GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Journal of Hypertension, Ovid Technologies (Wolters Kluwer Health), Vol. 39, No. Supplement 1 ( 2021-04), p. e257-
    Type of Medium: Online Resource
    ISSN: 0263-6352 , 1473-5598
    Language: English
    Publisher: Ovid Technologies (Wolters Kluwer Health)
    Publication Date: 2021
    detail.hit.zdb_id: 2017684-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Scientific Reports, Springer Science and Business Media LLC, Vol. 14, No. 1 ( 2024-01-22)
    Abstract: Vascular inflammation and fibrosis are hallmarks of hypertension and contribute to the development of cardiovascular disease and cognitive impairment. However, current anti-hypertensive drugs do not treat the underlying tissue damage, such as inflammation-associated fibrosis. Human amnion epithelial cells have several properties amenable for treating vascular pathology. This study tested the effect of amnion epithelial cells on vascular pathology and cognitive impairment during hypertension. Male C57Bl6 mice (8–12 weeks) were administered vehicle (saline; n = 58) or angiotensin II (0.7 mg/kg/d, n = 56) subcutaneously for 14 d. After surgery, a subset of mice were injected with 10 6 amnion epithelial cells intravenously. Angiotensin II infusion increased systolic blood pressure, aortic pulse wave velocity, accumulation of aortic leukocytes, and aortic mRNA expression of collagen subtypes compared to vehicle-infused mice (n = 9–11, P 〈 0.05). Administration of amnion epithelial cells attenuated these effects of angiotensin II ( P 〈 0.05). Angiotensin II-induced cognitive impairment was prevented by amnion epithelial cell therapy (n = 7–9, P 〈 0.05). In the brain, amnion epithelial cells modulated some of the inflammatory genes that angiotensin II promoted differential expression of (n = 6, p-adjusted 〈 0.05). These findings suggest that amnion epithelial cells could be explored as a potential therapy to inhibit vascular pathology and cognitive impairment during hypertension.
    Type of Medium: Online Resource
    ISSN: 2045-2322
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2024
    detail.hit.zdb_id: 2615211-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    Frontiers Media SA ; 2023
    In:  Frontiers in Neuroscience Vol. 17 ( 2023-6-16)
    In: Frontiers in Neuroscience, Frontiers Media SA, Vol. 17 ( 2023-6-16)
    Abstract: Thrombolytic agents such as tissue plasminogen activator (tPA) are the only drug class approved to treat ischemic stroke and are usually administered within 4.5 h. However, only ~20% of ischemic stroke patients are eligible to receive the therapy. We previously demonstrated that early intravenous administration of human amnion epithelial cells (hAECs) can limit brain inflammation and infarct growth in experimental stroke. Here, we have tested whether hAECs exert cerebroprotective effects in combination with tPA in mice. Methods Male C57Bl/6 mice were subjected to middle cerebral artery occlusion for 60 min followed by reperfusion. Immediately following reperfusion, vehicle (saline, n  = 31) or tPA (10 mg/kg; n  = 73) was administered intravenously. After 30 min of reperfusion, tPA-treated mice were injected intravenously with either hAECs (1×10 6 ; n  = 32) or vehicle (2% human serum albumin; n  = 41). A further 15 sham-operated mice were treated with vehicle ( n  = 7) or tPA + vehicle ( n  = 8). Mice were designated to be euthanised at 3, 6 or 24 h post-stroke ( n  = 21, 31, and 52, respectively), and brains were collected to assess infarct volume, blood–brain barrier (BBB) disruption, intracerebral bleeding and inflammatory cell content. Results There was no mortality within 6 h of stroke onset, but a high mortality occurred in tPA + saline-treated mice between 6 h and 24 h post-stroke in comparison to mice treated with tPA + hAECs (61% vs. 27%, p  = 0.04). No mortality occurred within 24 h of sham surgery in mice treated with tPA + vehicle. We focused on early infarct expansion within 6 h of stroke and found that infarction was ~50% larger in tPA + saline- than in vehicle-treated mice (23 ± 3 mm 3 vs. 15 ± 2 mm 3 , p  = 0.02) but not in mice receiving tPA + hAECs (13 ± 2 mm 3 , p   & lt; 0.01 vs. tPA + saline) in which intracerebral hAECs were detected. Similar to the profiles of infarct expansion, BBB disruption and intracerebral bleeding in tPA + saline-treated mice at 6 h was 50–60% greater than in vehicle-treated controls (2.6 ± 0.5 vs. 1.6 ± 0.2, p  = 0.05) but not after tPA + hAECs treatment (1.7 ± 0.2, p  = 0.10 vs. tPA + saline). No differences in inflammatory cell content were detected between treatment groups. Conclusion When administered following tPA in acute stroke, hAECs improve safety and attenuate infarct growth in association with less BBB disruption and lower 24 h mortality.
    Type of Medium: Online Resource
    ISSN: 1662-453X
    Language: Unknown
    Publisher: Frontiers Media SA
    Publication Date: 2023
    detail.hit.zdb_id: 2411902-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Frontiers in Neuroscience, Frontiers Media SA, Vol. 17 ( 2023-7-31)
    Type of Medium: Online Resource
    ISSN: 1662-453X
    Language: Unknown
    Publisher: Frontiers Media SA
    Publication Date: 2023
    detail.hit.zdb_id: 2411902-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Physiology, American Physiological Society, Vol. 38, No. S1 ( 2023-05)
    Abstract: Introduction: Vascular stiffening and inflammation are hallmarks of hypertension and contribute to the development of cardiovascular disease and cognitive impairment. Current treatments for hypertension may reduce blood pressure but do not target the associated pathological changes and end-organ damage. It is also inconclusive from clinical trials whether anti-hypertensive medication can improve cognitive impairment. Cell therapy has great therapeutic potential as unlike single pharmacological agents, cells can deliver multiple mediators which could more effectively target complex disease mechanisms. Human amnion epithelial cells (AECs) have many properties (eg. anti-inflammatory, anti-fibrotic, and immunologically inert) that make them attractive candidates for a cell-based therapy for vascular and brain pathology. Aim: To test the potential of AECs to treat vascular and brain pathology in angiotensin II-induced hypertension. Methods. Male C57Bl6 mice (8-12 weeks) were administered vehicle (saline; n=35) or angiotensin II (0.7 mg/kg/d, n=37) for 14 d via an osmotic minipump. After minipump implantation, a subset of mice were injected with 10 6 of AECs intravenously. Systolic blood pressure was measured using tail-cuff; pulse wave velocity was measured using ultrasound, inflammation was assessed using flow cytometry and RNA sequencing, markers of fibrosis using quantitative PCR and cognitive function using novel object recognition test. Results: Angiotensin II infusion increased blood pressure and aortic pulse wave velocity (n=6-7, P 〈 0.05). In aorta, angiotensin II promoted accumulation of leukocytes, specifically macrophages and monocytes, which were elevated by ~3-fold compared to vehicle-infused mice (n=9-11, P 〈 0.05). Angiotensin II also increased aortic mRNA expression of collagen type 1 alpha 1 ( Col1a1) by ~4-fold and collagen type 5 alpha 1 ( Col5a1) by ~7-fold compared to vehicle (n=6-8, P 〈 0.05). Co-administration of AECs limited the development of hypertension and aortic stiffening by angiotensin II (185±5 mmHg vs 165±4 mmHg; n=9-11, P 〈 0.05), as well as the aortic infiltration of macrophages and monocytes (n=9-11, P 〈 0.05) and expression of Col1a1 and Col5a1 (n=6-8, P 〈 0.05). Infusion with angiotensin II also impaired memory which was improved by AECs (n=7-9, P 〈 0.05). RNA sequencing revealed that in the brain, angiotensin II increased expression of genes involved in inflammation ( Ptcra, Blk, Bpifb1), fibrosis ( Col6a5) and apoptosis ( Fas) and co-administration of AECs caused downregulation of these genes. Conclusions: Intravenous administration of AECs blunted angiotensin II-induced hypertension, aortic stiffening, inflammation and cognitive impairment in male mice. The beneficial effects of AECs on blood pressure and aortic stiffening could have contributed to the improvement in cognition. This study suggests that AECs or their cellular products could be explored as treatments for vascular and brain pathology during hypertension. La Trobe University Start Up grant This is the full abstract presented at the American Physiology Summit 2023 meeting and is only available in HTML format. There are no additional versions or additional content available for this abstract. Physiology was not involved in the peer review process.
    Type of Medium: Online Resource
    ISSN: 1548-9213 , 1548-9221
    Language: English
    Publisher: American Physiological Society
    Publication Date: 2023
    detail.hit.zdb_id: 3115360-4
    detail.hit.zdb_id: 2005759-3
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Frontiers in Neuroscience, Frontiers Media SA, Vol. 17 ( 2023-5-9)
    Abstract: We proposed a Phase I dose escalation trial to assess the safety of allogeneic human amniotic epithelial cells (hAECs) in stroke patients with a view to informing the design for a Phase II trial. Methods The design is based on 3 + 3 dose escalation design with additional components for measuring MR signal of efficacy as well as the effect of hAECs (2–8 × 10 6 /kg, i.v.) on preventing immunosuppression after stroke. Results Eight patients (six males) were recruited within 24 h of ischemic stroke onset and were infused with hAECs. We were able to increase the dose of hAECs to 8 × 10 6 cells/kg (2 × 10 6 /kg, n = 3; 4 × 10 6 /kg, n = 3; 8 × 10 6 /kg, n = 2). The mean age is 68.0 ± 10.9 (mean ± SD). The frequencies of hypertension and hyperlipidemia were 87.5%, diabetes was 37.5%, atrial fibrillation was 50%, ischemic heart disease was 37.5% and ever-smoker was 25%. Overall, baseline NIHSS was 7.5 ± 3.1, 7.8 ± 7.2 at 24 h, and 4.9 ± 5.4 at 1 week ( n = 8). The modified Rankin scale at 90 days was 2.1 ± 1.2. Supplemental oxygen was given in five patients during hAEC infusion. Using pre-defined criteria, two serious adverse events occurred. One patient developed recurrent stroke and another developed pulmonary embolism whilst in rehabilitation. For the last four patients, infusion of hAECs was split across separate infusions on subsequent days to reduce the risk for fluid overload. Conclusion Our Phase I trial demonstrates that a maximal dose of 2 × 10 6 /kg hAECs given intravenously each day over 2 days (a total of 4 × 10 6 /kg) is safe and optimal for use in a Phase II trial. Clinical trial registration ClinicalTrials.gov , identifier ACTRN12618000076279P.
    Type of Medium: Online Resource
    ISSN: 1662-453X
    Language: Unknown
    Publisher: Frontiers Media SA
    Publication Date: 2023
    detail.hit.zdb_id: 2411902-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...