Changing stroke rehab and research worldwide now.Time is Brain! trillions and trillions of neurons that DIE each day because there are NO effective hyperacute therapies besides tPA(only 12% effective). I have 523 posts on hyperacute therapy, enough for researchers to spend decades proving them out. These are my personal ideas and blog on stroke rehabilitation and stroke research. Do not attempt any of these without checking with your medical provider. Unless you join me in agitating, when you need these therapies they won't be there.

What this blog is for:

My blog is not to help survivors recover, it is to have the 10 million yearly stroke survivors light fires underneath their doctors, stroke hospitals and stroke researchers to get stroke solved. 100% recovery. The stroke medical world is completely failing at that goal, they don't even have it as a goal. Shortly after getting out of the hospital and getting NO information on the process or protocols of stroke rehabilitation and recovery I started searching on the internet and found that no other survivor received useful information. This is an attempt to cover all stroke rehabilitation information that should be readily available to survivors so they can talk with informed knowledge to their medical staff. It lays out what needs to be done to get stroke survivors closer to 100% recovery. It's quite disgusting that this information is not available from every stroke association and doctors group.

Wednesday, March 10, 2021

Hyperbaric oxygen therapy increases telomere length and decreases immunosenescence in isolated blood cells: a prospective trial

 I don't necessarily trust this since it was done by Shai Efrati. Shai Efrati has conflicts, he is the director of the Hyperbaric Oxygen Institute at the Assaf Harofeh Medical Center.

Hyperbaric oxygen therapy increases telomere length and decreases immunosenescence in isolated blood cells: a prospective trial

Yafit Hachmo1, * , Amir Hadanny2,3,4, * , Ramzia Abu Hamed1 , Malka Daniel-Kotovsky2 , Merav Catalogna2 , Gregory Fishlev2 , Erez Lang2 , Nir Polak2 , Keren Doenyas2 , Mony Friedman2 , Yonatan Zemel2 , Yair Bechor2 , Shai Efrati1,2,3,5

  • 1 Research and Development Unit, Shamir Medical Center, Zerifin, Israel
  • 2 The Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf-Harofeh) Medical Center, Zerifin, Israel
  • 3 Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
  • 4 Bar Ilan University, Ramat-Gan, Israel
  • 5 Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, Israel
* Equal contribution

Received: September 3, 2020       Accepted: October 22, 2020       Published: November 18, 2020

https://doi.org/10.18632/aging.202188
How to Cite

Copyright: © 2020 Yafit et al. This is an open access article distributed under the terms of the Creative Commons Attribution License (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Abstract

Introduction:  

Aging is characterized by the progressive loss of physiological capacity. At the cellular level, two key hallmarks of the aging process include telomere length (TL) shortening and cellular senescence. Repeated intermittent hyperoxic exposures, using certain hyperbaric oxygen therapy (HBOT) protocols, can induce regenerative effects which normally occur during hypoxia. The aim of the current study was to evaluate whether HBOT affects TL and senescent cell concentrations in a normal, non-pathological, aging adult population.

Methods: 

Thirty-five healthy independently living adults, aged 64 and older, were enrolled to receive 60 daily HBOT exposures. Whole blood samples were collected at baseline, at the 30th and 60th session, and 1-2 weeks following the last HBOT session. Peripheral blood mononuclear cells (PBMCs) telomeres length and senescence were assessed.

Results: 

Telomeres length of T helper, T cytotoxic, natural killer and B cells increased significantly by over 20% following HBOT. The most significant change was noticed in B cells which increased at the 30th session, 60th session and post HBOT by 25.68%±40.42 (p=0.007), 29.39%±23.39 (p=0.0001) and 37.63%±52.73 (p=0.007), respectively.

There was a significant decrease in the number of senescent T helpers by -37.30%±33.04 post-HBOT (P<0.0001). T-cytotoxic senescent cell percentages decreased significantly by -10.96%±12.59 (p=0.0004) post-HBOT.

In conclusion, the study indicates that HBOT may induce significant senolytic effects including significantly increasing telomere length and clearance of senescent cells in the aging populations.

Introduction

Aging can be characterized by the progressive loss of physiological integrity, resulting in impaired functions and susceptibility for diseases and death. This biological deterioration is considered a major risk factor for cancer, cardiovascular diseases, diabetes and Alzheimer’s disease among others. At the cellular level, there are two key hallmarks of the aging process: shortening of telomere length and cellular senescence [1].

Telomeres are tandem nucleotide repeats located at the end of the chromosomes which maintain genomic stability. Telomeres shorten during replication (mitosis) due to the inherent inability to fully replicate the end part of the lagging DNA strand [2]. Telomere length (TL), measuring between 4 to 15 kilobases, gradually shorten by ~20-40 bases per year and is associated with different diseases, low physical performance and cortical thinning of the brain [35]. When TL reaches a critical length, cells cannot replicate and progress to senescence or programmed cell death [6]. Goglin et al. demonstrated that adults with shorter TLs have increased mortality rates [7]. Shortened TLs can be a direct inherited trait, but several environmental factors have also been associated with shortening TL including stress, lack of physical endurance activity, excess body mass index, smoking, chronic inflammation, vitamins deficiency and oxidative stress [2, 8, 9].

Cellular senescence is an arrest of the cell cycle which can be caused by telomere shortening [10], as well as other aging associated stimuli independent of TL such as non-telomeric DNA damage [1]. The primary purpose of senescence is to prevent propagation of damaged cells by triggering their elimination via the immune system. The accumulation of senescent cells with aging reflects either an increase in the generation of these cells and/or a decrease in their clearance, which in turn aggravates the damage and contributes to aging [1].

A growing body of research has found several pharmacological agents that can reduce the telomere shortening rate [11, 12]. Several lifestyle interventions including endurance training, diets and supplements targeting cell metabolism and oxidative stress have reported relatively small effects (2-5%) on TL3, [2, 8, 9].

Hyperbaric oxygen therapy (HBOT) utilizes 100% oxygen in an environmental pressure higher than one absolute atmospheres (ATA) to enhance the amount of oxygen dissolved in body’s tissues. Repeated intermittent hyperoxic exposures, using certain HBOT protocols, can induce physiological effects which normally occur during hypoxia in a hyperoxic environment, the so called hyperoxic-hypoxic paradox [1316]. In addition, it was recently demonstrated that HBOT can induce cognitive enhancements in healthy aging adults via mechanisms involving regional changes in cerebral blood flow [17]. On the cellular level, it was demonstrated that HBOT can induce the expression of hypoxia induced factor (HIF), vascular endothelial growth factor (VEGF) and sirtuin (SIRT), stem cell proliferation, mitochondrial biogenesis, angiogenesis and neurogenesis [18]. However, no study to date has examined HBOT’s effects on TL and senescent cell accumulation.

The aim of the current study was to evaluate whether HBOT affects TL and senescence-like T-cells population in aging adults.

Results

Thirty-five individuals were assigned to HBOT. Five patients did not complete baseline assessments and were excluded. All 30 patients who completed baseline evaluations completed the interventions. Due to the low quality of blood samples (low number of cells or technician error), four patients were excluded from the telomere analysis and 10 patients from senescent cell analysis (Figure 1). The baseline characteristics and comparison of the cohorts following exclusion of the patients are provided in Table 1. There were no significant differences between the three groups (Table 1).

Patient flowchart.

Figure 1. Patient flowchart.

Table 1. Baseline characteristics.



HBOTTelomere analysisSenescent analysisP-value
N
3025 (83.3%)20 (66.6%)
Age (years)
68.41±13.267.56±14.3566.70±16.000.917
BMI
26.77±3.2026.89±3.3427.14±3.810.946
Males
16 (53.3%)13 (52.0%)10 (50.0%)0.987
Females
14 (47.7%)12 (48.0%)10 (50.0%)0.987
Complete blood count





Hemoglobin6.33±1.256.57±1.156.58±1.290.707

White blood cells14.02±1.4013.92±1.3513.97±1.490.969

%PBMC39.96±6.7539.25±6.6438.59±6.630.774

Platelets239.87±1.39244.08±43.0254.05±41.40.559
Chronic medical conditions




Atrial fibrillation4 (13.3%)4 (16.0%)2 (10.0%)0.841

Hypothyroidism4 (13.3%)4 (16.0%)3 (15.8%)0.956

Obstructive sleep apnea4 (13.3%)4 (16.0%)3 (15.0%)0.961

Asthma1 (3.3%)1 (4.0%)00.680

BPH7 (23.3%)5 (20.0%)6 (30.0%)0.733

GERD3 (10%)2 (8.0%)2 (10.0%)0.961

Osteoporosis5 (16.7%)5 (20.0%)4 (20.0%)0.936

Rheumatic arthritis1 (3.3%)01 (5.0%)0.561

Osteoarthritis7 (23.3%)4 (16.0%)5 (25.0%)0.755

Diabetes mellitus3 (10%)3 (12.0%)2 (10.0%)0.966

Hypertension7 (23.3%)5 (20.0%)5 (25.0%)0.918

Dyslipidemia16 (53.3%)14 (56.0%)12 (60.0%)0.897

Ischemic heart disease2 (6.7%)1 (4.0%)2 (10.0%)0.725

History of smoking10 (33.3%)8 (32.0%)7 (35.0%)0.978
Chronic medications




Anti-aggregation8 (26.7%)6 (24.0%)5 (25.0%)0.974

ACE-Inhibitors/ARB blockers6 (20%)6 (24.0%)6 (30.0%)0.720

Beta blockers5 (16.7%)5 (20.0%)3 (15.0%)0.901

Calcium blockers3 (10%)3 (12.0%)2 (10.0%)0.966

Alpha blockers7 (23.3%)5 (20.0%)6 (30.0%)0.733

Diuretics2 (6.7%)1 (4.0%) 1 (5.0%)0.906

Statins10 (33.3%)9 (36.0%)7 (35.0%)0.978

Oral hypoglycemic1 (3.3%)1 (4.0%)1 (5.0%)0.958

Bisphosphonates1 (3.3%)1 (4.0%)1 (5.0%)0.958

Proton pump inhibitors3 (10%)3 (12.0%)3 (15.0%)0.726

Hormones3 (10%)3 (12.0%)2 (10.0%)0.966

Benzodiazepines3 (10%)2 (8.0%)1 (5.0%)0.816

SSRI5 (16.7%)5 (20.0%)3 (15.0%)0.990

Telomere length

Compared to the baseline, the T-helper telomere lengths were significantly increased at the 30th session and post-HBOT by 21.70±40.05 (p=0.042), 23.69%±39.54 (p=0.012) and 29.30±38.51 (p=0.005), respectively (Figure 2). However, repeated measures analysis shows a non-significant trend (F=4.663, p=0.06, Table 2 and Figure 2).

Telomere length changes with HBOT. Mean+SEM *p

Figure 2. Telomere length changes with HBOT. Mean+SEM *p<0.05, **p<0.01, ***p<0.001.

Table 2. Telomere length and senescent cell changes post-HBOT.



Absolute changesRelative changes (%)Repeated measures F (p)
PBMCBaseline30th Session
60th SessionPost HBOT30th session60th sessionPost-HBOT
PBMC ((N=25)2.55±0.53


-0.15±0.40

-4.91±16.701.987 (t) 0.09
PBMC (N=20)2.50±0.53


-0.13±0.31

-4.21±11.991.810 (t) 0.07
Relative telomeres length (N=25)
Natural killer9.27±1.9111.77±5.14 (0.045)
10.73±2.73 (0.013)11.75±4.22 (0.06)25.02±51.4220.56±33.3522.16±44.810.812 (0.391)
B-cells8.36±2.0210.22±3.04 (0.007)
11.23±3.58 (0.0001)11.17±2.98 (0.007)25.68±40.4229.39±23.3937.63±52.737.390 (0.017)
T Helper8.04±1.829.92±3.68 (0.042)
9.63±2.17 (0.012)10.20±2.77 (0.005)21.70±40.0523.69±39.5429.30±38.514.663 (0.063)
T Cytotoxic8.26±1.549.83±4.08 (0.11)
10.08±3.33 (0.019)10.15±2.74 (0.023)18.29±45.6224.13±40.8819.59±33.981.159 (0.310)
Senescent cells (% of T cells) (N=20)
T Helper10.29±5.427.84±7.09 (0.09)
8.51±7.45 (0.20)6.22±4.88 (<0.0001)-19.66±80.03-11.67±94.30-37.30±33.048.548 (0.01)
T Cytotoxic52.19±21.0745.53±19.91 (<0.0001)
45.45±18.81 (0.002)46.59±21.91 (0.0004)-12.21±8.74-9.81±9.50-10.96±12.596.916 (0.018)
P-values shown in () compared to baseline.
P-values in bold <0.05.

Compared to baseline, telomere lengths of B cells increased significantly at the 30th session, 60th session and post-HBOT by 25.68%±40.42 (p=0.007), 29.39%±23.39 (p=0.0001) and 37.63%±52.73 (p=0.007), respectively (Figure 2). Repeated measures analysis shows a significant within-group effect (F=0.390, p=0.017, Table 2 and Figure 2).

Compared to baseline, natural killer cells telomer lengths significantly increased at the 30th session (p=0.045) and at the 60th session by 20.56% ±33.35 (p=0.013). Post-HBOT, telomere lengths increased by 22.16%±44.81 post-HBOT (p=0.06, Table 2 and Figure 2). Repeated measures analysis indicates that there was no additional significant effect after the 30th session (F=0.812, p=0.391).

Compared to baseline, cytotoxic T-cells had a non-significant increase at the 30th session by 18.29%±45.62 (p=0.11), followed by a significant increase of 24.13%±40.88 at the 60th session (p=0.0019) and 19.59%±33.98 post-HBOT (p=0.023). Repeated measures analysis indicates that there was no additional significant effect after the 30th session (F=1.159, p=0.310, Table 2 and Figure 2).

Senescent cells

There was a non-significant decrease in the number of senescent T-helpers at the 30th session and 60th session by -19.66%±80.03 (p=0.09) and -11.67%±94.30 (p=0.20) respectively. However, there was a significant drop in the number of senescent T helpers by -37.30%±33.04 post-HBOT (P<0.0001, Figure 3). Repeated measures analysis showed a significant continuous effect even after the 30th session, with a within-group effect (F=8.547, p=0.01, Table 2 and Figure 3).

Senescent cell changes with HBOT. Mean+SEM *p
 

No comments:

Post a Comment