12.005] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/21/2020] [Accepted: 12/30/2020] [Indexed: 12/11/2022]
Abstract
SARS-COV-2 infection represents the greatest pandemic of the world, counting daily increasing number of subjects positive to the virus and, sadly, increasing number of deaths. Current studies reported that the cytokine/chemokine network is crucial in the onset and maintenance of the "cytokine storm", the event occurring in those patients in whom the progression of COVID-19 will progress, in most cases, to a very severe and potentially threatening disease. Detecting a possible "immune signature" in patients, as assessed by chemokines status in patients with COVID-19, could be helpful for individual risk stratification for developing a more or less severe clinical course of the disease. The present review is specifically aimed at overviewing current evidences provided by in vitro and in vivo studies addressing the issue of which chemokines seems to be involved, at least at present, in COVID-19. Currently available experimental and clinical studies regarding those chemokines more deeply studied in COVID-19, with a specific focus on their role in the cytokine storm and ultimately with their ability to predict the clinical course of the disease, will be taken into account. Moreover, similarities and differences between chemokines and cytokines, which both contribute to the onset of the pro-inflammatory loop characterizing SARS-COV-2 infection, will be briefly discussed. Future studies will rapidly accumulate in the next months and their results will hopefully provide more insights as to the complex physiopathology of COVID-19-related cytokine storm. This will likely make the present review somehow "dated" in a short time, but still the present review provides an overview of the scenario of the current knowledge on this topic.
Collapse
Affiliation(s)
- Francesca Coperchini
- Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, 27100 Pavia PV, Italy
| | - Luca Chiovato
- Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, 27100 Pavia PV, Italy; Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia PV, Italy
| | - Gianluca Ricci
- Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, 27100 Pavia PV, Italy
| | - Laura Croce
- Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, 27100 Pavia PV, Italy; Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia PV, Italy
| | - Flavia Magri
- Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, 27100 Pavia PV, Italy; Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia PV, Italy
| | - Mario Rotondi
- Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, 27100 Pavia PV, Italy; Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia PV, Italy.
| |
Collapse
|
22
|
Slater O, Miller B, Kontoyianni M. Decoding Protein-protein Interactions: An Overview. Curr Top Med Chem 2021; 20:855-882. [PMID: 32101126 DOI: 10.2174/1568026620666200226105312] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 11/27/2019] [Accepted: 11/27/2019] [Indexed: 12/24/2022]
Abstract
Drug discovery has focused on the paradigm "one drug, one target" for a long time. However, small molecules can act at multiple macromolecular targets, which serves as the basis for drug repurposing. In an effort to expand the target space, and given advances in X-ray crystallography, protein-protein interactions have become an emerging focus area of drug discovery enterprises. Proteins interact with other biomolecules and it is this intricate network of interactions that determines the behavior of the system and its biological processes. In this review, we briefly discuss networks in disease, followed by computational methods for protein-protein complex prediction. Computational methodologies and techniques employed towards objectives such as protein-protein docking, protein-protein interactions, and interface predictions are described extensively. Docking aims at producing a complex between proteins, while interface predictions identify a subset of residues on one protein that could interact with a partner, and protein-protein interaction sites address whether two proteins interact. In addition, approaches to predict hot spots and binding sites are presented along with a representative example of our internal project on the chemokine CXC receptor 3 B-isoform and predictive modeling with IP10 and PF4.
Collapse
Affiliation(s)
- Olivia Slater
- Department of Pharmaceutical Sciences, Southern Illinois University, Edwardsville, IL 62026, United States
| | - Bethany Miller
- Department of Pharmaceutical Sciences, Southern Illinois University, Edwardsville, IL 62026, United States
| | - Maria Kontoyianni
- Department of Pharmaceutical Sciences, Southern Illinois University, Edwardsville, IL 62026, United States
| |
Collapse
|
23
|
Swogger J, Conner IP, Happ-Smith C, Kemmerer MC, Julian DR, Davis R, Wells A, Schuman JS, Yates CC. Novel combination therapy reduces subconjunctival fibrosis after glaucoma filtration surgery in the rabbit model. Clin Exp Ophthalmol 2021; 49:60-69. [PMID: 33426793 DOI: 10.1111/ceo.13884] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 10/08/2020] [Accepted: 11/06/2020] [Indexed: 01/28/2023]
Abstract
BACKGROUND Glaucoma filtration surgery (GFS) is limited by subconjunctival, episcleral and scleral fibrosis sealing the trabeculectomy and scarring the filtering bleb. Mitomycin-C (MMC) is commonly applied intraoperatively to the subconjunctival and/or intrascleral space to reduce scarring and promotes GFS success but is associated with postoperative scleral melting and bleb leaks. IP-10 peptide (IP-10p), an ELR-negative CXC chemokine mimetic and inhibitor of fibroblast function, may be an alternative or adjunct to current postoperative GFS treatments. This study sought to determine if IP-10p produces histological changes in tissue remodelling, vascularity and fibrosis that enhance bleb survival after GFS. METHODS Rabbits underwent tube-assisted filtration surgery on the right eye with either: (a) IP-10p injected into bleb at time of surgery and postoperative days 2, 4 and 7, (b) intraoperative MMC or (c) intraoperative MMC plus IP-10p injected into bleb at time of surgery and postoperative days 2, 4 and 7. Left contralateral eyes were treated with balanced salt solution (BSS). RESULTS IP-10p-treated blebs demonstrated reduced collagen deposition, cellularity and overall reduction of scar formation compared to BSS-control. Bleb vascularity was reduced compared to BSS-control and MMC treatment groups. Additionally, IP-10p/MMC treated eyes demonstrated an increased number of conjunctival goblet cells in bleb histology compared to the dramatic loss seen with MMC treatment alone. CONCLUSIONS This study demonstrates that IP-10p significantly reduces histological scarring compared to BSS or MMC alone, does not damage the conjunctiva to the extent of current standards, and may be an alternative or adjunct to MMC for those undergoing GFS.
Collapse
Affiliation(s)
- John Swogger
- Department of Ophthalmology, UPMC Eye Center, Eye and Ear Institute, Ophthalmology and Visual Science Research Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Ian P Conner
- Department of Ophthalmology, UPMC Eye Center, Eye and Ear Institute, Ophthalmology and Visual Science Research Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Carrie Happ-Smith
- Department of Ophthalmology, UPMC Eye Center, Eye and Ear Institute, Ophthalmology and Visual Science Research Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Megan C Kemmerer
- Department of Health Promotion and Development, University of Pittsburgh School of Nursing, Pittsburgh, Pennsylvania, USA
| | - Dana R Julian
- Department of Health Promotion and Development, University of Pittsburgh School of Nursing, Pittsburgh, Pennsylvania, USA
| | - Rachel Davis
- Department of Ophthalmology, UNM Eye Center, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Alan Wells
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Joel S Schuman
- Department of Ophthalmology, UPMC Eye Center, Eye and Ear Institute, Ophthalmology and Visual Science Research Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,New York University (NYU) Langone Eye Center, NYU Langone Medical Center, Department of Ophthalmology, NYU School of Medicine, New York, New York, USA
| | - Cecelia C Yates
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Health Promotion and Development, University of Pittsburgh School of Nursing, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
24
|
Aslam S, Khan I, Jameel F, Zaidi MB, Salim A. Umbilical cord-derived mesenchymal stem cells preconditioned with isorhamnetin: potential therapy for burn wounds. World J Stem Cells 2020; 12:1652-1666. [PMID: 33505606 PMCID: PMC7789118 DOI: 10.4252/wjsc.v12.i12.1652] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 10/01/2020] [Accepted: 10/26/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Impaired wound healing can be associated with different pathological states. Burn wounds are the most common and detrimental injuries and remain a major health issue worldwide. Mesenchymal stem cells (MSCs) possess the ability to regenerate tissues by secreting factors involved in promoting cell migration, proliferation and differentiation, while suppressing immune reactions. Preconditioning of MSCs with small molecules having cytoprotective properties can enhance the potential of these cells for their use in cell-based therapeutics. AIM To enhance the therapeutic potential of MSCs by preconditioning them with isorhamnetin for second degree burn wounds in rats. METHODS Human umbilical cord MSCs (hU-MSCs) were isolated and characterized by surface markers, CD105, vimentin and CD90. For preconditioning, hU-MSCs were treated with isorhamnetin after selection of the optimized concentration (5 µmol/L) by cytotoxicity analysis. The migration potential of these MSCs was analyzed by the in vitro scratch assay. The healing potential of normal, and preconditioned hU-MSCs was compared by transplanting these MSCs in a rat model of a second degree burn wound. Normal, and preconditioned MSCs (IH + MSCs) were transplanted after 72 h of burn injury and observed for 2 wk. Histological and gene expression analyses were performed on day 7 and 14 after cell transplantation to determine complete wound healing. RESULTS The scratch assay analysis showed a significant reduction in the scratch area in the case of IH + MSCs compared to the normal untreated MSCs at 24 h, while complete closure of the scratch area was observed at 48 h. Histological analysis showed reduced inflammation, completely remodeled epidermis and dermis without scar formation and regeneration of hair follicles in the group that received IH + MSCs. Gene expression analysis was time dependent and more pronounced in the case of IH + MSCs. Interleukin (IL)-1β, IL-6 and Bcl-2 associated X genes showed significant downregulation, while transforming growth factor β, vascular endothelial growth factor, Bcl-2 and matrix metallopeptidase 9 showed significant upregulation compared to the burn wound, showing increased angiogenesis and reduced inflammation and apoptosis. CONCLUSION Preconditioning of hU-MSCs with isorhamnetin decreases wound progression by reducing inflammation, and improving tissue architecture and wound healing. The study outcome is expected to lead to an improved cell-based therapeutic approach for burn wounds.
Collapse
Affiliation(s)
- Shazmeen Aslam
- Stem Cell Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 74700, Sindh, Pakistan
| | - Irfan Khan
- Stem Cell Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 74700, Sindh, Pakistan
| | - Fatima Jameel
- Stem Cell Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 74700, Sindh, Pakistan
| | - Midhat Batool Zaidi
- Stem Cell Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 74700, Sindh, Pakistan
| | - Asmat Salim
- Stem Cell Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 74700, Sindh, Pakistan.
| |
Collapse
|
25
|
Nienhold R, Ciani Y, Koelzer VH, Tzankov A, Haslbauer JD, Menter T, Schwab N, Henkel M, Frank A, Zsikla V, Willi N, Kempf W, Hoyler T, Barbareschi M, Moch H, Tolnay M, Cathomas G, Demichelis F, Junt T, Mertz KD. Two distinct immunopathological profiles in autopsy lungs of COVID-19. Nat Commun 2020; 11:5086. [PMID: 33033248 PMCID: PMC7546638 DOI: 10.1038/s41467-020-18854-2] [Citation(s) in RCA: 207] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/17/2020] [Indexed: 12/15/2022] Open
Abstract
Coronavirus Disease 19 (COVID-19) is a respiratory disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which has grown to a worldwide pandemic with substantial mortality. Immune mediated damage has been proposed as a pathogenic factor, but immune responses in lungs of COVID-19 patients remain poorly characterized. Here we show transcriptomic, histologic and cellular profiles of post mortem COVID-19 (n = 34 tissues from 16 patients) and normal lung tissues (n = 9 tissues from 6 patients). Two distinct immunopathological reaction patterns of lethal COVID-19 are identified. One pattern shows high local expression of interferon stimulated genes (ISGhigh) and cytokines, high viral loads and limited pulmonary damage, the other pattern shows severely damaged lungs, low ISGs (ISGlow), low viral loads and abundant infiltrating activated CD8+ T cells and macrophages. ISGhigh patients die significantly earlier after hospitalization than ISGlow patients. Our study may point to distinct stages of progression of COVID-19 lung disease and highlights the need for peripheral blood biomarkers that inform about patient lung status and guide treatment.
Collapse
Affiliation(s)
- Ronny Nienhold
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Yari Ciani
- Laboratory of Computational and Functional Oncology, Department for Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento, Italy
| | - Viktor H Koelzer
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
- Department of Oncology and Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Alexandar Tzankov
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Jasmin D Haslbauer
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Thomas Menter
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Nathalie Schwab
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Maurice Henkel
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Angela Frank
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Veronika Zsikla
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Niels Willi
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Werner Kempf
- Kempf und Pfaltz Histologische Diagnostik, Zurich, Switzerland
| | - Thomas Hoyler
- Novartis Institutes for BioMedical Research (NIBR), Basel, Switzerland
| | - Mattia Barbareschi
- Anatomia ed Istologia Patologica, Ospedale S. Chiara di Trento, Trento, Italy
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Markus Tolnay
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Gieri Cathomas
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Francesca Demichelis
- Laboratory of Computational and Functional Oncology, Department for Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento, Italy
- Caryl and Israel Englander Institute for Precision Medicine, Institute for Computational Biomedicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY, USA
| | - Tobias Junt
- Novartis Institutes for BioMedical Research (NIBR), Basel, Switzerland
| | - Kirsten D Mertz
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland.
| |
Collapse
|
26
|
Bi Y, Shirure VS, Liu R, Cunningham C, Ding L, Meacham JM, Goedegebuure SP, George SC, Fields RC. Tumor-on-a-chip platform to interrogate the role of macrophages in tumor progression. Integr Biol (Camb) 2020; 12:221-232. [PMID: 32930334 DOI: 10.1093/intbio/zyaa017] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/20/2020] [Accepted: 08/15/2020] [Indexed: 02/06/2023]
Abstract
Tumor-infiltrating leukocytes, in particular macrophages, play an important role in tumor behavior and clinical outcome. The spectrum of macrophage subtypes ranges from antitumor 'M1'-type to protumor 'M2'-type macrophages. Tumor-associated macrophages (TAMs) typically display phenotypic features of both M1 and M2, and the population distribution is thought to be dynamic and evolves as the tumor progresses. However, our understanding of how TAMs impact the tumor microenvironment remains limited by the lack of appropriate 3D in vitro models that can capture cell-cell dynamics at high spatial and temporal resolution. Using our recently developed microphysiological 'tumor-on-a-chip' (TOC) device, we present here our findings on the impact of defined macrophage subsets on tumor behavior. The TOC device design contains three adjacent and connected chambers in which both the upper and lower chambers are loaded with tumor cells, whereas the central chamber contains a dynamic, perfused, living microvascular network. Introduction of human pancreatic or colorectal cancer cells together with M1-polarized macrophages significantly inhibited tumor growth and tumor-induced angiogenesis. Protein analysis and antibody-based neutralization studies confirmed that these effects were mediated through production of C-X-C motif chemokines (CXCL9), CXCL10 and CXCL11. By contrast, M2-macrophages mediated increased tumor cell migration into the vascularized chamber and did not inhibit tumor growth or angiogenesis. In fact, single-cell RNA sequencing showed that M2 macrophages further segregated endothelial cells into two distinct subsets, corresponding to static cells in vessels versus active cells involved in angiogenesis. The impact of M2 macrophages was mediated mostly by production of matrix metalloproteinase 7 and angiopoietin 2. In summary, our data demonstrate the utility of the TOC device to mechanistically probe biological questions in a 3D in vitro microenvironment.
Collapse
Affiliation(s)
- Ye Bi
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Venktesh S Shirure
- Department of Biomedical Engineering, University of California Davis, Davis, CA 95616, USA
| | - Ruiyang Liu
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA.,McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Cassandra Cunningham
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Li Ding
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA.,McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - J Mark Meacham
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - S Peter Goedegebuure
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA.,Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Steven C George
- Department of Biomedical Engineering, University of California Davis, Davis, CA 95616, USA
| | - Ryan C Fields
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA.,Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
27
|
Ishiuchi-Sato Y, Hiraiwa E, Shinozaki A, Nedachi T. The effects of glucose and fatty acids on CXCL10 expression in skeletal muscle cells. Biosci Biotechnol Biochem 2020; 84:2448-2457. [PMID: 32877316 DOI: 10.1080/09168451.2020.1814127] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Skeletal muscles produce secretory factors termed as myokines, which alter physiological functions of target tissues. We recently identified C-X-C chemokine ligand 10 (CXCL10) as a novel myokine, which is downregulated in response to exercise. In the present study, we investigated whether the nutritional changes affect CXCL10 expression in mouse skeletal muscle. Expression of CXCL10 was evaluated in mice fed a normal diet or a high fat diet for 10 weeks. In animals fed on HFD, Cxcl10 expression was significantly induced in fast-twitched muscles, and was accompanied by increased blood glucose and free fatty acid levels. In vitro experiments using C2C12 myotubes suggested that the increased levels of glucose and palmitic acids directly enhanced CXCL10 expression. Interestingly, the effect of palmitic acids was attenuated by palmitoleic acids. Considering its potent angiostatic activity, induction of CXCL10 by nutritional changes may contribute to the impairment of microvascular networks in skeletal muscles.
Collapse
Affiliation(s)
| | - Erika Hiraiwa
- Faculty of Life Sciences, Toyo University , Gunma, Japan
| | | | - Taku Nedachi
- Graduate School of Life Sciences, Toyo University , Gunma, Japan.,Faculty of Life Sciences, Toyo University , Gunma, Japan
| |
Collapse
|
28
|
Groblewska M, Litman-Zawadzka A, Mroczko B. The Role of Selected Chemokines and Their Receptors in the Development of Gliomas. Int J Mol Sci 2020; 21:ijms21103704. [PMID: 32456359 PMCID: PMC7279280 DOI: 10.3390/ijms21103704] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/21/2020] [Accepted: 05/22/2020] [Indexed: 02/07/2023] Open
Abstract
Among heterogeneous primary tumors of the central nervous system (CNS), gliomas are the most frequent type, with glioblastoma multiforme (GBM) characterized with the worst prognosis. In their development, certain chemokine/receptor axes play important roles and promote proliferation, survival, metastasis, and neoangiogenesis. However, little is known about the significance of atypical receptors for chemokines (ACKRs) in these tumors. The objective of the study was to present the role of chemokines and their conventional and atypical receptors in CNS tumors. Therefore, we performed a thorough search for literature concerning our investigation via the PubMed database. We describe biological functions of chemokines/chemokine receptors from various groups and their significance in carcinogenesis, cancer-related inflammation, neo-angiogenesis, tumor growth, and metastasis. Furthermore, we discuss the role of chemokines in glioma development, with particular regard to their function in the transition from low-grade to high-grade tumors and angiogenic switch. We also depict various chemokine/receptor axes, such as CXCL8-CXCR1/2, CXCL12-CXCR4, CXCL16-CXCR6, CX3CL1-CX3CR1, CCL2-CCR2, and CCL5-CCR5 of special importance in gliomas, as well as atypical chemokine receptors ACKR1-4, CCRL2, and PITPMN3. Additionally, the diagnostic significance and usefulness of the measurement of some chemokines and their receptors in the blood and cerebrospinal fluid (CSF) of glioma patients is also presented.
Collapse
Affiliation(s)
- Magdalena Groblewska
- Department of Biochemical Diagnostics, University Hospital in Białystok, 15-269 Białystok, Poland;
| | - Ala Litman-Zawadzka
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, 15-269 Białystok, Poland;
| | - Barbara Mroczko
- Department of Biochemical Diagnostics, University Hospital in Białystok, 15-269 Białystok, Poland;
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, 15-269 Białystok, Poland;
- Correspondence: ; Tel.: +48-85-831-8785
| |
Collapse
|
29
|
Özkan A, Stolley D, Cressman ENK, McMillin M, DeMorrow S, Yankeelov TE, Rylander MN. The Influence of Chronic Liver Diseases on Hepatic Vasculature: A Liver-on-a-chip Review. MICROMACHINES 2020; 11:E487. [PMID: 32397454 PMCID: PMC7281532 DOI: 10.3390/mi11050487] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/02/2020] [Accepted: 05/04/2020] [Indexed: 12/13/2022]
Abstract
In chronic liver diseases and hepatocellular carcinoma, the cells and extracellular matrix of the liver undergo significant alteration in response to chronic injury. Recent literature has highlighted the critical, but less studied, role of the liver vasculature in the progression of chronic liver diseases. Recent advancements in liver-on-a-chip systems has allowed in depth investigation of the role that the hepatic vasculature plays both in response to, and progression of, chronic liver disease. In this review, we first introduce the structure, gradients, mechanical properties, and cellular composition of the liver and describe how these factors influence the vasculature. We summarize state-of-the-art vascularized liver-on-a-chip platforms for investigating biological models of chronic liver disease and their influence on the liver sinusoidal endothelial cells of the hepatic vasculature. We conclude with a discussion of how future developments in the field may affect the study of chronic liver diseases, and drug development and testing.
Collapse
Affiliation(s)
- Alican Özkan
- Department of Mechanical Engineering, The University of Texas, Austin, TX 78712, USA
| | - Danielle Stolley
- Department of Biomedical Engineering, The University of Texas, Austin, TX 78712, USA
| | - Erik N K Cressman
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Matthew McMillin
- Department of Internal Medicine, Dell Medical School, The University of Texas at Austin, Austin, TX 78713, USA
- Central Texas Veterans Health Care System, Temple, TX 76504, USA
| | - Sharon DeMorrow
- Department of Internal Medicine, Dell Medical School, The University of Texas at Austin, Austin, TX 78713, USA
- Central Texas Veterans Health Care System, Temple, TX 76504, USA
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Thomas E Yankeelov
- Department of Biomedical Engineering, The University of Texas, Austin, TX 78712, USA
- Oden Institute for Computational Engineering and Sciences, The University of Texas, Austin, TX 78712, USA
- Departments of Diagnostic Medicine, The University of Texas, Austin, TX 78712, USA
- Department of Oncology, The University of Texas, Austin, TX 78712, USA
- Livestrong Cancer Institutes, Dell Medical School, The University of Texas, Austin, TX 78712, USA
| | - Marissa Nichole Rylander
- Department of Mechanical Engineering, The University of Texas, Austin, TX 78712, USA
- Department of Biomedical Engineering, The University of Texas, Austin, TX 78712, USA
- Oden Institute for Computational Engineering and Sciences, The University of Texas, Austin, TX 78712, USA
| |
Collapse
|
30
|
Okonkwo UA, Chen L, Ma D, Haywood VA, Barakat M, Urao N, DiPietro LA. Compromised angiogenesis and vascular Integrity in impaired diabetic wound healing. PLoS One 2020; 15:e0231962. [PMID: 32324828 PMCID: PMC7179900 DOI: 10.1371/journal.pone.0231962] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 04/03/2020] [Indexed: 12/19/2022] Open
Abstract
Vascular deficits are a fundamental contributing factor of diabetes-associated diseases. Although previous studies have demonstrated that the pro-angiogenic phase of wound healing is blunted in diabetes, a comprehensive understanding of the mechanisms that regulate skin revascularization and capillary stabilization in diabetic wounds is lacking. Using a mouse model of diabetic wound healing, we performed microCT analysis of the 3-dimensional architecture of the capillary bed. As compared to wild type, vessel surface area, branch junction number, total vessel length, and total branch number were significantly decreased in wounds of diabetic mice as compared to WT mice. Diabetic mouse wounds also had significantly increased capillary permeability and decreased pericyte coverage of capillaries. Diabetic wounds exhibited significant perturbations in the expression of factors that affect vascular regrowth, maturation and stability. Specifically, the expression of VEGF-A, Sprouty2, PEDF, LRP6, Thrombospondin 1, CXCL10, CXCR3, PDGFR-β, HB-EGF, EGFR, TGF-β1, Semaphorin3a, Neuropilin 1, angiopoietin 2, NG2, and RGS5 were down-regulated in diabetic wounds. Together, these studies provide novel information about the complexity of the perturbation of angiogenesis in diabetic wounds. Targeting factors responsible for wound resolution and vascular pruning, as well those that affect pericyte recruitment, maturation, and stability may have the potential to improve diabetic skin wound healing.
Collapse
Affiliation(s)
- Uzoagu A. Okonkwo
- Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Lin Chen
- Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Da Ma
- Guangdong Provincial Key Laboratory of Stomatology, Stomatological Hospital, Guanghua School of Stomatology, SunYat-sen University, Guangzhou, Guangdong, China
| | - Veronica A. Haywood
- Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL, United States of America
| | - May Barakat
- Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Norifumi Urao
- Department of Pharmacology, Upstate Medical University, Syracuse, NY, United States of America
| | - Luisa A. DiPietro
- Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL, United States of America
- * E-mail:
| |
Collapse
|
31
|
Nazari A, Ahmadi Z, Hassanshahi G, Abbasifard M, Taghipour Z, Falahati-Pour SK, Khorramdelazad H. Effective Treatments for Bladder Cancer Affecting CXCL9/CXCL10/CXCL11/CXCR3 Axis: A Review. Oman Med J 2020; 35:e103. [PMID: 32181005 PMCID: PMC7064791 DOI: 10.5001/omj.2020.21] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 01/14/2019] [Indexed: 02/06/2023] Open
Abstract
Bladder cancer (BC) originates mainly from the epithelial compartment of the bladder, which is defined as transitional cell carcinoma or urothelial cell carcinoma. About 70% of patients with BC will survive five years from diagnosis. Previous studies revealed that the immune system and its mediators, particularly chemokines, play a crucial role in modulating responses against BC. Chemokines, which serve as chemoattractants for leukocytes, are small proteins that can initiate inflammatory and anti-inflammatory immune responses and also are associated with many aspects of both regulation and progression of mentioned responses. Additionally, these immune mediators can interfere with the other tumor-related processes, including tumor proliferation, neovascularization, and metastases. Among these chemokines, CXC chemokines, including CXCL9, CXCL10, and CXCL11, are recognized as the main ligands of C-X-C motif chemokine receptor 3 (CXCR3) and contribute to related immune responses after therapeutic strategies for BC. Evidence suggests that the production of these chemokines can have two important implications. First, these mediators can trigger the accumulation of CD8+ T cells that can contribute to the elimination of the tumor. Secondly, the production of these chemokines by tumor tissue may trigger the migration and activation of immune cells including myeloid-derived suppressor cells and regulatory T cells, which act in favor of the tumor and its progress. Therefore, in this review, we describe the latest therapeutic approaches based on targeting this axis's components and subsequent immune phenomenon.
Collapse
Affiliation(s)
- Alireza Nazari
- Non Communicable Diseases Research Center, Rafsanjan University of Medical Science, Rafsanjan, Iran.,Department of Surgery, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Zahra Ahmadi
- Pistachio Safety Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Gholamhossein Hassanshahi
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.,Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mitra Abbasifard
- Department of Internal Medicine, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Zahra Taghipour
- Department of Anatomy, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | | | - Hossein Khorramdelazad
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.,Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.,Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
32
|
Dókus LE, Yousef M, Bánóczi Z. Modulators of calpain activity: inhibitors and activators as potential drugs. Expert Opin Drug Discov 2020; 15:471-486. [DOI: 10.1080/17460441.2020.1722638] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Levente Endre Dókus
- Department of Organic Chemistry, Institute of Chemistry, Eötvös Loránd University, Budapest, Hungary
| | - Mo’ath Yousef
- Department of Organic Chemistry, Institute of Chemistry, Eötvös Loránd University, Budapest, Hungary
| | - Zoltán Bánóczi
- Department of Organic Chemistry, Institute of Chemistry, Eötvös Loránd University, Budapest, Hungary
| |
Collapse
|
33
|
Michl TD, Tran DTT, Kuckling HF, Zhalgasbaikyzy A, Ivanovská B, González García LE, Visalakshan RM, Vasilev K. It takes two for chronic wounds to heal: dispersing bacterial biofilm and modulating inflammation with dual action plasma coatings. RSC Adv 2020; 10:7368-7376. [PMID: 35492196 PMCID: PMC9049834 DOI: 10.1039/c9ra09875e] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/06/2020] [Indexed: 11/21/2022] Open
Abstract
Chronic wounds are affecting increasingly larger portions of the general population and their treatment has essentially remained unchanged for the past century. This lack of progress is due to the complex problem that chronic wounds are simultaneously infected and inflamed. Both aspects need to be addressed together to achieve a better healing outcome. Hence, we hereby demonstrate that the stable nitroxide radical (2,2,6,6-tetramethylpiperidin-1-yl)oxyl (TEMPO) can be plasma polymerized into smooth coatings (TEMPOpp), as seen via atomic force microscopy, X-ray photoelectron spectroscopy and ellipsometry. Upon contact with water, these coatings leach nitroxides into aqueous supernatant, as measured via EPR. We then exploited the known cell-signalling qualities of TEMPO to change the cellular behaviour of bacteria and human cells that come into contact with the surfaces. Specifically, the TEMPOpp coatings not only suppressed biofilm formation of the opportunistic bacterium Staphylococcus epidermidis but also dispersed already formed biofilm in a dose-dependent manner; a crucial aspect in treating chronic wounds that contain bacterial biofilm. Thus the coatings' microbiological efficacy correlated with their thickness and the thickest coating was the most efficient. Furthermore, this dose-dependent effect was mirrored in significant cytokine reduction of activated THP-1 macrophages for the four cytokines TNF-α, IL-1β, IL-6 and IP-10. At the same time, the THP-1 cells retained their ability to adhere and colonize the surfaces, as verified via SEM imaging. Thus, summarily, we have exploited the unique qualities of plasma polymerized TEMPO coatings in targeting both infection and inflammation simultaneously; demonstrating a novel alternative to how chronic wounds could be treated in the future. We plasma polymerized the stable nitroxide radical TEMPO into thin coatings and exploited the coatings' unique qualities in targeting both infection and inflammation simultaneously; demonstrating a novel alternative as to how chronic wounds could be treated in the future.![]()
Collapse
Affiliation(s)
| | | | | | | | - Barbora Ivanovská
- School of Engineering
- University of South Australia
- Mawson Lakes
- Australia
| | | | | | - Krasimir Vasilev
- School of Engineering
- University of South Australia
- Mawson Lakes
- Australia
| |
Collapse
|
34
|
Patel S, Pragati, Dwivedi SD, Yadav K, Kanwar JR, Singh MR, Singh D. Pathogenesis and Molecular Targets in Treatment of Diabetic Wounds. OBESITY AND DIABETES 2020:747-758. [DOI: 10.1007/978-3-030-53370-0_55] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
35
|
Xu W, Puligandla M, Manola J, Bullock AJ, Tamasauskas D, McDermott DF, Atkins MB, Haas NB, Flaherty K, Uzzo RG, Dutcher JP, DiPaola RS, Bhatt RS. Angiogenic Factor and Cytokine Analysis among Patients Treated with Adjuvant VEGFR TKIs in Resected Renal Cell Carcinoma. Clin Cancer Res 2019; 25:6098-6106. [PMID: 31471309 DOI: 10.1158/1078-0432.ccr-19-0818] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 06/06/2019] [Accepted: 07/09/2019] [Indexed: 12/30/2022]
Abstract
PURPOSE The use of VEGFR TKIs for the adjuvant treatment of renal cell carcinoma (RCC) remains controversial. We investigated the effects of adjuvant VEGFR TKIs on circulating cytokines in the ECOG-ACRIN 2805 (ASSURE) trial. EXPERIMENTAL DESIGN Patients with resected high-risk RCC were randomized to sunitinib, sorafenib, or placebo. Plasma from 413 patients was analyzed from post-nephrectomy baseline, 4 weeks, and 6 weeks after treatment initiation. Mixed effects and Cox proportional hazards models were used to test for changes in circulating cytokines and associations between disease-free survival (DFS) and cytokine levels. RESULTS VEGF and PlGF increased after 4 weeks on sunitinib or sorafenib (P < 0.0001 for both) and returned to baseline at 6 weeks on sunitinib (corresponding to the break in the sunitinib schedule) but not sorafenib (which was administered continuously). sFLT-1 decreased after 4 weeks on sunitinib and 6 weeks on sorafenib (P < 0.0001). sVEGFR-2 decreased after both 4 and 6 weeks of treatment on sunitinib or sorafenib (P < 0.0001). Patients receiving placebo had no significant changes in cytokine levels. CXCL10 was elevated at 4 and 6 weeks on sunitinib and sorafenib but not on placebo. Higher baseline CXCL10 was associated with worse DFS (HR 1.41 per log increase in CXCL10, Bonferroni-adjusted P = 0.003). This remained significant after adjustment for T-stage, Fuhrman grade, and ECOG performance status. CONCLUSIONS Among patients treated with adjuvant VEGFR TKIs for RCC, drug-host interactions mediate changes in circulating cytokines. Elevated baseline CXCL10 was associated with worse DFS. Studies to understand functional consequences of these changes are under way.
Collapse
Affiliation(s)
- Wenxin Xu
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Maneka Puligandla
- Dana-Farber Cancer Institute, ECOG-ACRIN Biostatistics Center, Boston, Massachusetts
| | - Judith Manola
- Dana-Farber Cancer Institute, ECOG-ACRIN Biostatistics Center, Boston, Massachusetts
| | | | | | | | - Michael B Atkins
- MedStar Georgetown University Hospital, Washington, District of Columbia
| | - Naomi B Haas
- Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | | | | | - Rupal S Bhatt
- Beth Israel Deaconess Medical Center, Boston, Massachusetts.
| |
Collapse
|
36
|
Butler CT, Kennedy SA, Buckley A, Doyle R, Conroy E, Gallagher WM, O'Sullivan J, Kennedy BN. 1,4-dihydroxy quininib attenuates growth of colorectal cancer cells and xenografts and regulates the TIE-2 signaling pathway in patient tumours. Oncotarget 2019; 10:3725-3744. [PMID: 31217905 PMCID: PMC6557215 DOI: 10.18632/oncotarget.26966] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 04/21/2019] [Indexed: 12/20/2022] Open
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer associated deaths in developed countries. Cancer progression and metastatic spread is reliant on new blood vasculature, or angiogenesis. Tumour-related angiogenesis is regulated by pro- and anti-angiogenic factors secreted from malignant tissue in a stepwise process. Previously we structurally modified the small anti-angiogenic molecule quininib and discovered a more potent anti-angiogenic compound 1, 4 dihydroxy quininib (Q8), an antagonist of cysteinyl leukotriene receptor-1 with VEGF-independent bioactivity. Here, Q8, quininib (Q1) and five structural analogues were assayed for anti-tumorigenic effects in pre-clinical cancer models. Q8 reduced clone formation of the human colorectal cancer cell line HT29-Luc2. Gene silencing of CysLT1 in HT29-Luc2 cells significantly reduced expression of calpain-2. In human ex vivo colorectal cancer tumour explants, Q8 significantly decreased the secretion of both TIE-2 and VCAM-1 expression. In vivo Q8 was well tolerated up to 50 mg/kg by Balb/C mice and significantly more effective at reducing tumour volume in colorectal tumour xenografts compared to the parent drug quininib. In tumour xenografts, Q8 significantly reduced expression of the angiogenic marker calpain-2. In summary, we propose Q8 may act on the TIE-2-Angiopoietin signalling pathway to significantly inhibit the process of tumour angiogenesis in colorectal cancer.
Collapse
Affiliation(s)
- Clare T Butler
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, Dublin, Ireland
| | - Susan A Kennedy
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James's Hospital, Dublin, Ireland
| | - Amy Buckley
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James's Hospital, Dublin, Ireland
| | - Ronan Doyle
- Department of Histopathology, Trinity College Dublin Central Pathology Laboratory, St James's Hospital, Dublin, Ireland
| | - Emer Conroy
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, Dublin, Ireland
| | - William M Gallagher
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, Dublin, Ireland
| | - Jacintha O'Sullivan
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James's Hospital, Dublin, Ireland.,These authors contributed equally to this work
| | - Breandán N Kennedy
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, Dublin, Ireland.,These authors contributed equally to this work
| |
Collapse
|
37
|
Ridiandries A, Tan JTM, Bursill CA. The Role of Chemokines in Wound Healing. Int J Mol Sci 2018; 19:ijms19103217. [PMID: 30340330 PMCID: PMC6214117 DOI: 10.3390/ijms19103217] [Citation(s) in RCA: 303] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Revised: 10/14/2018] [Accepted: 10/15/2018] [Indexed: 12/15/2022] Open
Abstract
Wound healing is a multistep process with four overlapping but distinct stages: hemostasis, inflammation, proliferation, and remodeling. An alteration at any stage may lead to the development of chronic non-healing wounds or excessive scar formation. Impaired wound healing presents a significant health and economic burden to millions of individuals worldwide, with diabetes mellitus and aging being major risk factors. Ongoing understanding of the mechanisms that underly wound healing is required for the development of new and improved therapies that increase repair. Chemokines are key regulators of the wound healing process. They are involved in the promotion and inhibition of angiogenesis and the recruitment of inflammatory cells, which release growth factors and cytokines to facilitate the wound healing process. Preclinical research studies in mice show that the administration of CCL2, CCL21, CXCL12, and a CXCR4 antagonist as well as broad-spectrum inhibition of the CC-chemokine class improve the wound healing process. The focus of this review is to highlight the contributions of chemokines during each stage of wound healing and to discuss the related molecular pathologies in complex and chronic non-healing wounds. We explore the therapeutic potential of targeting chemokines as a novel approach to overcome the debilitating effects of impaired wound healing.
Collapse
Affiliation(s)
- Anisyah Ridiandries
- Department of Cardiology, Kolling Institute, Northern Sydney Local Health District, St Leonards, NSW 2065, Australia.
- Sydney Medical School Northern, University of Sydney, Sydney, NSW 2006, Australia.
| | - Joanne T M Tan
- Heart Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, SA 5000, Australia.
- Adelaide Medical School, Faculty of Health & Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia.
| | - Christina A Bursill
- Heart Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, SA 5000, Australia.
- Adelaide Medical School, Faculty of Health & Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia.
| |
Collapse
|
38
|
Bradshaw A, Sylakowski K, Wells A. The Pro-reparative Engine: Stem Cells Aid Healing by Dampening Inflammation. CURRENT PATHOBIOLOGY REPORTS 2018; 6:109-115. [PMID: 30271682 DOI: 10.1007/s40139-018-0167-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Purpose of Review Stem cells have been proposed as sources for tissue replacement when healing does not occur. These cells could contribute directly to skin structures via differentiation, or via producing trophic factors that would 'educate' the micro-environment to encourage tissue repair. Studies in animals have supported both mechanisms, but translation to humans has been challenged by poor cell survival after transplantation. However, the improvement noted with even transient existence suggests another new possibility, that of suppressing the inflammatory response that limits regenerative healing. Herein, we will propose that this immunomodulatory aspect holds promise for promoting skin healing. Recent Findings We have found that stem cell transplantation into wounds can dampen both acute and chronic inflammation, leading to more regenerative-like healing and diminished scarring. Summary Wound healing could be improved by dampening inflammation both initially to allow for tissue replacement to proceed and late to reduce scarring.
Collapse
Affiliation(s)
- Andrew Bradshaw
- Departments of Pathology and Bioengineering, and the McGowan, Institute for Regenerative Medicine, University of Pittsburgh, and VA Pittsburgh Health System, Pittsburgh PA 15213 USA
| | - Kyle Sylakowski
- Departments of Pathology and Bioengineering, and the McGowan, Institute for Regenerative Medicine, University of Pittsburgh, and VA Pittsburgh Health System, Pittsburgh PA 15213 USA
| | - Alan Wells
- Departments of Pathology and Bioengineering, and the McGowan, Institute for Regenerative Medicine, University of Pittsburgh, and VA Pittsburgh Health System, Pittsburgh PA 15213 USA
| |
Collapse
|
39
|
Jia H, Thelwell C, Dilger P, Bird C, Daniels S, Wadhwa M. Endothelial cell functions impaired by interferon in vitro: Insights into the molecular mechanism of thrombotic microangiopathy associated with interferon therapy. Thromb Res 2018; 163:105-116. [PMID: 29407621 DOI: 10.1016/j.thromres.2018.01.039] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 12/22/2017] [Accepted: 01/22/2018] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Interferon (IFN)-α and IFN-β approved for treatment of chronic hepatitis C viral infection and multiple sclerosis respectively have been linked to thrombotic microangiopathy (TMA) affecting renal function. Since the molecular mechanisms underlying this severe complication remain largely unclear, we aimed to investigate whether IFN affects directly in vitro endothelial cell functions associated with angiogenesis and blood haemostasis, as well as endothelial cell-derived vasodilators of nitric oxide (NO) and prostacyclin. METHODS Proliferation and survival of human umbilical vein endothelial cells (HUVECs) were measured by BrdU incorporation and alamarBlue assays. Angiogenesis was evaluated in co-cultures of HUVECs and human dermal fibroblasts. Fibrinolysis molecules were measured with ELISA. NO and prostacyclin were measured using a fluorescent NO-specific probe and a competitive enzyme immunoassay, respectively. RESULTS HUVEC proliferation was dose-dependently inhibited by IFN-β1a and IFN-β1b, but not by IFN-α2a and IFN-α2b. Consistently, IFN-β1a and IFN-β1b also reduced survival of HUVECs, but this again was not observed with IFN-α. However, both IFN subtypes inhibited VEGF-induced development of capillary-like structures, but the effect of IFN-α was less potent than IFN-β. In addition, both IFN subtypes upregulated interferon inducible protein 10 production from treated co-cultures while suppressing angiogenesis. Furthermore, intracellular NO generation was reduced by IFN-α2a and IFN-β1a, whereas prostacyclin release from HUVECs was not affected by IFN. Importantly, both IFN-β1a- and IFN-β1b-treated HUVECs showed a marked reduction in urokinase-type plasminogen activator release and a much greater secretion of plasminogen activator inhibitor-1 than tissue-type plasminogen activator compared with untreated cells, suggesting decreased fibrinolytic activity. IFN-α, however was less effective in modulating the fibrinolysis system. CONCLUSIONS We demonstrate the detrimental effects of IFN on endothelial cell functions mediated with angiogenesis and fibrinolysis, which could potentially cause the loss of physiological endothelium thromboresistance and facilitate the development of vascular complications in a clinical setting. Mechanistically, our findings have implications for understanding how IFN therapy can foster the development of TMA.
Collapse
Affiliation(s)
- Haiyan Jia
- Section of Cytokines and Growth Factors, Division of Biotherapeutics, National Institute for Biological Standards and Control, United Kingdom.
| | - Craig Thelwell
- Section of Haemostasis, Division of Biotherapeutics, National Institute for Biological Standards and Control, United Kingdom
| | - Paula Dilger
- Section of Cytokines and Growth Factors, Division of Biotherapeutics, National Institute for Biological Standards and Control, United Kingdom
| | - Chris Bird
- Section of Cytokines and Growth Factors, Division of Biotherapeutics, National Institute for Biological Standards and Control, United Kingdom
| | - Sarah Daniels
- Section of Haemostasis, Division of Biotherapeutics, National Institute for Biological Standards and Control, United Kingdom
| | - Meenu Wadhwa
- Section of Cytokines and Growth Factors, Division of Biotherapeutics, National Institute for Biological Standards and Control, United Kingdom
| |
Collapse
|
40
|
Beckwitt CH, Clark AM, Wheeler S, Taylor DL, Stolz DB, Griffith L, Wells A. Liver 'organ on a chip'. Exp Cell Res 2018; 363:15-25. [PMID: 29291400 PMCID: PMC5944300 DOI: 10.1016/j.yexcr.2017.12.023] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 12/21/2017] [Accepted: 12/27/2017] [Indexed: 12/14/2022]
Abstract
The liver plays critical roles in both homeostasis and pathology. It is the major site of drug metabolism in the body and, as such, a common target for drug-induced toxicity and is susceptible to a wide range of diseases. In contrast to other solid organs, the liver possesses the unique ability to regenerate. The physiological importance and plasticity of this organ make it a crucial system of study to better understand human physiology, disease, and response to exogenous compounds. These aspects have impelled many to develop liver tissue systems for study in isolation outside the body. Herein, we discuss these biologically engineered organoids and microphysiological systems. These aspects have impelled many to develop liver tissue systems for study in isolation outside the body. Herein, we discuss these biologically engineered organoids and microphysiological systems.
Collapse
Affiliation(s)
- Colin H Beckwitt
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA; The McGowan Institute of Regenerative Medicine University of Pittsburgh, Pittsburgh, PA 15213, USA; Research and Development Service, VA Pittsburgh Health System, Pittsburgh, PA 15240, USA
| | - Amanda M Clark
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Sarah Wheeler
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - D Lansing Taylor
- Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA; The McGowan Institute of Regenerative Medicine University of Pittsburgh, Pittsburgh, PA 15213, USA; Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Donna B Stolz
- Cell Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA; The McGowan Institute of Regenerative Medicine University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Linda Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Alan Wells
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA; Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA; The McGowan Institute of Regenerative Medicine University of Pittsburgh, Pittsburgh, PA 15213, USA; Research and Development Service, VA Pittsburgh Health System, Pittsburgh, PA 15240, USA.
| |
Collapse
|
41
|
Bünemann E, Hoff NP, Buhren BA, Wiesner U, Meller S, Bölke E, Müller-Homey A, Kubitza R, Ruzicka T, Zlotnik A, Homey B, Gerber PA. Chemokine ligand-receptor interactions critically regulate cutaneous wound healing. Eur J Med Res 2018; 23:4. [PMID: 29338773 PMCID: PMC5771017 DOI: 10.1186/s40001-017-0299-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 12/22/2017] [Indexed: 01/25/2023] Open
Abstract
Background Wound healing represents a dynamic process involving directional migration of different cell types. Chemokines, a family of chemoattractive proteins, have been suggested to be key players in cell-to-cell communication and essential for directed migration of structural cells. Today, the role of the chemokine network in cutaneous wound healing is not fully understood. Unraveling the chemokine-driven communication pathways in this complex process could possibly lead to new therapeutic strategies in wound healing disorders. Methods We performed a systematic, comprehensive time-course analysis of the expression and function of a broad variety of cytokines, growth factors, adhesion molecules, matrixmetalloproteinases and chemokines in a murine cutaneous wound healing model. Results Strikingly, chemokines were found to be among the most highly regulated genes and their expression was found to coincide with the expression of their matching receptors. Accordingly, we could show that resting and activated human primary keratinocytes (CCR3, CCR4, CCR6, CXCR1, CXCR3), dermal fibroblasts (CCR3, CCR4, CCR10) and dermal microvascular endothelial cells (CCR3, CCR4, CCR6, CCR8, CCR9, CCR10, CXCR1, CXCR2, CXCR3) express a distinct and functionally active repertoire of chemokine receptors. Furthermore, chemokine ligand–receptor interactions markedly improved the wound repair of structural skin cells in vitro. Conclusion Taken together, we here present the most comprehensive analysis of mediators critically involved in acute cutaneous wound healing. Our findings suggest therapeutic approaches for the management of wound closure by targeting the chemokine network. Electronic supplementary material The online version of this article (10.1186/s40001-017-0299-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Erich Bünemann
- Department of Dermatology, University Clinic Duesseldorf, 40225, Duesseldorf, Germany
| | - Norman-Philipp Hoff
- Department of Dermatology, University Clinic Duesseldorf, 40225, Duesseldorf, Germany
| | | | - Ulrike Wiesner
- Department of Dermatology, University Clinic Duesseldorf, 40225, Duesseldorf, Germany
| | - Stephan Meller
- Department of Dermatology, University Clinic Duesseldorf, 40225, Duesseldorf, Germany
| | - Edwin Bölke
- Department of Radiation Oncology, University Clinic Duesseldorf, 40225, Duesseldorf, Germany
| | - Anja Müller-Homey
- Department of Radiation Oncology, University Clinic Duesseldorf, 40225, Duesseldorf, Germany
| | - Robert Kubitza
- Department of Dermatology, University Clinic Duesseldorf, 40225, Duesseldorf, Germany
| | - Thomas Ruzicka
- Department of Dermatology, Ludwig-Maximilian-University of Munich, 80539, Munich, Germany
| | - Albert Zlotnik
- Department of Biology, Senomyx, Inc, 4767 Nexus Center Drive, San Diego, CA, 92121, USA
| | - Bernhard Homey
- Department of Dermatology, University Clinic Duesseldorf, 40225, Duesseldorf, Germany.
| | - Peter Arne Gerber
- Department of Dermatology, University Clinic Duesseldorf, 40225, Duesseldorf, Germany
| |
Collapse
|
42
|
Rubina KA, Semina EV, Tkachuk VA. Guidance molecules and chemokines in angiogenesis and vascular remodeling. J EVOL BIOCHEM PHYS+ 2017. [DOI: 10.1134/s0022093017050015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
43
|
Kammertoens T, Friese C, Arina A, Idel C, Briesemeister D, Rothe M, Ivanov A, Szymborska A, Patone G, Kunz S, Sommermeyer D, Engels B, Leisegang M, Textor A, Fehling HJ, Fruttiger M, Lohoff M, Herrmann A, Yu H, Weichselbaum R, Uckert W, Hübner N, Gerhardt H, Beule D, Schreiber H, Blankenstein T. Tumour ischaemia by interferon-γ resembles physiological blood vessel regression. Nature 2017; 545:98-102. [PMID: 28445461 PMCID: PMC5567674 DOI: 10.1038/nature22311] [Citation(s) in RCA: 198] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 03/30/2017] [Indexed: 12/11/2022]
Abstract
The relative contribution of the effector molecules produced by T cells to tumour rejection is unclear, but interferon-γ (IFNγ) is critical in most of the analysed models. Although IFNγ can impede tumour growth by acting directly on cancer cells, it must also act on the tumour stroma for effective rejection of large, established tumours. However, which stroma cells respond to IFNγ and by which mechanism IFNγ contributes to tumour rejection through stromal targeting have remained unknown. Here we use a model of IFNγ induction and an IFNγ-GFP fusion protein in large, vascularized tumours growing in mice that express the IFNγ receptor exclusively in defined cell types. Responsiveness to IFNγ by myeloid cells and other haematopoietic cells, including T cells or fibroblasts, was not sufficient for IFNγ-induced tumour regression, whereas responsiveness of endothelial cells to IFNγ was necessary and sufficient. Intravital microscopy revealed IFNγ-induced regression of the tumour vasculature, resulting in arrest of blood flow and subsequent collapse of tumours, similar to non-haemorrhagic necrosis in ischaemia and unlike haemorrhagic necrosis induced by tumour necrosis factor. The early events of IFNγ-induced tumour ischaemia resemble non-apoptotic blood vessel regression during development, wound healing or IFNγ-mediated, pregnancy-induced remodelling of uterine arteries. A better mechanistic understanding of how solid tumours are rejected may aid the design of more effective protocols for adoptive T-cell therapy.
Collapse
Affiliation(s)
- Thomas Kammertoens
- Institute of Immunology, Charité Campus Buch, 13125 Berlin, Germany
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Christian Friese
- Institute of Immunology, Charité Campus Buch, 13125 Berlin, Germany
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Ainhoa Arina
- Department of Radiation and Cellular Oncology, Ludwig Center for Metastasis Research, The University of Chicago, Chicago, Illinois 60637, USA
| | - Christian Idel
- Department of Pathology, The University of Chicago, Chicago, Illinois 60637, USA
| | - Dana Briesemeister
- Institute of Immunology, Charité Campus Buch, 13125 Berlin, Germany
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Michael Rothe
- Institute of Immunology, Charité Campus Buch, 13125 Berlin, Germany
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Andranik Ivanov
- Berlin Institute of Health, 10117 Berlin, Germany
- Charité - Universitätsmedizin, 10117 Berlin, Germany
| | - Anna Szymborska
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Giannino Patone
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Severine Kunz
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | | | - Boris Engels
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Matthias Leisegang
- Institute of Immunology, Charité Campus Buch, 13125 Berlin, Germany
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
- Berlin Institute of Health, 10117 Berlin, Germany
| | - Ana Textor
- Institute of Immunology, Charité Campus Buch, 13125 Berlin, Germany
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | | | - Marcus Fruttiger
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Michael Lohoff
- Institute for Medical Microbiology, University of Marburg, 35032 Marburg, Germany
| | - Andreas Herrmann
- Beckman Research Institute at the Comprehensive Cancer Center City of Hope, Los Angeles, California 91010-3000, USA
| | - Hua Yu
- Beckman Research Institute at the Comprehensive Cancer Center City of Hope, Los Angeles, California 91010-3000, USA
| | - Ralph Weichselbaum
- Department of Radiation and Cellular Oncology, Ludwig Center for Metastasis Research, The University of Chicago, Chicago, Illinois 60637, USA
| | - Wolfgang Uckert
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
- Berlin Institute of Health, 10117 Berlin, Germany
| | - Norbert Hübner
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
- Charité - Universitätsmedizin, 10117 Berlin, Germany
- DZHK (German Center for Cardiovascular Research), partner site Berlin, 13347 Berlin, Germany
| | - Holger Gerhardt
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
- Berlin Institute of Health, 10117 Berlin, Germany
- DZHK (German Center for Cardiovascular Research), partner site Berlin, 13347 Berlin, Germany
| | - Dieter Beule
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
- Berlin Institute of Health, 10117 Berlin, Germany
| | - Hans Schreiber
- Institute of Immunology, Charité Campus Buch, 13125 Berlin, Germany
- Department of Pathology, The University of Chicago, Chicago, Illinois 60637, USA
- Berlin Institute of Health, 10117 Berlin, Germany
| | - Thomas Blankenstein
- Institute of Immunology, Charité Campus Buch, 13125 Berlin, Germany
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
- Berlin Institute of Health, 10117 Berlin, Germany
| |
Collapse
|
44
|
Guimarães E, Machado R, Fonseca MDC, França A, Carvalho C, Araújo e Silva AC, Almeida B, Cassini P, Hissa B, Drumond L, Gonçalves C, Fernandes G, De Brot M, Moraes M, Barcelos L, Ortega JM, Oliveira A, Leite MF. Inositol 1, 4, 5-trisphosphate-dependent nuclear calcium signals regulate angiogenesis and cell motility in triple negative breast cancer. PLoS One 2017; 12:e0175041. [PMID: 28376104 PMCID: PMC5380351 DOI: 10.1371/journal.pone.0175041] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 03/20/2017] [Indexed: 01/19/2023] Open
Abstract
Increases in nuclear calcium concentration generate specific biological outcomes that differ from those resulting from increased cytoplasmic calcium. Nuclear calcium effects on tumor cell proliferation are widely appreciated; nevertheless, its involvement in other steps of tumor progression is not well understood. Therefore, we evaluated whether nuclear calcium is essential in other additional stages of tumor progression, including key steps associated with the formation of the primary tumor or with the metastatic cascade. We found that nuclear calcium buffering impaired 4T1 triple negative breast cancer growth not just by decreasing tumor cell proliferation, but also by enhancing tumor necrosis. Moreover, nuclear calcium regulates tumor angiogenesis through a mechanism that involves the upregulation of the anti-angiogenic C-X-C motif chemokine 10 (CXCL10-IP10). In addition, nuclear calcium buffering regulates breast tumor cell motility, culminating in less cell invasion, likely due to enhanced vinculin expression, a focal adhesion structural protein. Together, our results show that nuclear calcium is essential for triple breast cancer angiogenesis and cell migration and can be considered as a promising strategic target for triple negative breast cancer therapy.
Collapse
Affiliation(s)
- Erika Guimarães
- Department of Molecular Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Rodrigo Machado
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Matheus de Castro Fonseca
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil
- Brazilian National Laboratory for Biosciences, Center for Research in Energy and Materials, Campinas, Brazil
| | - Andressa França
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Clarissa Carvalho
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Brígida Almeida
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Puebla Cassini
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Bárbara Hissa
- Department of Physics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Luciana Drumond
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Carlos Gonçalves
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Gabriel Fernandes
- Genomics Sciences and Biotechnology of Universidade Católica de Brasília, Brasília, Brazil
| | - Marina De Brot
- Department of Pathological Anatomy, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Márcio Moraes
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Lucíola Barcelos
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - José Miguel Ortega
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - André Oliveira
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - M. Fátima Leite
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
45
|
Zhang Y, Liu NM, Wang Y, Youn JY, Cai H. Endothelial cell calpain as a critical modulator of angiogenesis. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1326-1335. [PMID: 28366876 DOI: 10.1016/j.bbadis.2017.03.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 03/04/2017] [Accepted: 03/28/2017] [Indexed: 12/13/2022]
Abstract
Calpains are a family of calcium-dependent non-lysosomal cysteine proteases. In particular, calpains residing in the endothelial cells play important roles in angiogenesis. It has been shown that calpain activity can be increased in endothelial cells by growth factors, primarily vascular endothelial growth factor (VEGF). VEGF/VEGFR2 induces calpain 2 dependent activation of PI3K/AMPK/Akt/eNOS pathway, and consequent nitric oxide production and physiological angiogenesis. Under pathological conditions such as tumor angiogenesis, endothelial calpains can be activated by hypoxia. This review focuses on the molecular regulatory mechanisms of calpain activation, and the newly identified mechanistic roles and downstream signaling events of calpains in physiological angiogenesis, and in the conditions of pathological tumor angiogenesis and diabetic wound healing, as well as retinopathy and atherosclerosis that are also associated with an increase in calpain activity. Further discussed include the differential strategies of modulating angiogenesis through manipulating calpain expression/activity in different pathological settings. Targeted limitation of angiogenesis in cancer and targeted promotion of angiogenesis in diabetic wound healing via modulations of calpains and calpain-dependent signaling mechanisms are of significant translational potential. Emerging strategies of tissue-specific targeting, environment-dependent targeting, and genome-targeted editing may turn out to be effective regimens for targeted manipulation of angiogenesis through calpain pathways, for differential treatments including both attenuation of tumor angiogenesis and potentiation of diabetic angiogenesis.
Collapse
Affiliation(s)
- Yixuan Zhang
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA; Division of Cardiology, Department Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA
| | - Norika Mengchia Liu
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA; Division of Cardiology, Department Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA
| | - Yongchen Wang
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA; Division of Cardiology, Department Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA
| | - Ji Youn Youn
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA; Division of Cardiology, Department Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA
| | - Hua Cai
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA; Division of Cardiology, Department Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA.
| |
Collapse
|
46
|
Ha Y, Liu H, Zhu S, Yi P, Liu W, Nathanson J, Kayed R, Loucas B, Sun J, Frishman LJ, Motamedi M, Zhang W. Critical Role of the CXCL10/C-X-C Chemokine Receptor 3 Axis in Promoting Leukocyte Recruitment and Neuronal Injury during Traumatic Optic Neuropathy Induced by Optic Nerve Crush. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 187:352-365. [PMID: 27960090 DOI: 10.1016/j.ajpath.2016.10.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 09/14/2016] [Accepted: 10/13/2016] [Indexed: 12/11/2022]
Abstract
Traumatic optic neuropathy (TON) is an acute injury of the optic nerve secondary to trauma. Loss of retinal ganglion cells (RGCs) is a key pathological process in TON, yet mechanisms responsible for RGC death remain unclear. In a mouse model of TON, real-time noninvasive imaging revealed a dramatic increase in leukocyte rolling and adhesion in veins near the optic nerve (ON) head at 9 hours after ON injury. Although RGC dysfunction and loss were not detected at 24 hours after injury, massive leukocyte infiltration was observed in the superficial retina. These cells were identified as T cells, microglia/monocytes, and neutrophils but not B cells. CXCL10 is a chemokine that recruits leukocytes after binding to its receptor C-X-C chemokine receptor (CXCR) 3. The levels of CXCL10 and CXCR3 were markedly elevated in TON, and up-regulation of CXCL10 was mediated by STAT1/3. Deleting CXCR3 in leukocytes significantly reduced leukocyte recruitment, and prevented RGC death at 7 days after ON injury. Treatment with CXCR3 antagonist attenuated TON-induced RGC dysfunction and cell loss. In vitro co-culture of primary RGCs with leukocytes resulted in increased RGC apoptosis, which was exaggerated in the presence of CXCL10. These results indicate that leukocyte recruitment in retinal vessels near the ON head is an early event in TON and the CXCL10/CXCR3 axis has a critical role in recruiting leukocytes and inducing RGC death.
Collapse
Affiliation(s)
- Yonju Ha
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas
| | - Hua Liu
- Center for Biomedical Engineering, University of Texas Medical Branch, Galveston, Texas
| | - Shuang Zhu
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas
| | - Panpan Yi
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Wei Liu
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas
| | - Jared Nathanson
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas
| | - Rakez Kayed
- Department of Neurology, University of Texas Medical Branch, Galveston, Texas
| | - Bradford Loucas
- Department of Radiation Oncology, University of Texas Medical Branch, Galveston, Texas
| | - Jiaren Sun
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | | | - Massoud Motamedi
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas; Center for Biomedical Engineering, University of Texas Medical Branch, Galveston, Texas
| | - Wenbo Zhang
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas; Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas.
| |
Collapse
|
47
|
Huen AC, Marathi A, Nam PK, Wells A. CXCL11 Expression by Keratinocytes Occurs Transiently Between Reaching Confluence and Cellular Compaction. Adv Wound Care (New Rochelle) 2016; 5:517-526. [PMID: 28078185 DOI: 10.1089/wound.2015.0680] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 02/01/2016] [Indexed: 12/31/2022] Open
Abstract
Objective: To investigate whether differentiation or cellular confluence is responsible for CXCL11 expression patterns in re-epithelialization. Approach:In vitro model systems of re-epithelialization using the HaCaT keratinocyte cell line were utilized in monitoring expression of differentiation markers, including desmoplakin and various cytokeratins while evaluating for an association with chemokine CXCL11 expression. Results: CXCL11 expression was elevated in sparse culture with peak expression near the time of confluence. This somewhat followed the accumulation of desmoplakin in detergent-insoluble pool of proteins. However, in postconfluent, despite continued accumulation of desmoplakin within cells, CXCL11 expression decreased to baseline levels. This biphasic pattern was also seen in low calcium culture, an environment that inhibits keratinocyte differentiation and accumulation of desmosomal proteins. Highest CXCL11-expressing areas best correlated with newly confluent areas within culture expressing basal keratin 14, but also activated keratin 6. Innovation: Achievement of a threshold cellular density induces cell signaling cascade through CXCR3 that, in addition to other undiscovered pathways, can progress cutaneous wounds from the proliferative into the remodeling phases of cutaneous wound healing. Conclusion: These results suggest that the achievement of confluence with increased cellular density by migrating keratinocytes at the wound edge triggers expression of CXCL11. Since CXCR3 stimulation in endothelial cells results in apoptosis and causes neovascular pruning, whereas stimulation of CXCR3 in fibroblasts results decreased motility and cellular contraction, we speculate that CXCL11 expression by epidermal cells upon achieving cellular confluence could be the source of CXCR3 stimulation in the dermis ushering a transition from proliferative to remodeling phases of wound healing.
Collapse
Affiliation(s)
- Arthur C. Huen
- Department of Dermatology, McGowan Institute for Regenerative Medicine, VA Pittsburgh Health System, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Archana Marathi
- Department of Dermatology, McGowan Institute for Regenerative Medicine, VA Pittsburgh Health System, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Peter K. Nam
- Department of Dermatology, McGowan Institute for Regenerative Medicine, VA Pittsburgh Health System, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Alan Wells
- Department of Pathology, McGowan Institute for Regenerative Medicine, VA Pittsburgh Health System, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
48
|
Zhang J, Yang J, Huang T, Shu Y, Chen L. Identification of novel proliferative diabetic retinopathy related genes on protein–protein interaction network. Neurocomputing 2016. [DOI: 10.1016/j.neucom.2015.09.136] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
49
|
MicroRNA regulation of endothelial TREX1 reprograms the tumour microenvironment. Nat Commun 2016; 7:13597. [PMID: 27886180 PMCID: PMC5133658 DOI: 10.1038/ncomms13597] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 10/18/2016] [Indexed: 02/07/2023] Open
Abstract
Rather than targeting tumour cells directly, elements of the tumour microenvironment can be modulated to sensitize tumours to the effects of therapy. Here we report a unique mechanism by which ectopic microRNA-103 can manipulate tumour-associated endothelial cells to enhance tumour cell death. Using gain-and-loss of function approaches, we show that miR-103 exacerbates DNA damage and inhibits angiogenesis in vitro and in vivo. Local, systemic or vascular-targeted delivery of miR-103 in tumour-bearing mice decreased angiogenesis and tumour growth. Mechanistically, miR-103 regulation of its target gene TREX1 in endothelial cells governs the secretion of pro-inflammatory cytokines into the tumour microenvironment. Our data suggest that this inflammatory milieu may potentiate tumour cell death by supporting immune activation and inducing tumour expression of Fas and TRAIL receptors. Our findings reveal miR-mediated crosstalk between vasculature and tumour cells that can be exploited to improve the efficacy of chemotherapy and radiation. The tumour microenvironment can be modulated to sensitize tumours to the effects of therapy. Here the authors show that radiation induced miR-103 downregulates TREX1 in endothelial cells, decreases angiogenesis and leads to the secretion of proinflammatory mediators that reduce tumour growth.
Collapse
|
50
|
Ligi D, Mosti G, Croce L, Raffetto JD, Mannello F. Chronic venous disease – Part I: Inflammatory biomarkers in wound healing. Biochim Biophys Acta Mol Basis Dis 2016; 1862:1964-74. [DOI: 10.1016/j.bbadis.2016.07.018] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 07/20/2016] [Accepted: 07/26/2016] [Indexed: 01/12/2023]
|